1
|
Lan T, Li Y, Chen X, Wang W, Wang C, Lou H, Chen S, Yu S. Exercise-Activated mPFC Tri-Synaptic Pathway Ameliorates Depression-Like Behaviors in Mouse. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408618. [PMID: 39574315 PMCID: PMC11744721 DOI: 10.1002/advs.202408618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/03/2024] [Indexed: 01/21/2025]
Abstract
Exercise is considered as playing a pivotal role in the modulation of emotional responses. However, a precise circuit that mediates the effects of exercise on depression have yet to be elucidated. Here, a molecularly defined tri-synaptic pathway circuit is identified that correlates motor inputs with antidepressant effects. With this pathway, initial inputs from neurons within the dorsal root ganglia (DRG) project to excitatory neurons in the gracile nucleus (GR), which in turn connect with 5-HTergic neurons in the dorsal raphe nucleus (DRN), eventually coursing to excitatory pyramidal neurons within the medial prefrontal cortex (mPFC). Exercise activates this pathway, with the result that depressive- and anxiety-like behaviors in mice are significantly reduced. In addition, it is found that exercise may exert antidepressant effects through regulating synaptic plasticity within this tri-synaptic pathway. These findings reveal a hindbrain-to-forebrain neuronal circuit that specifically modulates depression and provides a potential mechanism for the antidepressant effects of exercise.
Collapse
Affiliation(s)
- Tian Lan
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Ye Li
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Xiao Chen
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Wenjing Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Changmin Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Haiyan Lou
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PharmacologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Shihong Chen
- Department of Endocrinology and MetabolismThe Second Hospital of Shandong UniversityJinanShandong250033China
| | - Shuyan Yu
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
- Department of Medical Psychology and EthicsSchool of Basic Medical sciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| |
Collapse
|
2
|
Hazlett MF, Hall VL, Patel E, Halvorsen A, Calakos N, West AE. The Perineuronal Net Protein Brevican Acts in Nucleus Accumbens Parvalbumin-Expressing Interneurons of Adult Mice to Regulate Excitatory Synaptic Inputs and Motivated Behaviors. Biol Psychiatry 2024; 96:694-707. [PMID: 38346480 PMCID: PMC11315813 DOI: 10.1016/j.biopsych.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Experience-dependent functional adaptation of nucleus accumbens (NAc) circuitry underlies the development and expression of reward-motivated behaviors. Parvalbumin-expressing GABAergic (gamma-aminobutyric acidergic) interneurons (PVINs) within the NAc are required for this process. Perineuronal nets (PNNs) are extracellular matrix structures enriched around PVINs that arise during development and have been proposed to mediate brain circuit stability. However, their function in the adult NAc is largely unknown. Here, we studied the developmental emergence and adult regulation of PNNs in the NAc of male and female mice and examined the cellular and behavioral consequences of reducing the PNN component brevican in NAc PVINs. METHODS We characterized the expression of PNN components in mouse NAc using immunofluorescence and RNA in situ hybridization. We lowered brevican in NAc PVINs of adult mice using an intersectional viral and genetic method and quantified the effects on synaptic inputs to NAc PVINs and reward-motivated learning. RESULTS PNNs around NAc PVINs were developmentally regulated and appeared during adolescence. In the adult NAc, PVIN PNNs were also dynamically regulated by cocaine. Transcription of the gene that encodes brevican was regulated in a cell type- and isoform-specific manner in the NAc, with the membrane-tethered form of brevican being highly enriched in PVINs. Lowering brevican in NAc PVINs of adult mice decreased their excitatory inputs and enhanced both short-term novel object recognition and cocaine-induced conditioned place preference. CONCLUSIONS Regulation of brevican in NAc PVINs of adult mice modulates their excitatory synaptic drive and sets experience thresholds for the development of motivated behaviors driven by rewarding stimuli.
Collapse
Affiliation(s)
- Mariah F Hazlett
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Victoria L Hall
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Esha Patel
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Aaron Halvorsen
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Nicole Calakos
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina; Department of Neurology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
3
|
Wang SH, Lee DS, Kim TH, Kim JE, Kang TC. Reciprocal regulation of oxidative stress and mitochondrial fission augments parvalbumin downregulation through CDK5-DRP1- and GPx1-NF-κB signaling pathways. Cell Death Dis 2024; 15:707. [PMID: 39349423 PMCID: PMC11443148 DOI: 10.1038/s41419-024-07050-5] [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: 02/25/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 10/02/2024]
Abstract
Loss of parvalbumin (PV) expressing neurons (PV neurons) is relevant to the underlying mechanisms of the pathogenesis of neurological and psychiatric diseases associated with the dysregulation of neuronal excitatory networks and brain metabolism. Although PV modulates mitochondrial morphology, volume and dynamics, it is largely unknown whether mitochondrial dynamics affect PV expression and what the molecular events are responsible for PV neuronal degeneration. In the present study, L-buthionine sulfoximine (BSO, an inhibitor of glutathione synthesis) did not degenerate PV neurons under physiological condition. However, BSO-induced oxidative stress decreased PV expression and facilitated cyclin-dependent kinase 5 (CDK5) tyrosine (Y) 15 phosphorylation, dynamin-related protein 1 (DRP1)-mediated mitochondrial fission and glutathione peroxidase-1 (GPx1) downregulation in PV neurons. Co-treatment of roscovitine (a CDK5 inhibitor) or mitochondrial division inhibitor-1 (Mdivi-1, an inhibitor of mitochondrial fission) attenuated BSO-induced PV downregulation. WY14643 (an inducer of mitochondrial fission) reduced PV expression without affecting CDK5 Y15 phosphorylation. Following status epilepticus (SE), CDK5 Y15 phosphorylation and mitochondrial fission were augmented in PV neurons. These were accompanied by reduced GPx1-mediated inhibition of NF-κB p65 serine (S) 536 phosphorylation. N-acetylcysteine (NAC), roscovitine and Mdivi-1 ameliorated SE-induced PV neuronal degeneration by mitigating CDK5 Y15 hyperphosphorylation, aberrant mitochondrial fragmentation and reduced GPx1-mediated NF-κB inhibition. Furthermore, SN50 (a NF-κB inhibitor) alleviated SE-induced PV neuronal degeneration, independent of dysregulation of mitochondrial fission, CDK5 hyperactivation and GPx1 downregulation. These findings provide an evidence that oxidative stress may activate CDK5-DRP1- and GPx1-NF-κB-mediated signaling pathways, which would be possible therapeutic targets for preservation of PV neurons in various diseases.
Collapse
Affiliation(s)
- Su Hyeon Wang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
4
|
Kushinsky D, Tsivourakis E, Apelblat D, Roethler O, Breger-Mikulincer M, Cohen-Kashi Malina K, Spiegel I. Daily light-induced transcription in visual cortex neurons drives downward firing rate homeostasis and stabilizes sensory processing. Cell Rep 2024; 43:114701. [PMID: 39244753 DOI: 10.1016/j.celrep.2024.114701] [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: 12/08/2023] [Revised: 05/05/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Balancing plasticity and stability in neural circuits is essential for an animal's ability to learn from its environment while preserving proper processing and perception of sensory information. However, unlike the mechanisms that drive plasticity in neural circuits, the activity-induced molecular mechanisms that convey functional stability remain poorly understood. Focusing on the visual cortex of adult mice and combining transcriptomics, electrophysiology, and in vivo calcium imaging, we find that the daily appearance of light induces, in excitatory neurons, a large gene program along with rapid and transient increases in the ratio of excitation and inhibition (E/I ratio) and neural activity. Furthermore, we find that the light-induced transcription factor NPAS4 drives these daily normalizations of the E/I ratio and neural activity rates and that it stabilizes the neurons' response properties. These findings indicate that daily sensory-induced transcription normalizes the E/I ratio and drives downward firing rate homeostasis to maintain proper sensory processing and perception.
Collapse
Affiliation(s)
- Dahlia Kushinsky
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Emmanouil Tsivourakis
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Daniella Apelblat
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Roethler
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | | | - Katayun Cohen-Kashi Malina
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Ivo Spiegel
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
5
|
Bian Y, Kawabata R, Enwright JF, Tsubomoto M, Okuda T, Kamikawa K, Kimoto S, Kikuchi M, Lewis DA, Hashimoto T. Expression of activity-regulated transcripts in pyramidal neurons across the cortical visuospatial working memory network in unaffected comparison individuals and individuals with schizophrenia. Psychiatry Res 2024; 339:116084. [PMID: 39033685 DOI: 10.1016/j.psychres.2024.116084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
Visuospatial working memory (vsWM), which is impaired in schizophrenia (SZ), is mediated by multiple cortical regions including the primary (V1) and association (V2) visual, posterior parietal (PPC) and dorsolateral prefrontal (DLPFC) cortices. In these regions, parvalbumin (PV) or somatostatin (SST) GABA neurons are altered in SZ as reflected in lower levels of activity-regulated transcripts. As PV and SST neurons receive excitatory inputs from neighboring pyramidal neurons, we hypothesized that levels of activity-regulated transcripts are also lower in pyramidal neurons in these regions. Thus, we quantified levels of four activity-regulated, pyramidal neuron-selective transcripts, namely adenylate cyclase-activating polypeptide-1 (ADCYAP1), brain-derived neurotrophic factor (BDNF), neuronal pentraxin-2 (NPTX2) and neuritin-1 (NRN1) mRNAs, in V1, V2, PPC and DLPFC from unaffected comparison and SZ individuals. In SZ, BDNF and NPTX2 mRNA levels were lower across all four regions, whereas ADCYAP1 and NRN1 mRNA levels were lower in V1 and V2. The regional pattern of deficits in BDNF and NPTX2 mRNAs was similar to that in transcripts in PV and SST neurons in SZ. These findings suggest that lower activity of pyramidal neurons expressing BDNF and/or NPTX2 mRNAs might contribute to alterations in PV and SST neurons across the vsWM network in SZ.
Collapse
Affiliation(s)
- Yufan Bian
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - John F Enwright
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Takeshi Okuda
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan; Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan; Research Center for Child Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; National Hospital Organization Hokuriku Hospital, Nanto, 939-1893, Japan.
| |
Collapse
|
6
|
McCarthy CI, Kavalali ET. Nano-organization of synaptic calcium signaling. Biochem Soc Trans 2024; 52:1459-1471. [PMID: 38752834 PMCID: PMC11346461 DOI: 10.1042/bst20231385] [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: 03/04/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 06/27/2024]
Abstract
Recent studies suggest an exquisite structural nano-organization within single synapses, where sites of evoked fusion - marked by clustering of synaptic vesicles, active zone proteins and voltage-gated calcium channels - are directly juxtaposed to postsynaptic receptor clusters within nanocolumns. This direct nanometer scale alignment between presynaptic fusion apparatus and postsynaptic receptors is thought to ensure the fidelity of synaptic signaling and possibly allow multiple distinct signals to occur without interference from each other within a single active zone. The functional specificity of this organization is made possible by the inherent nano-organization of calcium signals, where all the different calcium sources such as voltage-gated calcium channels, intracellular stores and store-operated calcium entry have dedicated local targets within their nanodomain to ensure precision of action. Here, we discuss synaptic nano-organization from the perspective of calcium signals, where some of the principal findings from early work in the 1980s continue to inspire current studies that exploit new genetic tools and super-resolution imaging technologies.
Collapse
Affiliation(s)
- Clara I. McCarthy
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
| |
Collapse
|
7
|
Concina G, Milano L, Renna A, Manassero E, Stabile F, Sacchetti B. Hippocampus-to-amygdala pathway drives the separation of remote memories of related events. Cell Rep 2024; 43:114151. [PMID: 38656872 DOI: 10.1016/j.celrep.2024.114151] [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: 05/24/2023] [Revised: 02/21/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
The mammalian brain can store and retrieve memories of related events as distinct memories and remember common features of those experiences. How it computes this function remains elusive. Here, we show in rats that recent memories of two closely timed auditory fear events share overlapping neuronal ensembles in the basolateral amygdala (BLA) and are functionally linked. However, remote memories have reduced neuronal overlap and are functionally independent. The activity of parvalbumin (PV)-expressing neurons in the BLA plays a crucial role in forming separate remote memories. Chemogenetic blockade of PV preserves individual remote memories but prevents their segregation, resulting in reciprocal associations. The hippocampus drives this process through specific excitatory connections with BLA GABAergic interneurons. These findings provide insights into the neuronal mechanisms that minimize the overlap between distinct remote memories and enable the retrieval of related memories separately.
Collapse
Affiliation(s)
- Giulia Concina
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy
| | - Luisella Milano
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy
| | - Annamaria Renna
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy
| | - Eugenio Manassero
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy
| | - Francesca Stabile
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy
| | - Benedetto Sacchetti
- Rita Levi-Montalcini Department of Neuroscience, University of Turin, 10125 Turin, Italy.
| |
Collapse
|
8
|
Izumi H, Demura M, Imai A, Ogawa R, Fukuchi M, Okubo T, Tabata T, Mori H, Yoshida T. Developmental synapse pathology triggered by maternal exposure to the herbicide glufosinate ammonium. Front Mol Neurosci 2023; 16:1298238. [PMID: 38098940 PMCID: PMC10720911 DOI: 10.3389/fnmol.2023.1298238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Environmental and genetic factors influence synapse formation. Numerous animal experiments have revealed that pesticides, including herbicides, can disturb normal intracellular signals, gene expression, and individual animal behaviors. However, the mechanism underlying the adverse outcomes of pesticide exposure remains elusive. Herein, we investigated the effect of maternal exposure to the herbicide glufosinate ammonium (GLA) on offspring neuronal synapse formation in vitro. Cultured cerebral cortical neurons prepared from mouse embryos with maternal GLA exposure demonstrated impaired synapse formation induced by synaptic organizer neuroligin 1 (NLGN1)-coated beads. Conversely, the direct administration of GLA to the neuronal cultures exhibited negligible effect on the NLGN1-induced synapse formation. The comparison of the transcriptomes of cultured neurons from embryos treated with maternal GLA or vehicle and a subsequent bioinformatics analysis of differentially expressed genes (DEGs) identified "nervous system development," including "synapse," as the top-ranking process for downregulated DEGs in the GLA group. In addition, we detected lower densities of parvalbumin (Pvalb)-positive neurons at the postnatal developmental stage in the medial prefrontal cortex (mPFC) of offspring born to GLA-exposed dams. These results suggest that maternal GLA exposure induces synapse pathology, with alterations in the expression of genes that regulate synaptic development via an indirect pathway distinct from the effect of direct GLA action on neurons.
Collapse
Affiliation(s)
- Hironori Izumi
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Maina Demura
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Ayako Imai
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Ryohei Ogawa
- Department of Radiology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Mamoru Fukuchi
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, Gunma, Japan
| | - Taisaku Okubo
- Laboratory for Biological Information Processing, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Toshihide Tabata
- Laboratory for Biological Information Processing, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Research Center for Pre-Disease Science, University of Toyama, Toyama, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| |
Collapse
|
9
|
Ma H, Khaled HG, Wang X, Mandelberg NJ, Cohen SM, He X, Tsien RW. Excitation-transcription coupling, neuronal gene expression and synaptic plasticity. Nat Rev Neurosci 2023; 24:672-692. [PMID: 37773070 DOI: 10.1038/s41583-023-00742-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Excitation-transcription coupling (E-TC) links synaptic and cellular activity to nuclear gene transcription. It is generally accepted that E-TC makes a crucial contribution to learning and memory through its role in underpinning long-lasting synaptic enhancement in late-phase long-term potentiation and has more recently been linked to late-phase long-term depression: both processes require de novo gene transcription, mRNA translation and protein synthesis. E-TC begins with the activation of glutamate-gated N-methyl-D-aspartate-type receptors and voltage-gated L-type Ca2+ channels at the membrane and culminates in the activation of transcription factors in the nucleus. These receptors and ion channels mediate E-TC through mechanisms that include long-range signalling from the synapse to the nucleus and local interactions within dendritic spines, among other possibilities. Growing experimental evidence links these E-TC mechanisms to late-phase long-term potentiation and learning and memory. These advances in our understanding of the molecular mechanisms of E-TC mean that future efforts can focus on understanding its mesoscale functions and how it regulates neuronal network activity and behaviour in physiological and pathological conditions.
Collapse
Affiliation(s)
- Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China.
| | - Houda G Khaled
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Xiaohan Wang
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Nataniel J Mandelberg
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Samuel M Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
10
|
Guo Z, Ni H, Cui Z, Zhu Z, Kang J, Wang D, Ke Z. Pain sensitivity related to gamma oscillation of parvalbumin interneuron in primary somatosensory cortex in Dync1i1 -/- mice. Neurobiol Dis 2023:106170. [PMID: 37257662 DOI: 10.1016/j.nbd.2023.106170] [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: 02/08/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023] Open
Abstract
Cytoplasmic dynein is an important intracellular motor protein that plays an important role in neuronal growth, axonal polarity formation, dendritic differentiation, and dendritic spine development among others. The intermediate chain of dynein, encoded by Dync1i1, plays a vital role in the dynein complex. Therefore, we assessed the behavioral and related neuronal activities in mice with dync1i1 gene knockout. Neuronal activities in primary somatosensory cortex were recorded by in vivo electrophysiology and manipulated by optogenetic and chemogenetics. Nociception of mechanical, thermal, and cold pain in Dync1i1-/- mice were impaired. The activities of parvalbumin (PV) interneurons and gamma oscillation in primary somatosensory were also impaired when exposed to mechanical nociceptive stimulation. This neuronal dysfunction was rescued by optogenetic activation of PV neurons in Dync1i1-/- mice, and mimicked by suppressing PV neurons using chemogenetics in WT mice. Impaired pain sensations in Dync1i1-/- mice were correlated with impaired gamma oscillations due to a loss of interneurons, especially the PV type. This genotype-driven approach revealed an association between impaired pain sensation and cytoplasmic dynein complex.
Collapse
Affiliation(s)
- Zhongzhao Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong Ni
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengyu Cui
- Department of Traditional Chinese Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 201203, China
| | - Zilu Zhu
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiansheng Kang
- Clinical Systems Biology Laboratories East District of The first affiliated hospital of ZhengZhou University, Zhengzhou 450018, China
| | - Deheng Wang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zunji Ke
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
11
|
Malik AM, Wu JJ, Gillies CA, Doctrove QA, Li X, Huang H, Tank EHM, Shakkottai VG, Barmada S. Neuronal activity regulates Matrin 3 abundance and function in a calcium-dependent manner through calpain-mediated cleavage and calmodulin binding. Proc Natl Acad Sci U S A 2023; 120:e2206217120. [PMID: 37011198 PMCID: PMC10104577 DOI: 10.1073/pnas.2206217120] [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: 04/08/2022] [Accepted: 02/14/2023] [Indexed: 04/05/2023] Open
Abstract
RNA-binding protein (RBP) dysfunction is a fundamental hallmark of amyotrophic lateral sclerosis (ALS) and related neuromuscular disorders. Abnormal neuronal excitability is also a conserved feature in ALS patients and disease models, yet little is known about how activity-dependent processes regulate RBP levels and functions. Mutations in the gene encoding the RBP Matrin 3 (MATR3) cause familial disease, and MATR3 pathology has also been observed in sporadic ALS, suggesting a key role for MATR3 in disease pathogenesis. Here, we show that glutamatergic activity drives MATR3 degradation through an NMDA receptor-, Ca2+-, and calpain-dependent mechanism. The most common pathogenic MATR3 mutation renders it resistant to calpain degradation, suggesting a link between activity-dependent MATR3 regulation and disease. We also demonstrate that Ca2+ regulates MATR3 through a nondegradative process involving the binding of Ca2+/calmodulin to MATR3 and inhibition of its RNA-binding ability. These findings indicate that neuronal activity impacts both the abundance and function of MATR3, underscoring the effect of activity on RBPs and providing a foundation for further study of Ca2+-coupled regulation of RBPs implicated in ALS and related neurological diseases.
Collapse
Affiliation(s)
- Ahmed M. Malik
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
| | - Josephine J. Wu
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI48109
| | - Christie A. Gillies
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI48109
| | - Quinlan A. Doctrove
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Postbac Research Education Program, University of Michigan, Ann Arbor, MI48109
| | - Xingli Li
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
| | - Haoran Huang
- University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | | | - Sami Barmada
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
12
|
Ryner RF, Derera ID, Armbruster M, Kansara A, Sommer ME, Pirone A, Noubary F, Jacob M, Dulla CG. Cortical Parvalbumin-Positive Interneuron Development and Function Are Altered in the APC Conditional Knockout Mouse Model of Infantile and Epileptic Spasms Syndrome. J Neurosci 2023; 43:1422-1440. [PMID: 36717229 PMCID: PMC9987578 DOI: 10.1523/jneurosci.0572-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/14/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023] Open
Abstract
Infantile and epileptic spasms syndrome (IESS) is a childhood epilepsy syndrome characterized by infantile or late-onset spasms, abnormal neonatal EEG, and epilepsy. Few treatments exist for IESS, clinical outcomes are poor, and the molecular and circuit-level etiologies of IESS are not well understood. Multiple human IESS risk genes are linked to Wnt/β-catenin signaling, a pathway that controls developmental transcriptional programs and promotes glutamatergic excitation via β-catenin's role as a synaptic scaffold. We previously showed that deleting adenomatous polyposis coli (APC), a component of the β-catenin destruction complex, in excitatory neurons (APC cKO mice, APCfl/fl x CaMKIIαCre) increased β-catenin levels in developing glutamatergic neurons and led to infantile behavioral spasms, abnormal neonatal EEG, and adult epilepsy. Here, we tested the hypothesis that the development of GABAergic interneurons (INs) is disrupted in APC cKO male and female mice. IN dysfunction is implicated in human IESS, is a feature of other rodent models of IESS, and may contribute to the manifestation of spasms and seizures. We found that parvalbumin-positive INs (PV+ INs), an important source of cortical inhibition, were decreased in number, underwent disproportionate developmental apoptosis, and had altered dendrite morphology at P9, the peak of behavioral spasms. PV+ INs received excessive excitatory input, and their intrinsic ability to fire action potentials was reduced at all time points examined (P9, P14, P60). Subsequently, GABAergic transmission onto pyramidal neurons was uniquely altered in the somatosensory cortex of APC cKO mice at all ages, with both decreased IPSC input at P14 and enhanced IPSC input at P9 and P60. These results indicate that inhibitory circuit dysfunction occurs in APC cKOs and, along with known changes in excitation, may contribute to IESS-related phenotypes.SIGNIFICANCE STATEMENT Infantile and epileptic spasms syndrome (IESS) is a devastating epilepsy with limited treatment options and poor clinical outcomes. The molecular, cellular, and circuit disruptions that cause infantile spasms and seizures are largely unknown, but inhibitory GABAergic interneuron dysfunction has been implicated in rodent models of IESS and may contribute to human IESS. Here, we use a rodent model of IESS, the APC cKO mouse, in which β-catenin signaling is increased in excitatory neurons. This results in altered parvalbumin-positive GABAergic interneuron development and GABAergic synaptic dysfunction throughout life, showing that pathology arising in excitatory neurons can initiate long-term interneuron dysfunction. Our findings further implicate GABAergic dysfunction in IESS, even when pathology is initiated in other neuronal types.
Collapse
Affiliation(s)
- Rachael F Ryner
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
- Cell, Molecular, and Developmental Biology Graduate Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts 02111
| | - Isabel D Derera
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Anar Kansara
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Mary E Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Antonella Pirone
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Farzad Noubary
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115
| | - Michele Jacob
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
13
|
Rigter PMF, de Konink C, van Woerden GM. Loss of CAMK2G affects intrinsic and motor behavior but has minimal impact on cognitive behavior. Front Neurosci 2023; 16:1086994. [PMID: 36685241 PMCID: PMC9853378 DOI: 10.3389/fnins.2022.1086994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/07/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction The gamma subunit of calcium/calmodulin-dependent protein kinase 2 (CAMK2G) is expressed throughout the brain and is associated with neurodevelopmental disorders. Research on the role of CAMK2G is limited and attributes different functions to specific cell types. Methods To further expand on the role of CAMK2G in brain functioning, we performed extensive phenotypic characterization of a Camk2g knockout mouse. Results We found different CAMK2G isoforms that show a distinct spatial expression pattern in the brain. Additionally, based on our behavioral characterization, we conclude that CAMK2G plays a minor role in hippocampus-dependent learning and synaptic plasticity. Rather, we show that CAMK2G is required for motor function and that the loss of CAMK2G results in impaired nest-building and marble burying behavior, which are innate behaviors that are associated with impaired neurodevelopment. Discussion Taken together, our results provide evidence for a unique function of this specific CAMK2 isozyme in the brain and further support the role of CAMK2G in neurodevelopment.
Collapse
Affiliation(s)
- Pomme M. F. Rigter
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
| | - Charlotte de Konink
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Geeske M. van Woerden
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
14
|
Miranda JM, Cruz E, Bessières B, Alberini CM. Hippocampal parvalbumin interneurons play a critical role in memory development. Cell Rep 2022; 41:111643. [PMID: 36384113 PMCID: PMC9737056 DOI: 10.1016/j.celrep.2022.111643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/16/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Episodic memories formed in early childhood rapidly decay, but their latent traces remain stored long term. These memories require the dorsal hippocampus (dHPC) and seem to undergo a developmental critical period. It remains to be determined whether the maturation of parvalbumin interneurons (PVIs), a major mechanism of critical periods, contributes to memory development. Here, we show that episodic infantile learning significantly increases the levels of parvalbumin in the dHPC 48 h after training. Chemogenetic inhibition of PVIs before learning indicated that these neurons are required for infantile memory formation. A bilateral dHPC injection of the γ-aminobutyric acid type A receptor agonist diazepam after training elicited long-term memory expression in infant rats, although direct PVI chemogenetic activation had no effect. Finally, PVI activity was required for brain-derived neurotrophic factor (BDNF)-dependent maturation of memory competence, i.e., adult-like long-term memory expression. Thus, dHPC PVIs are critical for the formation of infantile memories and for memory development.
Collapse
Affiliation(s)
- Janelle M Miranda
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Emmanuel Cruz
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Benjamin Bessières
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
15
|
Kimoto S, Hashimoto T, Berry KJ, Tsubomoto M, Yamaguchi Y, Enwright JF, Chen K, Kawabata R, Kikuchi M, Kishimoto T, Lewis DA. Expression of actin- and oxidative phosphorylation-related transcripts across the cortical visuospatial working memory network in unaffected comparison and schizophrenia subjects. Neuropsychopharmacology 2022; 47:2061-2070. [PMID: 35034100 PMCID: PMC9556568 DOI: 10.1038/s41386-022-01274-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 11/09/2022]
Abstract
Visuospatial working memory (vsWM), which is impaired in schizophrenia (SZ), is mediated by a distributed cortical network. In one node of this network, the dorsolateral prefrontal cortex (DLPFC), altered expression of transcripts for actin assembly and mitochondrial oxidative phosphorylation (OXPHOS) have been reported in SZ. To understand the relationship between these processes, and the extent to which similar alterations are present in other regions of vsWM network in SZ, a subset of actin- (CDC42, BAIAP2, ARPC3, and ARPC4) and OXPHOS-related (ATP5H, COX4I1, COX7B, and NDUFB3) transcripts were quantified in DLPFC by RNA sequencing in 139 SZ and unaffected comparison (UC) subjects, and in DLPFC and three other regions of the cortical vsWM network by qPCR in 20 pairs of SZ and UC subjects. By RNA sequencing, levels of actin- and OXPHOS-related transcripts were significantly altered in SZ, and robustly correlated in both UC and SZ subject groups. By qPCR, cross-regional expression patterns of these transcripts in UC subjects were consistent with greater actin assembly in DLPFC and higher OXPHOS activity in primary visual cortex (V1). In SZ, CDC42 and ARPC4 levels were lower in all regions, BAIAP2 levels higher only in V1, and ARPC3 levels unaltered across regions. All OXPHOS-related transcript levels were lower in SZ, with the disease effect decreasing from posterior to anterior regions. The differential alterations in markers of actin assembly and energy production across regions of the cortical vsWM network in SZ suggest that each region may make specific contributions to vsWM impairments in the illness.
Collapse
Affiliation(s)
- Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Research Center for Child Development, Kanazawa University, Kanazawa, 920-8640, Japan
| | - Kimberly J Berry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yasunari Yamaguchi
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - John F Enwright
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kehui Chen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Research Center for Child Development, Kanazawa University, Kanazawa, 920-8640, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
16
|
Glucocorticoid-Regulated Kinase CAMKIγ in the Central Amygdala Controls Anxiety-like Behavior in Mice. Int J Mol Sci 2022; 23:ijms232012328. [PMID: 36293185 PMCID: PMC9604347 DOI: 10.3390/ijms232012328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
The expression of the Calcium/Calmodulin-Dependent Protein Kinase I gamma (encoded by the Camk1g gene) depends on the activation of glucocorticoid receptors (GR) and is strongly regulated by stress. Since Camk1g is primarily expressed in neuronal cells of the limbic system in the brain, we hypothesized that it could be involved in signaling mechanisms that underlie the adaptive or maladaptive responses to stress. Here, we find that restraint-induced stress and the GR agonist dexamethasone robustly increase the expression of Camk1g in neurons of the amygdalar nuclei in the mouse brain. To assess the functional role of Camk1g expression, we performed a virally induced knock-down of the transcript. Mice with bilateral amygdala-specific Camk1g knock-down showed increased anxiety-like behaviors in the light-dark box, and an increase in freezing behavior after fear-conditioning, but normal spatial working memory during exploration of a Y-maze. Thus, we confirm that Camk1g is a neuron-specific GR-regulated transcript, and show that it is specifically involved in behaviors related to anxiety, as well as responses conditioned by aversive stimuli.
Collapse
|
17
|
Suzuki Y, Kurata T, Koide T, Okada I, Nakajima N, Imaizumi Y, Yamamura H. Local Ca<sup>2+</sup> Signals within Caveolae Cause Nuclear Translocation of CaMK1α in Mouse Vascular Smooth Muscle Cells. Biol Pharm Bull 2022; 45:1354-1363. [DOI: 10.1248/bpb.b22-00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshiaki Suzuki
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tomo Kurata
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tsukasa Koide
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Itsuki Okada
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Nanami Nakajima
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Yuji Imaizumi
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Hisao Yamamura
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
18
|
Mancini M, Patel JC, Affinati AH, Witkovsky P, Rice ME. Leptin Promotes Striatal Dopamine Release via Cholinergic Interneurons and Regionally Distinct Signaling Pathways. J Neurosci 2022; 42:6668-6679. [PMID: 35906070 PMCID: PMC9436012 DOI: 10.1523/jneurosci.0238-22.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
Dopamine (DA) is a critical regulator of striatal network activity and is essential for motor activation and reward-associated behaviors. Previous work has shown that DA is influenced by the reward value of food, as well as by hormonal factors that reguate food intake and energy expenditure. Changes in striatal DA signaling also have been linked to aberrant eating patterns. Here we test the effect of leptin, an adipocyte-derived hormone involved in feeding and energy homeostasis regulation, on striatal DA release and uptake. Immunohistochemical evaluation identified leptin receptor (LepR) expression throughout mouse striatum, including on striatal cholinergic interneurons (ChIs) and their extensive processes. Using fast-scan cyclic voltammetry (FSCV), we found that leptin causes a concentration-dependent increase in evoked extra-cellular DA concentration ([DA]o) in dorsal striatum (dStr) and nucleus accumbens (NAc) core and shell in male mouse striatal slices, and also an increase in the rate of DA uptake. Further, we found that leptin increases ChI excitability, and that the enhancing effect of leptin on evoked [DA]o is lost when nicotinic acetylcholine (ACh) receptors are antagonized or when examined in striatal slices from mice lacking ACh synthesis. Evaluation of signaling pathways underlying leptin's action revealed a requirement for intracellular Ca2+, and the involvement of different downstream pathways in dStr and NAc core versus NAc shell. These results provide the first evidence for dynamic regulation of DA release and uptake by leptin within brain motor and reward pathways, and highlight the involvement of ChIs in this process.SIGNIFICANCE STATEMENT Given the importance of striatal dopamine (DA) in reward, motivation, motor behavior and food intake, identifying the actions of metabolic hormones on DA release in striatal subregions should provide new insight into factors that influence DA-dependent motivated behaviors. We find that one of these hormones, leptin, boosts striatal DA release through a process involving striatal cholinergic interneurons (ChIs) and nicotinic acetylcholine (ACh) receptors. Moreover, we find that the intracellular cascades downstream from leptin receptor (LepR) activation that lead to enhanced DA release differ among striatal subregions. Thus, we not only show that leptin regulates DA release, but also identify characteristics of this process that could be harnessed to alter pathologic eating behaviors.
Collapse
Affiliation(s)
- Maria Mancini
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York 10016
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
| | - Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| | - Alison H Affinati
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| | - Margaret E Rice
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York 10016
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, New York 10016
| |
Collapse
|
19
|
Zhao A, Liu N, Jiang G, Xu L, Yao M, Zhang Y, Xue B, Ma B, Chang D, Feng Y, Jiang Y, Liu J, Zhou G. Combination of panax ginseng and ginkgo biloba extracts attenuate cerebral ischemia injury with modulation of NLRP3 inflammasome and CAMK4/CREB pathway. Front Pharmacol 2022; 13:980449. [PMID: 36091745 PMCID: PMC9452960 DOI: 10.3389/fphar.2022.980449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/03/2022] [Indexed: 11/22/2022] Open
Abstract
Stroke is a major cause of death and disability throughout the world. A combination of Panax Ginseng and Ginkgo biloba extracts (CGGE) is an effective treatment for nervous system diseases, but the neuroprotective mechanism underlying CGGE remains unclear. Both network analysis and experimental research were employed to explore the potential mechanism of CGGE in treating ischemic stroke (IS). Network analysis identified a total number of 133 potential targets for 34 active ingredients and 239 IS-related targets. What’s more, several processes that might involve the regulation of CGGE against IS were identified, including long-term potentiation, cAMP signaling pathway, neurotrophin signaling pathway, and Nod-like receptor signaling pathway. Our studies in animal models suggested that CGGE could reduce inflammatory response by inhibiting the activity of Nod-like receptor, pyrin containing 3 (NLRP3) inflammasome, and maintain the balance of glutamate (Glu)/gamma-aminobutyric acid (GABA) via activating calmodulin-dependent protein kinase type Ⅳ (CAMK4)/cyclic AMP-responsive element-binding protein (CREB) pathway. These findings indicated the neuroprotective effects of CGGE, possibly improving neuroinflammation and excitotoxicity by regulating the NLRP3 inflammasome and CAMK4/CREB pathway.
Collapse
Affiliation(s)
- Aimei Zhao
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Liu
- Beijing Increasepharm Safety and Efficacy Co. Ltd., Beijing, China
| | - Guozhi Jiang
- Shineway Pharmaceutical Group Co. Ltd., Shijiazhuang, China
| | - Li Xu
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingjie Xue
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Ma
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Dennis Chang
- NICM, Western Sydney University, Penrith, NSW, Australia
| | - Yujing Feng
- Department of Anesthesiology, Punan Hospital, Shanghai, China
| | - Yunyao Jiang
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
- *Correspondence: Yunyao Jiang, ; Jianxun Liu, ; Guoping Zhou,
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunyao Jiang, ; Jianxun Liu, ; Guoping Zhou,
| | - Guoping Zhou
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Yunyao Jiang, ; Jianxun Liu, ; Guoping Zhou,
| |
Collapse
|
20
|
Altered Development of Prefrontal GABAergic Functions and Anxiety-like Behavior in Adolescent Offspring Induced by Prenatal Stress. Brain Sci 2022; 12:brainsci12081015. [PMID: 36009078 PMCID: PMC9406165 DOI: 10.3390/brainsci12081015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Maternal stress can afflict fetal brain development, putting the offspring at risk of cognitive deficits, including anxiety. The prefrontal cortex (PFC), a protracted maturing region, is notably affected by prenatal stress (PS). However, it remains unclear how PS interferes with the maturation of the GABAergic system, considering its functional adjustment in the PFC during adolescence. The present study thus investigated the long-lasting consequences of PS on the prefrontal GABAergic functions of adolescent offspring. Pregnant Sprague–Dawley rats were divided into controls and the PS group, which underwent restraint stress during the last week of gestation. Male pups from postnatal days (PND) 40–42 were submitted to the elevated plus maze (EPM) test. Proteins essentially involved in GABAergic signaling were then examined in PFC tissues, including the K+-Cl− cotransporter (KCC2), Na+-K+-Cl− cotransporter (NKCC1), α1 and α5 subunits of GABA type A receptors (GABAA receptors), and parvalbumin (PV), along with cAMP response element-binding protein phosphorylation (pCREB), which reacts in the plasticity regulation of PV-positive interneurons. The results revealed that the higher anxiety-like behavior of PS adolescent rats concurred with the significant decreases of the KCC2 and α1 subunits, with PV- and pCREB-lowered levels. The findings suggested that PS disrupts the continuance of PFC maturity by reducing the essential elements of GABAergic functions. These changes likely underlie the anxiety emerging in adolescence, possibly progressing to mental disorders.
Collapse
|
21
|
Pumo GM, Kitazawa T, Rijli FM. Epigenetic and Transcriptional Regulation of Spontaneous and Sensory Activity Dependent Programs During Neuronal Circuit Development. Front Neural Circuits 2022; 16:911023. [PMID: 35664458 PMCID: PMC9158562 DOI: 10.3389/fncir.2022.911023] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Spontaneous activity generated before the onset of sensory transduction has a key role in wiring developing sensory circuits. From axonal targeting, to synapse formation and elimination, to the balanced integration of neurons into developing circuits, this type of activity is implicated in a variety of cellular processes. However, little is known about its molecular mechanisms of action, especially at the level of genome regulation. Conversely, sensory experience-dependent activity implements well-characterized transcriptional and epigenetic chromatin programs that underlie heterogeneous but specific genomic responses that shape both postnatal circuit development and neuroplasticity in the adult. In this review, we focus on our knowledge of the developmental processes regulated by spontaneous activity and the underlying transcriptional mechanisms. We also review novel findings on how chromatin regulates the specificity and developmental induction of the experience-dependent program, and speculate their relevance for our understanding of how spontaneous activity may act at the genomic level to instruct circuit assembly and prepare developing neurons for sensory-dependent connectivity refinement and processing.
Collapse
Affiliation(s)
- Gabriele M. Pumo
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| | - Taro Kitazawa
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Filippo M. Rijli
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
22
|
A molecular complex of Ca v1.2/CaMKK2/CaMK1a in caveolae is responsible for vascular remodeling via excitation-transcription coupling. Proc Natl Acad Sci U S A 2022; 119:e2117435119. [PMID: 35412911 PMCID: PMC9169798 DOI: 10.1073/pnas.2117435119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excitation–transcription (E-T) coupling can initiate and modulate essential physiological or pathological responses in cells, such as neurons and cardiac myocytes. Although vascular myocytes also exhibit E-T coupling in response to membrane depolarization, the underlying molecular mechanisms are unknown. Our study reveals that E-T coupling in vascular myocytes converts intracellular Ca2+ signals into selective gene transcription related to chemotaxis, leukocyte adhesion, and inflammation. Our discovery identifies a mechanism for vascular remodeling as an adaptation to increased circumferential stretch. Elevation of intracellular Ca2+ concentration ([Ca2+]i) activates Ca2+/calmodulin-dependent kinases (CaMK) and promotes gene transcription. This signaling pathway is referred to as excitation–transcription (E-T) coupling. Although vascular myocytes can exhibit E-T coupling, the molecular mechanisms and physiological/pathological roles are unknown. Multiscale analysis spanning from single molecules to whole organisms has revealed essential steps in mouse vascular myocyte E-T coupling. Upon a depolarizing stimulus, Ca2+ influx through Cav1.2 voltage-dependent Ca2+ channels activates CaMKK2 and CaMK1a, resulting in intranuclear CREB phosphorylation. Within caveolae, the formation of a molecular complex of Cav1.2/CaMKK2/CaMK1a is promoted in vascular myocytes. Live imaging using a genetically encoded Ca2+ indicator revealed direct activation of CaMKK2 by Ca2+ influx through Cav1.2 localized to caveolae. CaMK1a is phosphorylated by CaMKK2 at caveolae and translocated to the nucleus upon membrane depolarization. In addition, sustained depolarization of a mesenteric artery preparation induced genes related to chemotaxis, leukocyte adhesion, and inflammation, and these changes were reversed by inhibitors of Cav1.2, CaMKK2, and CaMK, or disruption of caveolae. In the context of pathophysiology, when the mesenteric artery was loaded by high pressure in vivo, we observed CREB phosphorylation in myocytes, macrophage accumulation at adventitia, and an increase in thickness and cross-sectional area of the tunica media. These changes were reduced in caveolin1-knockout mice or in mice treated with the CaMKK2 inhibitor STO609. In summary, E-T coupling depends on Cav1.2/CaMKK2/CaMK1a localized to caveolae, and this complex converts [Ca2+]i changes into gene transcription. This ultimately leads to macrophage accumulation and media remodeling for adaptation to increased circumferential stretch.
Collapse
|
23
|
Li KT, He X, Zhou G, Yang J, Li T, Hu H, Ji D, Zhou C, Ma H. Rational designing of oscillatory rhythmicity for memory rescue in plasticity-impaired learning networks. Cell Rep 2022; 39:110678. [PMID: 35417714 DOI: 10.1016/j.celrep.2022.110678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/19/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022] Open
Abstract
In the brain, oscillatory strength embedded in network rhythmicity is important for processing experiences, and this process is disrupted in certain psychiatric disorders. The use of rhythmic network stimuli can change these oscillations and has shown promise in terms of improving cognitive function, although the underlying mechanisms are poorly understood. Here, we combine a two-layer learning model, with experiments involving genetically modified mice, that provides precise control of experience-driven oscillations by manipulating long-term potentiation of excitatory synapses onto inhibitory interneurons (LTPE→I). We find that, in the absence of LTPE→I, impaired network dynamics and memory are rescued by activating inhibitory neurons to augment the power in theta and gamma frequencies, which prevents network overexcitation with less inhibitory rebound. In contrast, increasing either theta or gamma power alone was less effective. Thus, inducing network changes at dual frequencies is involved in memory encoding, indicating a potentially feasible strategy for optimizing network-stimulating therapies.
Collapse
Affiliation(s)
- Kwan Tung Li
- Department of Physics, Centre for Nonlinear Studies, Beijing-Hong Kong-Singapore Joint Centre for Nonlinear and Complex Systems (Hong Kong), Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong, China
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Guangjun Zhou
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jing Yang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hailan Hu
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Daoyun Ji
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Changsong Zhou
- Department of Physics, Centre for Nonlinear Studies, Beijing-Hong Kong-Singapore Joint Centre for Nonlinear and Complex Systems (Hong Kong), Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong, China; Department of Physics, Zhejiang University, Hangzhou 310027, China.
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
24
|
He X, Wang Y, Zhou G, Yang J, Li J, Li T, Hu H, Ma H. A Critical Role for γCaMKII in Decoding NMDA Signaling to Regulate AMPA Receptors in Putative Inhibitory Interneurons. Neurosci Bull 2022; 38:916-926. [PMID: 35290589 PMCID: PMC9352831 DOI: 10.1007/s12264-022-00840-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
CaMKII is essential for long-term potentiation (LTP), a process in which synaptic strength is increased following the acquisition of information. Among the four CaMKII isoforms, γCaMKII is the one that mediates the LTP of excitatory synapses onto inhibitory interneurons (LTPE→I). However, the molecular mechanism underlying how γCaMKII mediates LTPE→I remains unclear. Here, we show that γCaMKII is highly enriched in cultured hippocampal inhibitory interneurons and opts to be activated by higher stimulating frequencies in the 10-30 Hz range. Following stimulation, γCaMKII is translocated to the synapse and becomes co-localized with the postsynaptic protein PSD-95. Knocking down γCaMKII prevents the chemical LTP-induced phosphorylation and trafficking of AMPA receptors (AMPARs) in putative inhibitory interneurons, which are restored by overexpression of γCaMKII but not its kinase-dead form. Taken together, these data suggest that γCaMKII decodes NMDAR-mediated signaling and in turn regulates AMPARs for expressing LTP in inhibitory interneurons.
Collapse
Affiliation(s)
- Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yang Wang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Guangjun Zhou
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jing Yang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jiarui Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hailan Hu
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China.
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
25
|
Speigel IA, Hemmings Jr. HC. Relevance of Cortical and Hippocampal Interneuron Functional Diversity to General Anesthetic Mechanisms: A Narrative Review. Front Synaptic Neurosci 2022; 13:812905. [PMID: 35153712 PMCID: PMC8825374 DOI: 10.3389/fnsyn.2021.812905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/30/2021] [Indexed: 01/04/2023] Open
Abstract
General anesthetics disrupt brain processes involved in consciousness by altering synaptic patterns of excitation and inhibition. In the cerebral cortex and hippocampus, GABAergic inhibition is largely mediated by inhibitory interneurons, a heterogeneous group of specialized neuronal subtypes that form characteristic microcircuits with excitatory neurons. Distinct interneuron subtypes regulate specific excitatory neuron networks during normal behavior, but how these interneuron subtypes are affected by general anesthetics is unclear. This narrative review summarizes current principles of the synaptic architecture of cortical and interneuron subtypes, their contributions to different forms of inhibition, and their roles in distinct neuronal microcircuits. The molecular and cellular targets in these circuits that are sensitive to anesthetics are reviewed in the context of how anesthetics impact interneuron function in a subtype-specific manner. The implications of this functional interneuron diversity for mechanisms of anesthesia are discussed, as are their implications for anesthetic-induced changes in neural plasticity and overall brain function.
Collapse
Affiliation(s)
- Iris A. Speigel
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Iris A. Speigel
| | - Hugh C. Hemmings Jr.
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
26
|
Takasu K, Niidome K, Hasegawa M, Ogawa K. Histone Deacetylase Inhibitor Improves the Dysfunction of Hippocampal Gamma Oscillations and Fast Spiking Interneurons in Alzheimer's Disease Model Mice. Front Mol Neurosci 2021; 14:782206. [PMID: 35027883 PMCID: PMC8751405 DOI: 10.3389/fnmol.2021.782206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/01/2021] [Indexed: 12/05/2022] Open
Abstract
The hippocampal gamma oscillation is important for cognitive function, and its deficit is related to cognitive impairment in Alzheimer's disease (AD). Recently, it has been recognized that post-translational modification via histone acetylation is a fundamental molecular mechanism for regulating synaptic plasticity and cognitive function. However, little is known regarding the regulation of hippocampal gamma oscillation by histone acetylation. We investigated whether histone acetylation regulated kainate-induced gamma oscillations and their important regulator, fast-spiking interneurons, using acute hippocampal slices of AD model mice (PSAPP transgenic mice). We found a decrease in kainate-induced gamma oscillations in slices from PSAPP mice, accompanied with the increased activity of fast spiking interneurons in basal state and the decreased activity in activated state. The histone deacetylase (HDAC) inhibitor (SAHA, named vorinostat) restored deficits of gamma oscillation in PSAPP mice, accompanied with rescue of activity of fast spiking interneurons in basal and activated state. The effect of SAHA was different from that of the clinical AD drug donepezil, which rescued only function of fast spiking interneurons in basal state. Besides, activator of nuclear receptor family 4a (NR4a) receptor (cytosporone B), as one of the epigenetic modification related to HDAC inhibition, rescued the deficits in gamma oscillations in PSAPP mice. These results suggested a novel mechanism in which HDAC inhibition improved impairment of gamma oscillations in PSAPP mice by restoring the activity of fast spiking interneurons both in basal and activated state. The reversal of gamma oscillation deficits by HDAC inhibition and/or NR4a activation appears to be a potential therapeutic target for treating cognitive impairment in AD patients.
Collapse
Affiliation(s)
| | | | | | - Koichi Ogawa
- Pain and Neuroscience, Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
27
|
Inhibition in the auditory cortex. Neurosci Biobehav Rev 2021; 132:61-75. [PMID: 34822879 DOI: 10.1016/j.neubiorev.2021.11.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/20/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023]
Abstract
The auditory system provides us with extremely rich and precise information about the outside world. Once a sound reaches our ears, the acoustic information it carries travels from the cochlea all the way to the auditory cortex, where its complexity and nuances are integrated. In the auditory cortex, functional circuits are formed by subpopulations of intermingled excitatory and inhibitory cells. In this review, we discuss recent evidence of the specific contributions of inhibitory neurons in sound processing and integration. We first examine intrinsic properties of three main classes of inhibitory interneurons in the auditory cortex. Then, we describe how inhibition shapes the responsiveness of the auditory cortex to sound. Finally, we discuss how inhibitory interneurons contribute to the sensation and perception of sounds. Altogether, this review points out the crucial role of cortical inhibitory interneurons in integrating information about the context, history, or meaning of a sound. It also highlights open questions to be addressed for increasing our understanding of the staggering complexity leading to the subtlest auditory perception.
Collapse
|
28
|
Ippolito D, Thapliyal S, Glauser DA. Ca 2+/CaM binding to CaMKI promotes IMA-3 importin binding and nuclear translocation in sensory neurons to control behavioral adaptation. eLife 2021; 10:71443. [PMID: 34766550 PMCID: PMC8635976 DOI: 10.7554/elife.71443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Sensory and behavioral plasticity are essential for animals to thrive in changing environments. As key effectors of intracellular calcium signaling, Ca2+/calmodulin-dependent protein kinases (CaMKs) can bridge neural activation with the many regulatory processes needed to orchestrate sensory adaptation, including by relaying signals to the nucleus. Here, we elucidate the molecular mechanism controlling the cell activation-dependent nuclear translocation of CMK-1, the Caenorhabditis elegans ortholog of mammalian CaMKI/IV, in thermosensory neurons in vivo. We show that an intracellular Ca2+ concentration elevation is necessary and sufficient to favor CMK-1 nuclear import. The binding of Ca2+/CaM to CMK-1 increases its affinity for IMA-3 importin, causing a redistribution with a relatively slow kinetics, matching the timescale of sensory adaptation. Furthermore, we show that this mechanism enables the encoding of opposite nuclear signals in neuron types with opposite calcium-responses and that it is essential for experience-dependent behavioral plasticity and gene transcription control in vivo. Since CaMKI/IV are conserved regulators of adaptable behaviors, similar mechanisms could exist in other organisms and for other sensory modalities.
Collapse
Affiliation(s)
- Domenica Ippolito
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Saurabh Thapliyal
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
29
|
Phosphorylation of CREB at Serine 142 and 143 Is Essential for Visual Cortex Plasticity. eNeuro 2021; 8:ENEURO.0217-21.2021. [PMID: 34607805 PMCID: PMC8555886 DOI: 10.1523/eneuro.0217-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 11/21/2022] Open
Abstract
The transcription factor cAMP response element-binding protein (CREB) is involved in a myriad of cellular functions in the central nervous system. For instance, the role of CREB via phosphorylation at the amino-acid residue Serine (Ser)133 in expressing plasticity-related genes and activity-dependent neuronal plasticity processes has been extensively demonstrated. However, much less is known about the role of CREB phosphorylation at Ser142 and Ser143. Here, we employed a viral vector containing a dominant negative form of CREB, with serine-to-alanine mutations at residue 142 and 143 to specifically block phosphorylation at both sites. We then transfected this vector into primary neurons in vitro or intracortically injected it into mice in vivo, to test whether these phosphorylation events were important for activity-dependent plasticity. We demonstrated by immunohistochemistry of cortical neuronal cultures that the expression of Arc, a known plasticity-related gene, requires triple phosphorylation of CREB at Ser133, Ser142, and Ser143. Moreover, we recorded visually-evoked field potentials in awake mice before and after a 7-d period of monocular deprivation (MD) to show that, in addition to CREB phosphorylation at Ser133, ocular dominance plasticity (ODP) in the visual cortex also requires CREB phosphorylation at Ser142/143. Our findings suggest that Ser142/143 phosphorylation is an additional post-translational modification of CREB that triggers the expression of specific target genes and activity-dependent neuronal plasticity processes.
Collapse
|
30
|
Cummings KA, Lacagnina AF, Clem RL. GABAergic microcircuitry of fear memory encoding. Neurobiol Learn Mem 2021; 184:107504. [PMID: 34425220 DOI: 10.1016/j.nlm.2021.107504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 12/30/2022]
Abstract
The paradigm of fear conditioning is largely responsible for our current understanding of how memories are encoded at the cellular level. Its most fundamental underlying mechanism is considered to be plasticity of synaptic connections between excitatory projection neurons (PNs). However, recent studies suggest that while PNs execute critical memory functions, their activity at key stages of learning and recall is extensively orchestrated by a diverse array of GABAergic interneurons (INs). Here we review the contributions of genetically-defined INs to processing of threat-related stimuli in fear conditioning, with a particular focus on how synaptic interactions within interconnected networks of INs modulates PN activity through both inhibition and disinhibition. Furthermore, we discuss accumulating evidence that GABAergic microcircuits are an important locus for synaptic plasticity during fear learning and therefore a viable substrate for long-term memory. These findings suggest that further investigation of INs could unlock unique conceptual insights into the organization and function of fear memory networks.
Collapse
Affiliation(s)
- Kirstie A Cummings
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neurobiology, University of Alabama Birmingham School of Medicine, Birmingham, AL 35294, United States
| | - Anthony F Lacagnina
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Roger L Clem
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
31
|
Tuluc P, Theiner T, Jacobo-Piqueras N, Geisler SM. Role of High Voltage-Gated Ca 2+ Channel Subunits in Pancreatic β-Cell Insulin Release. From Structure to Function. Cells 2021; 10:2004. [PMID: 34440773 PMCID: PMC8393260 DOI: 10.3390/cells10082004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
The pancreatic islets of Langerhans secrete several hormones critical for glucose homeostasis. The β-cells, the major cellular component of the pancreatic islets, secrete insulin, the only hormone capable of lowering the plasma glucose concentration. The counter-regulatory hormone glucagon is secreted by the α-cells while δ-cells secrete somatostatin that via paracrine mechanisms regulates the α- and β-cell activity. These three peptide hormones are packed into secretory granules that are released through exocytosis following a local increase in intracellular Ca2+ concentration. The high voltage-gated Ca2+ channels (HVCCs) occupy a central role in pancreatic hormone release both as a source of Ca2+ required for excitation-secretion coupling as well as a scaffold for the release machinery. HVCCs are multi-protein complexes composed of the main pore-forming transmembrane α1 and the auxiliary intracellular β, extracellular α2δ, and transmembrane γ subunits. Here, we review the current understanding regarding the role of all HVCC subunits expressed in pancreatic β-cell on electrical activity, excitation-secretion coupling, and β-cell mass. The evidence we review was obtained from many seminal studies employing pharmacological approaches as well as genetically modified mouse models. The significance for diabetes in humans is discussed in the context of genetic variations in the genes encoding for the HVCC subunits.
Collapse
Affiliation(s)
- Petronel Tuluc
- Centre for Molecular Biosciences, Department of Pharmacology and Toxicology, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (T.T.); (N.J.-P.); (S.M.G.)
| | | | | | | |
Collapse
|
32
|
Unique dynamics and exocytosis properties of GABAergic synaptic vesicles revealed by three-dimensional single vesicle tracking. Proc Natl Acad Sci U S A 2021; 118:2022133118. [PMID: 33622785 DOI: 10.1073/pnas.2022133118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintaining the balance between neuronal excitation and inhibition is essential for proper function of the central nervous system. Inhibitory synaptic transmission plays an important role in maintaining this balance. Although inhibitory transmission has higher kinetic demands compared to excitatory transmission, its properties are poorly understood. In particular, the dynamics and exocytosis of single inhibitory vesicles have not been investigated, due largely to both technical and practical limitations. Using a combination of quantum dots (QDs) conjugated to antibodies against the luminal domain of the vesicular GABA transporter to selectively label GABAergic (i.e., predominantly inhibitory) vesicles together with dual-focus imaging optics, we tracked the real-time three-dimensional position of single GABAergic vesicles up to the moment of exocytosis (i.e., fusion). Using three-dimensional trajectories, we found that GABAergic synaptic vesicles traveled a shorter distance prior to fusion and had a shorter time to fusion compared to synaptotagmin-1 (Syt1)-labeled vesicles, which were mostly from excitatory neurons. Moreover, our analysis revealed that GABAergic synaptic vesicles move more straightly to their release sites than Syt1-labeled vesicles. Finally, we found that GABAergic vesicles have a higher prevalence of kiss-and-run fusion than Syt1-labeled vesicles. These results indicate that inhibitory synaptic vesicles have a unique set of dynamics and exocytosis properties to support rapid synaptic inhibition, thereby maintaining a tightly regulated coordination between excitation and inhibition in the central nervous system.
Collapse
|
33
|
CaMKIV Signaling Is Not Essential for the Maintenance of Intrinsic or Synaptic Properties in Mouse Visual Cortex. eNeuro 2021; 8:ENEURO.0135-21.2021. [PMID: 34001638 PMCID: PMC8260277 DOI: 10.1523/eneuro.0135-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 12/25/2022] Open
Abstract
Pyramidal neurons in rodent visual cortex homeostatically maintain their firing rates in vivo within a target range. In young cultured rat cortical neurons, Ca2+/calmodulin-dependent kinase IV (CaMKIV) signaling jointly regulates excitatory synaptic strength and intrinsic excitability to allow neurons to maintain their target firing rate. However, the role of CaMKIV signaling in regulating synaptic strength and intrinsic excitability in vivo has not been tested. Here, we show that in pyramidal neurons in visual cortex of juvenile male and female mice, CaMKIV signaling is not essential for the maintenance of basal synaptic or intrinsic properties. Neither CaMKIV conditional knock-down nor viral expression of dominant negative CaMKIV (dnCaMKIV) in vivo disrupts the intrinsic excitability or synaptic input strength of pyramidal neurons in primary visual cortex (V1), and CaMKIV signaling is not required for the increase in intrinsic excitability seen following monocular deprivation (MD). Viral expression of constitutively active CaMKIV (caCaMKIV) in vivo causes a complex disruption of the neuronal input/output function but does not affect synaptic input strength. Taken together, these results demonstrate that although augmented in vivo CaMKIV signaling can alter neuronal excitability, either endogenous CaMKIV signaling is dispensable for maintenance of excitability, or impaired CaMKIV signaling is robustly compensated.
Collapse
|
34
|
Fish KN, Rocco BR, DeDionisio AM, Dienel SJ, Sweet RA, Lewis DA. Altered Parvalbumin Basket Cell Terminals in the Cortical Visuospatial Working Memory Network in Schizophrenia. Biol Psychiatry 2021; 90:47-57. [PMID: 33892915 PMCID: PMC8243491 DOI: 10.1016/j.biopsych.2021.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/21/2021] [Accepted: 02/11/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Visuospatial working memory (vsWM), which is commonly impaired in schizophrenia, involves information processing across the primary visual cortex, association visual cortex, posterior parietal cortex, and dorsolateral prefrontal cortex (DLPFC). Within these regions, vsWM requires inhibition from parvalbumin-expressing basket cells (PVBCs). Here, we analyzed indices of PVBC axon terminals across regions of the vsWM network in schizophrenia. METHODS For 20 matched pairs of subjects with schizophrenia and unaffected comparison subjects, tissue sections from the primary visual cortex, association visual cortex, posterior parietal cortex, and DLPFC were immunolabeled for PV, the 65- and 67-kDa isoforms of glutamic acid decarboxylase (GAD65 and GAD67) that synthesize GABA (gamma-aminobutyric acid), and the vesicular GABA transporter. The density of PVBC terminals and of protein levels per terminal was quantified in layer 3 of each cortical region using fluorescence confocal microscopy. RESULTS In comparison subjects, all measures, except for GAD65 levels, exhibited a caudal-to-rostral decline across the vsWM network. In subjects with schizophrenia, the density of detectable PVBC terminals was significantly lower in all regions except the DLPFC, whereas PVBC terminal levels of PV, GAD67, and GAD65 proteins were lower in all regions. A composite measure of inhibitory strength was lower in subjects with schizophrenia, although the magnitude of the diagnosis effect was greater in the primary visual, association visual, and posterior parietal cortices than in the DLPFC. CONCLUSIONS In schizophrenia, alterations in PVBC terminals across the vsWM network suggest the presence of a shared substrate for cortical dysfunction during vsWM tasks. However, regional differences in the magnitude of the disease effect on an index of PVBC inhibitory strength suggest region-specific alterations in information processing during vsWM tasks.
Collapse
Affiliation(s)
- Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Brad R Rocco
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam M DeDionisio
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samuel J Dienel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Miyamae T, Hashimoto T, Abraham M, Kawabata R, Koshikizawa S, Bian Y, Nishihata Y, Kikuchi M, Ermentrout GB, Lewis DA, Gonzalez-Burgos G. Kcns3 deficiency disrupts Parvalbumin neuron physiology in mouse prefrontal cortex: Implications for the pathophysiology of schizophrenia. Neurobiol Dis 2021; 155:105382. [PMID: 33940180 PMCID: PMC8557947 DOI: 10.1016/j.nbd.2021.105382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 01/04/2023] Open
Abstract
The unique fast spiking (FS) phenotype of cortical parvalbumin-positive (PV) neurons depends on the expression of multiple subtypes of voltage-gated potassium channels (Kv). PV neurons selectively express Kcns3, the gene encoding Kv9.3 subunits, suggesting that Kcns3 expression is critical for the FS phenotype. KCNS3 expression is lower in PV neurons in the neocortex of subjects with schizophrenia, but the effects of this alteration are unclear, because Kv9.3 subunit function is poorly understood. Therefore, to assess the role of Kv9.3 subunits in PV neuron function, we combined gene expression analyses, computational modeling, and electrophysiology in acute slices from the cortex of Kcns3-deficient mice. Kcns3 mRNA levels were ~ 50% lower in cortical PV neurons from Kcns3-deficient relative to wildtype mice. While silent per se, Kv9.3 subunits are believed to amplify the Kv2.1 current in Kv2.1-Kv9.3 channel complexes. Hence, to assess the consequences of reducing Kv9.3 levels, we simulated the effects of decreasing the Kv2.1-mediated current in a computational model. The FS cell model with reduced Kv2.1 produced spike trains with irregular inter-spike intervals, or stuttering, and greater Na+ channel inactivation. As in the computational model, PV basket cells (PVBCs) from Kcns3-deficient mice displayed spike trains with strong stuttering, which depressed PVBC firing. Moreover, Kcns3 deficiency impaired the recruitment of PVBC firing at gamma frequency by stimuli mimicking synaptic input observed during cortical UP states. Our data indicate that Kv9.3 subunits are critical for PVBC physiology and suggest that KCNS3 deficiency in schizophrenia could impair PV neuron firing, possibly contributing to deficits in cortical gamma oscillations in the illness.
Collapse
Affiliation(s)
- Takeaki Miyamae
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Takanori Hashimoto
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan.
| | - Monica Abraham
- Department of Mathematics, Faculty of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Sho Koshikizawa
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Yufan Bian
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Yosuke Nishihata
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - G Bard Ermentrout
- Department of Mathematics, Faculty of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Guillermo Gonzalez-Burgos
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
36
|
Puentes-Mestril C, Delorme J, Wang L, Donnelly M, Popke D, Jiang S, Aton SJ. Sleep Loss Drives Brain Region-Specific and Cell Type-Specific Alterations in Ribosome-Associated Transcripts Involved in Synaptic Plasticity and Cellular Timekeeping. J Neurosci 2021; 41:5386-5398. [PMID: 34001629 PMCID: PMC8221591 DOI: 10.1523/jneurosci.1883-20.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/07/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022] Open
Abstract
Sleep and sleep loss are thought to impact synaptic plasticity, and recent studies have shown that sleep and sleep deprivation (SD) differentially affect gene transcription and protein translation in the mammalian forebrain. However, much less is known regarding how sleep and SD affect these processes in different microcircuit elements within the hippocampus and neocortex, for example, in inhibitory versus excitatory neurons. Here, we use translating ribosome affinity purification (TRAP) and in situ hybridization to characterize the effects of sleep versus SD on abundance of ribosome-associated transcripts in Camk2a-expressing (Camk2a+) pyramidal neurons and parvalbumin-expressing (PV+) interneurons in the hippocampus and neocortex of male mice. We find that while both Camk2a+ neurons and PV+ interneurons in neocortex show concurrent SD-driven increases in ribosome-associated transcripts for activity-regulated effectors of plasticity and transcriptional regulation, these transcripts are minimally affected by SD in hippocampus. Similarly, we find that while SD alters several ribosome-associated transcripts involved in cellular timekeeping in neocortical Camk2a+ and PV+ neurons, effects on circadian clock transcripts in hippocampus are minimal, and restricted to Camk2a+ neurons. Taken together, our results indicate that SD effects on transcripts associated with translating ribosomes are both cell type-specific and brain region-specific, and that these effects are substantially more pronounced in the neocortex than the hippocampus. We conclude that SD-driven alterations in the strength of synapses, excitatory-inhibitory (E-I) balance, and cellular timekeeping are likely more heterogeneous than previously appreciated.SIGNIFICANCE STATEMENT Sleep loss-driven changes in transcript and protein abundance have been used as a means to better understand the function of sleep for the brain. Here, we use translating ribosome affinity purification (TRAP) to characterize changes in abundance of ribosome-associated transcripts in excitatory and inhibitory neurons in mouse hippocampus and neocortex after a brief period of sleep or sleep loss. We show that these changes are not uniform, but are generally more pronounced in excitatory neurons than inhibitory neurons, and more pronounced in neocortex than in hippocampus.
Collapse
Affiliation(s)
- Carlos Puentes-Mestril
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - James Delorme
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - Lijing Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - Marcus Donnelly
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - Donald Popke
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48019
| |
Collapse
|
37
|
Kasai H, Ziv NE, Okazaki H, Yagishita S, Toyoizumi T. Spine dynamics in the brain, mental disorders and artificial neural networks. Nat Rev Neurosci 2021; 22:407-422. [PMID: 34050339 DOI: 10.1038/s41583-021-00467-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
In the brain, most synapses are formed on minute protrusions known as dendritic spines. Unlike their artificial intelligence counterparts, spines are not merely tuneable memory elements: they also embody algorithms that implement the brain's ability to learn from experience and cope with new challenges. Importantly, they exhibit structural dynamics that depend on activity, excitatory input and inhibitory input (synaptic plasticity or 'extrinsic' dynamics) and dynamics independent of activity ('intrinsic' dynamics), both of which are subject to neuromodulatory influences and reinforcers such as dopamine. Here we succinctly review extrinsic and intrinsic dynamics, compare these with parallels in machine learning where they exist, describe the importance of intrinsic dynamics for memory management and adaptation, and speculate on how disruption of extrinsic and intrinsic dynamics may give rise to mental disorders. Throughout, we also highlight algorithmic features of spine dynamics that may be relevant to future artificial intelligence developments.
Collapse
Affiliation(s)
- Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Noam E Ziv
- Technion Faculty of Medicine and Network Biology Research Labs, Technion City, Haifa, Israel
| | - Hitoshi Okazaki
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taro Toyoizumi
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan.,Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Diurnal changes in perineuronal nets and parvalbumin neurons in the rat medial prefrontal cortex. Brain Struct Funct 2021; 226:1135-1153. [PMID: 33585984 DOI: 10.1007/s00429-021-02229-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Perineuronal nets (PNNs) surrounding fast-spiking, parvalbumin (PV) interneurons provide excitatory:inhibitory balance, which is impaired in several disorders associated with altered diurnal rhythms, yet few studies have examined diurnal rhythms of PNNs or PV cells. We measured the intensity and number of PV cells and PNNs labeled with Wisteria floribunda agglutinin (WFA) and also the oxidative stress marker 8-oxo-deoxyguanosine (8-oxo-dG) in rat prelimbic medial prefrontal cortex (mPFC) at Zeitgeber times (ZT) ZT0 (lights-on, inactive phase), ZT6 (mid-inactive phase), ZT12 (lights-off, active phase), and ZT18 (mid-active phase). Relative to ZT0, the intensities of PNN and PV labeling were increased in the dark (active) phase compared with the light (inactive) phase. The intensity of 8-oxo-dG was decreased from ZT0 at all times (ZT6,12,18). We also measured GAD 65/67 and vGLUT1 puncta apposed to PV cells with and without PNNs. There were more excitatory puncta on PV cells with PNNs at ZT18 vs. ZT6, but no changes in PV cells without PNNs and no changes in inhibitory puncta. Whole-cell slice recordings in fast-spiking (PV) cells with PNNs showed an increased ratio of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor:N-methyl-D-aspartate receptor (AMPA: NMDA) at ZT18 vs. ZT6. The number of PV cells and PV/PNN cells containing orthodenticle homeobox 2 (OTX2), which maintains PNNs, showed a strong trend toward an increase from ZT6 to ZT18. Diurnal fluctuations in PNNs and PV cells are expected to alter cortical excitatory:inhibitory balance and provide new insights into treatments for diseases impacted by disturbances in sleep and circadian rhythms.
Collapse
|
39
|
He X, Li J, Zhou G, Yang J, McKenzie S, Li Y, Li W, Yu J, Wang Y, Qu J, Wu Z, Hu H, Duan S, Ma H. Gating of hippocampal rhythms and memory by synaptic plasticity in inhibitory interneurons. Neuron 2021; 109:1013-1028.e9. [PMID: 33548174 DOI: 10.1016/j.neuron.2021.01.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 12/17/2020] [Accepted: 01/14/2021] [Indexed: 12/31/2022]
Abstract
Mental experiences can become long-term memories if the hippocampal activity patterns that encode them are broadcast during network oscillations. The activity of inhibitory neurons is essential for generating these neural oscillations, but molecular control of this dynamic process during learning remains unknown. Here, we show that hippocampal oscillatory strength positively correlates with excitatory monosynaptic drive onto inhibitory neurons (E→I) in freely behaving mice. To establish a causal relationship between them, we identified γCaMKII as the long-sought mediator of long-term potentiation for E→I synapses (LTPE→I), which enabled the genetic manipulation of experience-dependent E→I synaptic input/plasticity. Deleting γCaMKII in parvalbumin interneurons selectively eliminated LTPE→I and disrupted experience-driven strengthening in theta and gamma rhythmicity. Behaviorally, this manipulation impaired long-term memory, for which the kinase activity of γCaMKII was required. Taken together, our data suggest that E→I synaptic plasticity, exemplified by LTPE→I, plays a gatekeeping role in tuning experience-dependent brain rhythms and mnemonic function.
Collapse
Affiliation(s)
- Xingzhi He
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Guangjun Zhou
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jing Yang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Sam McKenzie
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yanjun Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wenwen Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jun Yu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yang Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jing Qu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhiying Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hailan Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Huan Ma
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Mental Health Center, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
40
|
Filice F, Janickova L, Henzi T, Bilella A, Schwaller B. The Parvalbumin Hypothesis of Autism Spectrum Disorder. Front Cell Neurosci 2020; 14:577525. [PMID: 33390904 PMCID: PMC7775315 DOI: 10.3389/fncel.2020.577525] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of autism spectrum disorder (ASD)-a type of neurodevelopmental disorder-is increasing and is around 2% in North America, Asia, and Europe. Besides the known genetic link, environmental, epigenetic, and metabolic factors have been implicated in ASD etiology. Although highly heterogeneous at the behavioral level, ASD comprises a set of core symptoms including impaired communication and social interaction skills as well as stereotyped and repetitive behaviors. This has led to the suggestion that a large part of the ASD phenotype is caused by changes in a few and common set of signaling pathways, the identification of which is a fundamental aim of autism research. Using advanced bioinformatics tools and the abundantly available genetic data, it is possible to classify the large number of ASD-associated genes according to cellular function and pathways. Cellular processes known to be impaired in ASD include gene regulation, synaptic transmission affecting the excitation/inhibition balance, neuronal Ca2+ signaling, development of short-/long-range connectivity (circuits and networks), and mitochondrial function. Such alterations often occur during early postnatal neurodevelopment. Among the neurons most affected in ASD as well as in schizophrenia are those expressing the Ca2+-binding protein parvalbumin (PV). These mainly inhibitory interneurons present in many different brain regions in humans and rodents are characterized by rapid, non-adaptive firing and have a high energy requirement. PV expression is often reduced at both messenger RNA (mRNA) and protein levels in human ASD brain samples and mouse ASD (and schizophrenia) models. Although the human PVALB gene is not a high-ranking susceptibility/risk gene for either disorder and is currently only listed in the SFARI Gene Archive, we propose and present supporting evidence for the Parvalbumin Hypothesis, which posits that decreased PV level is causally related to the etiology of ASD (and possibly schizophrenia).
Collapse
Affiliation(s)
| | | | | | | | - Beat Schwaller
- Section of Medicine, Anatomy, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
41
|
Spiegel I. Experience-regulated molecular mechanisms in cortical GABAergic interneurons: from cellular functions to control over circuit plasticity. Curr Opin Neurobiol 2020; 67:145-154. [PMID: 33316573 DOI: 10.1016/j.conb.2020.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 01/30/2023]
Abstract
Experience-induced changes in GABAergic interneurons (INs) are thought to control the plasticity of neural circuits in the developing and adult cortex. However, it remains poorly understood how experience and the ensuing neuronal activity alter the properties and connectivity of specific IN subtypes and how these cellular changes, in turn, control the plasticity of cortical circuits. Here, I discuss recent experimental and theoretical studies that point to specific experience-induced changes in select IN subtypes as central regulators of plasticity in the cortex. In particular, I focus on the recent identification of several experience-regulated secreted molecules that modulate specific sets of synapses in IN subtypes. I argue that elucidating these molecular mechanisms will allow us to test experimentally the predictions made by theoretical models about the plasticity functions of specific IN subtypes.
Collapse
Affiliation(s)
- Ivo Spiegel
- Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| |
Collapse
|
42
|
Janickova L, Schwaller B. Parvalbumin-Deficiency Accelerates the Age-Dependent ROS Production in Pvalb Neurons in vivo: Link to Neurodevelopmental Disorders. Front Cell Neurosci 2020; 14:571216. [PMID: 33132847 PMCID: PMC7549402 DOI: 10.3389/fncel.2020.571216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
In neurodevelopmental disorders (NDDs) including autism spectrum disorder (ASD) and schizophrenia, impairment/malfunctioning of a subpopulation of interneurons expressing the calcium-binding protein parvalbumin (PV) -here termed Pvalb neurons- has gradually emerged as a possible cause. These neurons may represent a hub or point-of-convergence in the etiology of NDD. Increased oxidative stress associated with mitochondria impairment in Pvalb neurons is discussed as an essential step in schizophrenia etiology. Since PV downregulation is a common finding in ASD and schizophrenia individuals and PV-deficient (PV-/-) mice show a strong ASD-like behavior phenotype, we investigated the putative link between PV expression, alterations in mitochondria and oxidative stress. In a longitudinal study with 1, 3, and 6-months old PV-/- and wild type mice, oxidative stress was investigated in 9 Pvalb neuron subpopulations in the hippocampus, striatum, somatosensory cortex, medial prefrontal cortex, thalamic reticular nucleus (TRN) and cerebellum. In Pvalb neuron somata in the striatum and TRN, we additionally determined mitochondria volume and distribution at these three time points. In all Pvalb neuron subpopulations, we observed an age-dependent increase in oxidative stress and the increase strongly correlated with PV expression levels, but not with mitochondria density in these Pvalb neurons. Moreover, oxidative stress was elevated in Pvalb neurons of PV-/- mice and the magnitude of the effect was again correlated with PV expression levels in the corresponding wild type Pvalb neuron subpopulations. The PV-dependent effect was insignificant at 1 month and relative differences between WT and PV-/- Pvalb neurons were largest at 3 months. Besides the increase in mitochondria volume in PV's absence in TRN and striatal PV-/- Pvalb neurons fully present already at 1 month, we observed a redistribution of mitochondria from the perinuclear region toward the plasma membrane at all time points. We suggest that in absence of PV, slow Ca2+ buffering normally exerted by PV is compensated by a (mal)adaptive, mostly sub-plasmalemmal increase in mitochondria resulting in increased oxidative stress observed in 3- and 6-months old mice. Since PV-/- mice display core ASD-like symptoms already at 1 month, oxidative stress in Pvalb neurons is not a likely cause for their ASD-related behavior observed at this age.
Collapse
Affiliation(s)
| | - Beat Schwaller
- Department of Neurosciences amd Movement Science, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
43
|
Tsubomoto M, Kawabata R, Zhu X, Minabe Y, Chen K, Lewis DA, Hashimoto T. Expression of Transcripts Selective for GABA Neuron Subpopulations across the Cortical Visuospatial Working Memory Network in the Healthy State and Schizophrenia. Cereb Cortex 2020; 29:3540-3550. [PMID: 30247542 DOI: 10.1093/cercor/bhy227] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/01/2018] [Accepted: 08/22/2018] [Indexed: 01/21/2023] Open
Abstract
Visuospatial working memory (WM), which is impaired in schizophrenia, depends on a distributed network including visual, posterior parietal, and dorsolateral prefrontal cortical regions. Within each region, information processing is differentially regulated by subsets of γ-aminobutyric acid (GABA) neurons that express parvalbumin (PV), somatostatin (SST), or vasoactive intestinal peptide (VIP). In schizophrenia, WM impairments have been associated with alterations of PV and SST neurons in the dorsolateral prefrontal cortex. Here, we quantified transcripts selectively expressed in GABA neuron subsets across four cortical regions in the WM network from comparison and schizophrenia subjects. In comparison subjects, PV mRNA levels declined and SST mRNA levels increased from posterior to anterior regions, whereas VIP mRNA levels were comparable across regions except for the primary visual cortex (V1). In schizophrenia subjects, each transcript in PV and SST neurons exhibited similar alterations across all regions, whereas transcripts in VIP neurons were unaltered in any region except for V1. These findings suggest that the contribution of each GABA neuron subset to inhibitory regulation of local circuitry normally differs across cortical regions of the visuospatial WM network and that in schizophrenia alterations of PV and SST neurons are a shared feature across these regions, whereas VIP neurons are affected only in V1.
Collapse
Affiliation(s)
- Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Xiaonan Zhu
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yoshio Minabe
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Kehui Chen
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Janickova L, Rechberger KF, Wey L, Schwaller B. Absence of parvalbumin increases mitochondria volume and branching of dendrites in inhibitory Pvalb neurons in vivo: a point of convergence of autism spectrum disorder (ASD) risk gene phenotypes. Mol Autism 2020; 11:47. [PMID: 32517751 PMCID: PMC7285523 DOI: 10.1186/s13229-020-00323-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/27/2020] [Indexed: 01/01/2023] Open
Abstract
Background In fast firing, parvalbumin (PV)-expressing (Pvalb) interneurons, PV acts as an intracellular Ca2+ signal modulator with slow-onset kinetics. In Purkinje cells of PV−/− mice, adaptive/homeostatic mechanisms lead to an increase in mitochondria, organelles equally capable of delayed Ca2+ sequestering/buffering. An inverse regulation of PV and mitochondria likewise operates in cell model systems in vitro including myotubes, epithelial cells, and oligodendrocyte-like cells overexpressing PV. Whether such opposite regulation pertains to all Pvalb neurons is currently unknown. In oligodendrocyte-like cells, PV additionally decreases growth and branching of processes in a cell-autonomous manner. Methods The in vivo effects of absence of PV were investigated in inhibitory Pvalb neurons expressing EGFP, present in the somatosensory and medial prefrontal cortex, striatum, thalamic reticular nucleus, hippocampal regions DG, CA3, and CA1 and cerebellum of mice either wild-type or knockout (PV−/−) for the Pvalb gene. Changes in Pvalb neuron morphology and PV concentrations were determined using immunofluorescence, followed by 3D-reconstruction and quantitative image analyses. Results PV deficiency led to an increase in mitochondria volume and density in the soma; the magnitude of the effect was positively correlated with the estimated PV concentrations in the various Pvalb neuron subpopulations in wild-type neurons. The increase in dendrite length and branching, as well as thickness of proximal dendrites of selected PV−/− Pvalb neurons is likely the result of the observed increased density and length of mitochondria in these PV−/− Pvalb neuron dendrites. The increased branching and soma size directly linked to the absence of PV is assumed to contribute to the increased volume of the neocortex present in juvenile PV−/− mice. The extended dendritic branching is in line with the hypothesis of local hyperconnectivity in autism spectrum disorder (ASD) and ASD mouse models including PV−/− mice, which display all ASD core symptoms and several comorbidities including cortical macrocephaly at juvenile age. Conclusion PV is involved in most proposed mechanisms implicated in ASD etiology: alterations in Ca2+ signaling affecting E/I balance, changes in mitochondria structure/function, and increased dendritic length and branching, possibly resulting in local hyperconnectivity, all in a likely cell autonomous way.
Collapse
Affiliation(s)
- Lucia Janickova
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Karin Farah Rechberger
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Lucas Wey
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland
| | - Beat Schwaller
- Anatomy, Section of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland.
| |
Collapse
|
45
|
Li B, Suutari BS, Sun SD, Luo Z, Wei C, Chenouard N, Mandelberg NJ, Zhang G, Wamsley B, Tian G, Sanchez S, You S, Huang L, Neubert TA, Fishell G, Tsien RW. Neuronal Inactivity Co-opts LTP Machinery to Drive Potassium Channel Splicing and Homeostatic Spike Widening. Cell 2020; 181:1547-1565.e15. [PMID: 32492405 DOI: 10.1016/j.cell.2020.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 01/28/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.
Collapse
Affiliation(s)
- Boxing Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China; Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA.
| | - Benjamin S Suutari
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Simón(e) D. Sun
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Zhengyi Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China
| | - Chuanchuan Wei
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Nicolas Chenouard
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Nataniel J Mandelberg
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Guoan Zhang
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Brie Wamsley
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Guoling Tian
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sandrine Sanchez
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sikun You
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Lianyan Huang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Gordon Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
46
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
47
|
Synaptic GluN2A-Containing NMDA Receptors: From Physiology to Pathological Synaptic Plasticity. Int J Mol Sci 2020; 21:ijms21041538. [PMID: 32102377 PMCID: PMC7073220 DOI: 10.3390/ijms21041538] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-d-Aspartate Receptors (NMDARs) are ionotropic glutamate-gated receptors. NMDARs are tetramers composed by several homologous subunits of GluN1-, GluN2-, or GluN3-type, leading to the existence in the central nervous system of a high variety of receptor subtypes with different pharmacological and signaling properties. NMDAR subunit composition is strictly regulated during development and by activity-dependent synaptic plasticity. Given the differences between GluN2 regulatory subunits of NMDAR in several functions, here we will focus on the synaptic pool of NMDARs containing the GluN2A subunit, addressing its role in both physiology and pathological synaptic plasticity as well as the contribution in these events of different types of GluN2A-interacting proteins.
Collapse
|
48
|
In Vivo Imaging of the Coupling between Neuronal and CREB Activity in the Mouse Brain. Neuron 2019; 105:799-812.e5. [PMID: 31883788 DOI: 10.1016/j.neuron.2019.11.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
Sensory experiences cause long-term modifications of neuronal circuits by modulating activity-dependent transcription programs that are vital for regulation of long-term synaptic plasticity and memory. However, it has not been possible to precisely determine the interaction between neuronal activity patterns and transcription factor activity. Here we present a technique using two-photon fluorescence lifetime imaging (2pFLIM) with new FRET biosensors to chronically image in vivo signaling of CREB, an activity-dependent transcription factor important for synaptic plasticity, at single-cell resolution. Simultaneous imaging of the red-shifted CREB sensor and GCaMP permitted exploration of how experience shapes the interplay between CREB and neuronal activity in the neocortex of awake mice. Dark rearing increased the sensitivity of CREB activity to Ca2+ elevations and prolonged the duration of CREB activation to more than 24 h in the visual cortex. This technique will allow researchers to unravel the transcriptional dynamics underlying experience-dependent plasticity in the brain.
Collapse
|
49
|
Malik R, Pai ELL, Rubin AN, Stafford AM, Angara K, Minasi P, Rubenstein JL, Sohal VS, Vogt D. Tsc1 represses parvalbumin expression and fast-spiking properties in somatostatin lineage cortical interneurons. Nat Commun 2019; 10:4994. [PMID: 31676823 PMCID: PMC6825152 DOI: 10.1038/s41467-019-12962-4] [Citation(s) in RCA: 32] [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: 04/13/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022] Open
Abstract
Medial ganglionic eminence (MGE)-derived somatostatin (SST)+ and parvalbumin (PV)+ cortical interneurons (CINs), have characteristic molecular, anatomical and physiological properties. However, mechanisms regulating their diversity remain poorly understood. Here, we show that conditional loss of the Tuberous Sclerosis Complex (TSC) gene, Tsc1, which inhibits the mammalian target of rapamycin (MTOR), causes a subset of SST+ CINs, to express PV and adopt fast-spiking (FS) properties, characteristic of PV+ CINs. Milder intermediate phenotypes also occur when only one allele of Tsc1 is deleted. Notably, treatment of adult mice with rapamycin, which inhibits MTOR, reverses the phenotypes. These data reveal novel functions of MTOR signaling in regulating PV expression and FS properties, which may contribute to TSC neuropsychiatric symptoms. Moreover, they suggest that CINs can exhibit properties intermediate between those classically associated with PV+ or SST+ CINs, which may be dynamically regulated by the MTOR signaling.
Collapse
Affiliation(s)
- Ruchi Malik
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Emily Ling-Lin Pai
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Neuroscience Program, UCSF, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Anna N Rubin
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - April M Stafford
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA
| | - Kartik Angara
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA
| | - Petros Minasi
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
| | - John L Rubenstein
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Neuroscience Program, UCSF, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Vikaas S Sohal
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA.
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA.
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA.
| | - Daniel Vogt
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
50
|
Reinhard SM, Rais M, Afroz S, Hanania Y, Pendi K, Espinoza K, Rosenthal R, Binder DK, Ethell IM, Razak KA. Reduced perineuronal net expression in Fmr1 KO mice auditory cortex and amygdala is linked to impaired fear-associated memory. Neurobiol Learn Mem 2019; 164:107042. [PMID: 31326533 PMCID: PMC7519848 DOI: 10.1016/j.nlm.2019.107042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/20/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Fragile X Syndrome (FXS) is a leading cause of heritable intellectual disability and autism. Humans with FXS show anxiety, sensory hypersensitivity and impaired learning. The mechanisms of learning impairments can be studied in the mouse model of FXS, the Fmr1 KO mouse, using tone-associated fear memory paradigms. Our previous study reported impaired development of parvalbumin (PV) positive interneurons and perineuronal nets (PNN) in the auditory cortex of Fmr1 KO mice. A recent study suggested PNN dynamics in the auditory cortex following tone-shock association is necessary for fear expression. Together these data suggest that abnormal PNN regulation may underlie tone-fear association learning deficits in Fmr1 KO mice. We tested this hypothesis by quantifying PV and PNN expression in the amygdala, hippocampus and auditory cortex of Fmr1 KO mice following fear conditioning. We found impaired tone-associated memory formation in Fmr1 KO mice. This was paralleled by impaired learning-associated regulation of PNNs in the superficial layers of auditory cortex in Fmr1 KO mice. PV cell density decreased in the auditory cortex in response to fear conditioning in both WT and Fmr1 KO mice. Learning-induced increase of PV expression in the CA3 hippocampus was only observed in WT mice. We also found reduced PNN density in the amygdala and auditory cortex of Fmr1 KO mice in all conditions, as well as reduced PNN intensity in CA2 hippocampus. There was a positive correlation between tone-associated memory and PNN density in the amygdala and auditory cortex, consistent with a tone-association deficit. Altogether our studies suggest a link between impaired PV and PNN regulation within specific regions of the fear conditioning circuit and impaired tone memory formation in Fmr1 KO mice.
Collapse
Affiliation(s)
- Sarah M Reinhard
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Sonia Afroz
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Yasmien Hanania
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Kasim Pendi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Katherine Espinoza
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Robert Rosenthal
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| | - Khaleel A Razak
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| |
Collapse
|