1
|
O'Shea MJ, Anversa RG, Ch'ng SS, Campbell EJ, Walker LC, Andrews ZB, Lawrence AJ, Brown RM. An Island of Reil excitation: Mapping glutamatergic (vGlut1+ and vGlut2+) connections in the medial insular cortex. Biochem Pharmacol 2024; 230:116637. [PMID: 39561925 DOI: 10.1016/j.bcp.2024.116637] [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: 05/06/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The insular cortex is a multifunctional and richly connected region of the cerebral cortex, critical in the neural integration of external stimuli and internal signals. Well-served for this role by a large network of afferent and efferent connections, the mouse insula can be simplified into an anterior, medial and posterior portion. Here we focus on the medial subregion, a once over-looked area that has gained recent attention for its involvement in an array of behaviours. Although the connections of medial insular cortex neurons have been previously identified, their precise glutamatergic phenotype remains undefined (typically defined by the presence of the subtype of vesicular glutamate transporters). Hence, we combined Cre knock-in mouse lines and adeno-associated viral tracing to distinguish between the expression of the two major vesicular glutamate transporters, type 1 (vGlut1) and 2 (vGlut2), in the subregion's neuronal inputs and outputs. Our results determined that the medial insula has extensive glutamatergic efferents expressing both vGlut1 and vGlut2 throughout the neuraxis. In contrast, a more conservative number of glutamatergic inputs were observed, with exclusively vGlut2+ projections received from hypothalamic and thalamic regions. Taken together, we demonstrate that vGlut1- and vGlut2-expressing networks of this insular subdivision have distinct connectivity patterns, including a greater abundance of vGlut1+ fibres innervating hypothalamic regions and the extended amygdala. These findings provide insight into the distinct chemo-architecture of this region, which may facilitate further investigation into the role of the medial insula in complex behaviour.
Collapse
Affiliation(s)
- Mia Jessica O'Shea
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Roberta Goncalves Anversa
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Sarah Sulaiman Ch'ng
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Erin Jane Campbell
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Leigh Clasina Walker
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Zane Bruce Andrews
- Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Andrew John Lawrence
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Robyn Mary Brown
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
2
|
Ceballos CC, Ma L, Qin M, Zhong H. Widespread co-release of glutamate and GABA throughout the mouse brain. Commun Biol 2024; 7:1502. [PMID: 39537846 PMCID: PMC11560972 DOI: 10.1038/s42003-024-07198-y] [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: 06/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Several brain neuronal populations transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA. However, it remains largely unknown whether these opposing neurotransmitters are co-released simultaneously or are independently transmitted at different times and locations. By recording from acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons. This observation cannot be explained by accidental coincidence of monophasic excitatory and inhibitory minis. Interestingly, these biphasic minis could either be an excitatory current leading an inhibitory current or vice versa. Deletion of dopaminergic neurons did not eliminate biphasic minis, indicating that they originate from another source. Importantly, we found that both types of biphasic minis were present in multiple striatal neuronal types and in nine out of ten other brain regions. Overall, co-release of glutamate and GABA appears to be a widespread mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
3
|
Maity S, Huang Y, Kilgore MD, Thurmon AN, Vaasjo LO, Galazo MJ, Xu X, Cao J, Wang X, Ning B, Liu N, Fan J. Mapping dynamic molecular changes in hippocampal subregions after traumatic brain injury through spatial proteomics. Clin Proteomics 2024; 21:32. [PMID: 38735925 PMCID: PMC11089002 DOI: 10.1186/s12014-024-09485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) often results in diverse molecular responses, challenging traditional proteomic studies that measure average changes at tissue levels and fail to capture the complexity and heterogeneity of the affected tissues. Spatial proteomics offers a solution by providing insights into sub-region-specific alterations within tissues. This study focuses on the hippocampal sub-regions, analyzing proteomic expression profiles in mice at the acute (1 day) and subacute (7 days) phases of post-TBI to understand subregion-specific vulnerabilities and long-term consequences. METHODS Three mice brains were collected from each group, including Sham, 1-day post-TBI and 7-day post-TBI. Hippocampal subregions were extracted using Laser Microdissection (LMD) and subsequently analyzed by label-free quantitative proteomics. RESULTS The spatial analysis reveals region-specific protein abundance changes, highlighting the elevation of FN1, LGALS3BP, HP, and MUG-1 in the stratum moleculare (SM), suggesting potential immune cell enrichment post-TBI. Notably, established markers of chronic traumatic encephalopathy, IGHM and B2M, exhibit specific upregulation in the dentate gyrus bottom (DG2) independent of direct mechanical injury. Metabolic pathway analysis identifies disturbances in glucose and lipid metabolism, coupled with activated cholesterol synthesis pathways enriched in SM at 7-Day post-TBI and subsequently in deeper DG1 and DG2 suggesting a role in neurogenesis and the onset of recovery. Coordinated activation of neuroglia and microtubule dynamics in DG2 suggest recovery mechanisms in less affected regions. Cluster analysis revealed spatial variations post-TBI, indicative of dysregulated neuronal plasticity and neurogenesis and further predisposition to neurological disorders. TBI-induced protein upregulation (MUG-1, PZP, GFAP, TJP, STAT-1, and CD44) across hippocampal sub-regions indicates shared molecular responses and links to neurological disorders. Spatial variations were demonstrated by proteins dysregulated in both or either of the time-points exclusively in each subregion (ELAVL2, CLIC1 in PL, CD44 and MUG-1 in SM, and SHOC2, LGALS3 in DG). CONCLUSIONS Utilizing advanced spatial proteomics techniques, the study unveils the dynamic molecular responses in distinct hippocampal subregions post-TBI. It uncovers region-specific vulnerabilities and dysregulated neuronal processes, and potential recovery-related pathways that contribute to our understanding of TBI's neurological consequences and provides valuable insights for biomarker discovery and therapeutic targets.
Collapse
Affiliation(s)
- Sudipa Maity
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuanyu Huang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mitchell D Kilgore
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Abbigail N Thurmon
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | | | - Maria J Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jing Cao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane University Translational Sciences Institute, New Orleans, LA, USA.
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
4
|
Ceballos CC, Ma L, Qin M, Zhong H. Prevalent co-release of glutamate and GABA throughout the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587069. [PMID: 38585864 PMCID: PMC10996720 DOI: 10.1101/2024.03.27.587069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Several neuronal populations in the brain transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA, to downstream neurons. However, it remains largely unknown whether these opposing neurotransmitters are co-released onto the same postsynaptic neuron simultaneously or are independently transmitted at different time and locations (called co-transmission). Here, using whole-cell patch-clamp recording on acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons (SPNs). This observation cannot be explained by accidental coincidence of monophasic miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs, respectively), arguing for the co-release of glutamate and GABA. Interestingly, these biphasic minis could either be an mEPSC leading an mIPSC or vice versa. Although dopaminergic axons release both glutamate and GABA in the striatum, deletion of dopamine neurons did not eliminate biphasic minis, indicating that the co-release originates from another neuronal type. Importantly, we found that both types of biphasic minis were detected in other neuronal subtypes in the striatum as well as in nine out of ten additionally tested brain regions. Our results suggest that co-release of glutamate and GABA is a prevalent mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
5
|
Zhou J, Zhang Z, Wu M, Liu H, Pang Y, Bartlett A, Peng Z, Ding W, Rivkin A, Lagos WN, Williams E, Lee CT, Miyazaki PA, Aldridge A, Zeng Q, Salinda JLA, Claffey N, Liem M, Fitzpatrick C, Boggeman L, Yao Z, Smith KA, Tasic B, Altshul J, Kenworthy MA, Valadon C, Nery JR, Castanon RG, Patne NS, Vu M, Rashid M, Jacobs M, Ito T, Osteen J, Emerson N, Lee J, Cho S, Rink J, Huang HH, Pinto-Duartec A, Dominguez B, Smith JB, O'Connor C, Zeng H, Chen S, Lee KF, Mukamel EA, Jin X, Margarita Behrens M, Ecker JR, Callaway EM. Brain-wide correspondence of neuronal epigenomics and distant projections. Nature 2023; 624:355-365. [PMID: 38092919 PMCID: PMC10719087 DOI: 10.1038/s41586-023-06823-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023]
Abstract
Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.
Collapse
Affiliation(s)
- Jingtian Zhou
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
| | - Zhuzhu Zhang
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - May Wu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Yan Pang
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zihao Peng
- School of Mathematics and Computer Science, Nanchang University, Nanchang, China
- Henan Engineering Research Center of Intelligent Technology and Application, Henan University, Kaifeng, China
| | - Wubin Ding
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Angeline Rivkin
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Will N Lagos
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Elora Williams
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cheng-Ta Lee
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Paula Assakura Miyazaki
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Andrew Aldridge
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qiurui Zeng
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - J L Angelo Salinda
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Naomi Claffey
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michelle Liem
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Conor Fitzpatrick
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lara Boggeman
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Jordan Altshul
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mia A Kenworthy
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cynthia Valadon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rosa G Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Neelakshi S Patne
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Minh Vu
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mohammad Rashid
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Matthew Jacobs
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tony Ito
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Julia Osteen
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nora Emerson
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jasper Lee
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Silvia Cho
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jon Rink
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hsiang-Hsuan Huang
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - António Pinto-Duartec
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bertha Dominguez
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jared B Smith
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Carolyn O'Connor
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Shengbo Chen
- Henan Engineering Research Center of Intelligent Technology and Application, Henan University, Kaifeng, China
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Kuo-Fen Lee
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA
| | - Xin Jin
- Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science, New York University Shanghai, Shanghai, China
| | - M Margarita Behrens
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Edward M Callaway
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
6
|
Olivero G, Grilli M, Marchi M, Pittaluga A. Metamodulation of presynaptic NMDA receptors: New perspectives for pharmacological interventions. Neuropharmacology 2023; 234:109570. [PMID: 37146939 DOI: 10.1016/j.neuropharm.2023.109570] [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: 02/15/2023] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Metamodulation shifted the scenario of the central neuromodulation from a simplified unimodal model to a multimodal one. It involves different receptors/membrane proteins physically associated or merely colocalized that act in concert to control the neuronal functions influencing each other. Defects or maladaptation of metamodulation would subserve neuropsychiatric disorders or even synaptic adaptations relevant to drug dependence. Therefore, this "vulnerability" represents a main issue to be deeply analyzed to predict its aetiopathogenesis, but also to propose targeted pharmaceutical interventions. The review focusses on presynaptic release-regulating NMDA receptors and on some of the mechanisms of their metamodulation described in the literature. Attention is paid to the interactors, including both ionotropic and metabotropic receptors, transporters and intracellular proteins, which metamodulate their responsiveness in physiological conditions but also undergo adaptation that are relevant to neurological dysfunctions. All these structures are attracting more and more the interest as promising druggable targets for the treatment of NMDAR-related central diseases: these substances would not exert on-off control of the colocalized NMDA receptors (as usually observed with NMDAR full agonists/antagonists), but rather modulate their functions, with the promise of limiting side effects that would favor their translation from preclinic to clinic.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy; Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 16148, Genoa, Italy.
| | - Mario Marchi
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy; Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 16148, Genoa, Italy
| |
Collapse
|
7
|
Cope EC, Wang SH, Waters RC, Gore IR, Vasquez B, Laham BJ, Gould E. Activation of the CA2-ventral CA1 pathway reverses social discrimination dysfunction in Shank3B knockout mice. Nat Commun 2023; 14:1750. [PMID: 36991001 PMCID: PMC10060401 DOI: 10.1038/s41467-023-37248-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Mutation or deletion of the SHANK3 gene, which encodes a synaptic scaffolding protein, is linked to autism spectrum disorder and Phelan-McDermid syndrome, conditions associated with social memory impairments. Shank3B knockout mice also exhibit social memory deficits. The CA2 region of the hippocampus integrates numerous inputs and sends a major output to the ventral CA1 (vCA1). Despite finding few differences in excitatory afferents to the CA2 in Shank3B knockout mice, we found that activation of CA2 neurons as well as the CA2-vCA1 pathway restored social recognition function to wildtype levels. vCA1 neuronal oscillations have been linked to social memory, but we observed no differences in these measures between wildtype and Shank3B knockout mice. However, activation of the CA2 enhanced vCA1 theta power in Shank3B knockout mice, concurrent with behavioral improvements. These findings suggest that stimulating adult circuitry in a mouse model with neurodevelopmental impairments can invoke latent social memory function.
Collapse
Affiliation(s)
- Elise C Cope
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Samantha H Wang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Renée C Waters
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Betsy Vasquez
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Blake J Laham
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
8
|
Fudge JL, Kelly EA, Hackett TA. Corticotropin Releasing Factor (CRF) Coexpression in GABAergic, Glutamatergic, and GABA/Glutamatergic Subpopulations in the Central Extended Amygdala and Ventral Pallidum of Young Male Primates. J Neurosci 2022; 42:8997-9010. [PMID: 36280261 PMCID: PMC9732834 DOI: 10.1523/jneurosci.1453-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The central extended amygdala (CEA) and ventral pallidum (VP) are involved in diverse motivated behaviors based on rodent models. These structures are conserved, but expanded, in higher primates, including human. Corticotropin releasing factor (CRF), a canonical "stress molecule" associated with the CEA and VP circuitry across species, is dynamically regulated by stress and drugs of abuse and misuse. CRF's effects on circuits critically depend on its colocation with primary "fast" transmitters, making this crucial for understanding circuit effects. We surveyed the distribution and colocalization of CRF-, VGluT2- (vesicular glutamate transporter 2), and VGAT- (vesicular GABA transporter) mRNA in specific subregions of the CEA and VP in young male monkeys. Although CRF-containing neurons were clustered in the lateral central bed nucleus (BSTLcn), the majority were broadly dispersed throughout other CEA subregions, and the VP. CRF/VGAT-only neurons were highest in the BSTLcn, lateral central amygdala nucleus (CeLcn), and medial central amygdala nucleus (CeM) (74%, 73%, and 85%, respectively). In contrast, lower percentages of CRF/VGAT only neurons populated the sublenticular extended amygdala (SLEAc), ventrolateral bed nucleus (BSTLP), and VP (53%, 54%, 17%, respectively), which had higher complements of CRF/VGAT/VGluT2-labeled neurons (33%, 29%, 67%, respectively). Thus, the majority of CRF-neurons at the "poles" (BSTLcn and CeLcn/CeM) of the CEA are inhibitory, while the "extended" BSTLP and SLEAc subregions, and neighboring VP, have a more complex profile with admixtures of "multiplexed" excitatory CRF neurons. CRF's colocalization with its various fast transmitters is likely circuit-specific, and relevant for understanding CRF actions on specific target sites.SIGNIFICANCE STATEMENT The central extended amygdala (CEA) and ventral pallidum (VP) regulate multiple motivated behaviors through differential downstream projections. The stress neuropeptide corticotropin releasing factor (CRF) is enriched in the CEA, and is thought to "set the gain" through modulatory effects on coexpressed primary transmitters. Using protein and transcript assays in monkey, we found that CRF neurons are broadly and diffusely distributed in CEA and VP. CRF mRNA+ neurons colocalize with VGAT (GABA) and VGluT2 (glutamate) mRNAs in different proportions depending on subregion. CRF mRNA was also coexpressed in a subpopulation of VGAT/VGluT2 mRNA ("multiplexed") cells, which were most prominent in the VP and "pallidal"-like parts of the CEA. Heterogeneous CRF and fast transmitter coexpression across CEA/VP subregions implies circuit-specific effects.
Collapse
Affiliation(s)
- Julie L Fudge
- Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642
| | - Emily A Kelly
- Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642
| | - Troy A Hackett
- Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
9
|
Ncube D, Tallafuss A, Serafin J, Bruckner J, Farnsworth DR, Miller AC, Eisen JS, Washbourne P. A conserved transcriptional fingerprint of multi-neurotransmitter neurons necessary for social behavior. BMC Genomics 2022; 23:675. [PMID: 36175871 PMCID: PMC9523972 DOI: 10.1186/s12864-022-08879-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Background An essential determinant of a neuron’s functionality is its neurotransmitter phenotype. We previously identified a defined subpopulation of cholinergic neurons required for social orienting behavior in zebrafish. Results We transcriptionally profiled these neurons and discovered that they are capable of synthesizing both acetylcholine and GABA. We also established a constellation of transcription factors and neurotransmitter markers that can be used as a “transcriptomic fingerprint” to recognize a homologous neuronal population in another vertebrate. Conclusion Our results suggest that this transcriptomic fingerprint and the cholinergic-GABAergic neuronal subtype that it defines are evolutionarily conserved. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08879-w.
Collapse
Affiliation(s)
- Denver Ncube
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Alexandra Tallafuss
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Jen Serafin
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Joseph Bruckner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Dylan R Farnsworth
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Judith S Eisen
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Philip Washbourne
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
10
|
Cristofari P, Desplanque M, Poirel O, Hébert A, Dumas S, Herzog E, Danglot L, Geny D, Gilles JF, Geeverding A, Bolte S, Canette A, Trichet M, Fabre V, Daumas S, Pietrancosta N, El Mestikawy S, Bernard V. Nanoscopic distribution of VAChT and VGLUT3 in striatal cholinergic varicosities suggests colocalization and segregation of the two transporters in synaptic vesicles. Front Mol Neurosci 2022; 15:991732. [PMID: 36176961 PMCID: PMC9513193 DOI: 10.3389/fnmol.2022.991732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/26/2022] [Indexed: 11/26/2022] Open
Abstract
Striatal cholinergic interneurons (CINs) use acetylcholine (ACh) and glutamate (Glut) to regulate the striatal network since they express vesicular transporters for ACh (VAChT) and Glut (VGLUT3). However, whether ACh and Glut are released simultaneously and/or independently from cholinergic varicosities is an open question. The answer to that question requires the multichannel detection of vesicular transporters at the level of single synaptic vesicle (SV). Here, we used super-resolution STimulated Emission Depletion microscopy (STED) to characterize and quantify the distribution of VAChT and VGLUT3 in CINs SVs. Nearest-neighbor distances analysis between VAChT and VGLUT3-immunofluorescent spots revealed that 34% of CINs SVs contain both VAChT and VGLUT3. In addition, 40% of SVs expressed only VAChT while 26% of SVs contain only VGLUT3. These results suggest that SVs from CINs have the potential to store simultaneously or independently ACh and/or Glut. Overall, these morphological findings support the notion that CINs varicosities can signal with either ACh or Glut or both with an unexpected level of complexity.
Collapse
Affiliation(s)
- Paola Cristofari
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Mazarine Desplanque
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Odile Poirel
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Alison Hébert
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | | | - Etienne Herzog
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Lydia Danglot
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging Facility, Paris, France
- GHU Paris Psychiatrie & Neurosciences, Paris, France
| | - David Geny
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging Facility, Paris, France
| | - Jean-François Gilles
- Imaging Facility of the Institut de Biologie Paris-Seine (IBPS)—Sorbonne Université, Paris, France
| | - Audrey Geeverding
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Service de Microscopie Électronique (IBPS-SME), Paris, France
| | - Susanne Bolte
- Imaging Facility of the Institut de Biologie Paris-Seine (IBPS)—Sorbonne Université, Paris, France
| | - Alexis Canette
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Service de Microscopie Électronique (IBPS-SME), Paris, France
| | - Michaël Trichet
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Service de Microscopie Électronique (IBPS-SME), Paris, France
| | - Véronique Fabre
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Stéphanie Daumas
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Nicolas Pietrancosta
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- Sorbonne Université—CNRS UMR 7203—Laboratoire des BioMolécules, Paris, France
| | - Salah El Mestikawy
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Véronique Bernard
- Sorbonne Université—CNRS UMR 8246—INSERM U1130—Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- *Correspondence: Véronique Bernard,
| |
Collapse
|
11
|
Perrenoud Q, Leclerc C, Geoffroy H, Vitalis T, Richetin K, Rampon C, Gallopin T. Molecular and electrophysiological features of GABAergic neurons in the dentate gyrus reveal limited homology with cortical interneurons. PLoS One 2022; 17:e0270981. [PMID: 35802727 PMCID: PMC9269967 DOI: 10.1371/journal.pone.0270981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
GABAergic interneurons tend to diversify into similar classes across telencephalic regions. However, it remains unclear whether the electrophysiological and molecular properties commonly used to define these classes are discriminant in the hilus of the dentate gyrus. Here, using patch-clamp combined with single cell RT-PCR, we compare the relevance of commonly used electrophysiological and molecular features for the clustering of GABAergic interneurons sampled from the mouse hilus and primary sensory cortex. While unsupervised clustering groups cortical interneurons into well-established classes, it fails to provide a convincing partition of hilar interneurons. Statistical analysis based on resampling indicates that hilar and cortical GABAergic interneurons share limited homology. While our results do not invalidate the use of classical molecular marker in the hilus, they indicate that classes of hilar interneurons defined by the expression of molecular markers do not exhibit strongly discriminating electrophysiological properties.
Collapse
Affiliation(s)
- Quentin Perrenoud
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Clémence Leclerc
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Hélène Geoffroy
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Tania Vitalis
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Kevin Richetin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Li H, Zhao J, Lai L, Xia Y, Wan C, Wei S, Liang J, Chen Y, Xu N. Loss of SST and PV Positive Interneurons in the Ventral Hippocampus Results in Anxiety-like Behavior in 5xFAD Mice. Neurobiol Aging 2022; 117:165-178. [DOI: 10.1016/j.neurobiolaging.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/13/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
|
13
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
14
|
Upmanyu N, Jin J, Emde HVD, Ganzella M, Bösche L, Malviya VN, Zhuleku E, Politi AZ, Ninov M, Silbern I, Leutenegger M, Urlaub H, Riedel D, Preobraschenski J, Milosevic I, Hell SW, Jahn R, Sambandan S. Colocalization of different neurotransmitter transporters on synaptic vesicles is sparse except for VGLUT1 and ZnT3. Neuron 2022; 110:1483-1497.e7. [PMID: 35263617 DOI: 10.1016/j.neuron.2022.02.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/08/2022] [Accepted: 02/10/2022] [Indexed: 12/26/2022]
Abstract
Vesicular transporters (VTs) define the type of neurotransmitter that synaptic vesicles (SVs) store and release. While certain mammalian neurons release multiple transmitters, it is not clear whether the release occurs from the same or distinct vesicle pools at the synapse. Using quantitative single-vesicle imaging, we show that a vast majority of SVs in the rodent brain contain only one type of VT, indicating specificity for a single neurotransmitter. Interestingly, SVs containing dual transporters are highly diverse (27 types) but small in proportion (2% of all SVs), excluding the largest pool that carries VGLUT1 and ZnT3 (34%). Using VGLUT1-ZnT3 SVs, we demonstrate that the transporter colocalization influences the SV content and synaptic quantal size. Thus, the presence of diverse transporters on the same vesicle is bona fide, and depending on the VT types, this may act to regulate neurotransmitter type, content, and release in space and time.
Collapse
Affiliation(s)
- Neha Upmanyu
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Jialin Jin
- European Neurosciences Institute, A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Göttingen 37077, Germany
| | - Henrik von der Emde
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Marcelo Ganzella
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Leon Bösche
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Viveka Nand Malviya
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Evi Zhuleku
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Antonio Zaccaria Politi
- Live-Cell Imaging Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Momchil Ninov
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Marcel Leutenegger
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Dietmar Riedel
- Department of Structural Dynamics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Julia Preobraschenski
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen 37075, Germany
| | - Ira Milosevic
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7BN, UK; Multidisciplinary Institute of Ageing, MIA-Portugal, University of Coimbra, Coimbra 3000-370, Portugal
| | - Stefan W Hell
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg 69028, Germany
| | - Reinhard Jahn
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sivakumar Sambandan
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany.
| |
Collapse
|
15
|
Woelfle S, Boeckers TM. Layer-Specific Vesicular Glutamate Transporter 1 Immunofluorescence Levels Delineate All Layers of the Human Hippocampus Including the Stratum lucidum. Front Cell Neurosci 2021; 15:789903. [PMID: 34955756 PMCID: PMC8696355 DOI: 10.3389/fncel.2021.789903] [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: 10/05/2021] [Accepted: 11/18/2021] [Indexed: 11/18/2022] Open
Abstract
The hippocampal formation consists of the Ammon’s horn (cornu Ammonis with its regions CA1-4), dentate gyrus, subiculum, and the entorhinal cortex. The rough extension of the regions CA1-3 is typically defined based on the density and size of the pyramidal neurons without clear-cut boundaries. Here, we propose the vesicular glutamate transporter 1 (VGLUT1) as a molecular marker for the CA3 region. This is based on its strong labeling of the stratum lucidum (SL) in fluorescently stained human hippocampus sections. VGLUT1 puncta of the intense SL band co-localize with synaptoporin (SPO), a protein enriched in mossy fibers (MFs). Owing to its specific intensity profile throughout all hippocampal layers, VGLUT1 could be implemented as a pendant to Nissl-staining in fluorescent approaches with the additional demarcation of the SL. Furthermore, by high-resolution confocal microscopy, we detected VGLUT2 in the human hippocampus, thus reconciling two previous studies. Finally, by VGLUT1/SPO co-staining, we provide evidence for the existence of infrapyramidal MFs in the human hippocampus and we show that SPO expression is not restricted to MF synapses as demonstrated for rodent tissue.
Collapse
Affiliation(s)
- Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,International Graduate School in Molecular Medicine Ulm (IGradU), Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| |
Collapse
|
16
|
Abbas Farishta R, Zouahi H, Casanova C. Distributions of vesicular glutamate transporters 1 and 2 in the visual thalamus and associated areas of the cat. J Comp Neurol 2021; 530:1112-1125. [PMID: 34468980 DOI: 10.1002/cne.25239] [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: 02/01/2021] [Revised: 07/20/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
Glutamate is packaged in vesicles via two main vesicular transporter (VGLUT) proteins, VGLUT1 and VGLUT2, which regulate its storage and release from synapses of excitatory neurons. Studies in rodents, primates, ferrets, and tree shrews suggest that these transporters may identify distinct subsets of excitatory projections in visual structures, particularly in thalamocortical pathways where they tend to correlate with modulatory and driver projections, respectively. Despite being a well-studied model of thalamocortical connectivity, little is known about their expression pattern in the cat visual system. To expand current knowledge on their distribution and how they correlated with known driver and modulator projecting sites, we examined the protein expression patterns of VGLUT1 and VGLUT2 in the visual thalamus of the cat (lateral geniculate nucleus and the pulvinar complex). We also studied their expression pattern in relevant visual structures projecting to or receiving significant thalamic projections, such as the primary visual cortex and the superior colliculus. Our results indicate that both VGLUTs are consistently present throughout the cat visual system and show laminar or nuclei specificity in their distribution, which suggests, as in other species, that VGLUT1 and VGLUT2 represent distinct populations of glutamatergic projections.
Collapse
Affiliation(s)
- Reza Abbas Farishta
- Laboratoire des neurosciences de la vision, École d'optométrie, Université de Montréal, Montréal, Québec, Canada
| | - Hadjer Zouahi
- Laboratoire des neurosciences de la vision, École d'optométrie, Université de Montréal, Montréal, Québec, Canada
| | - Christian Casanova
- Laboratoire des neurosciences de la vision, École d'optométrie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
Shank3 Deficiency is Associated With Altered Profile of Neurotransmission Markers in Pups and Adult Mice. Neurochem Res 2021; 46:3342-3355. [PMID: 34453663 DOI: 10.1007/s11064-021-03435-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/04/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the balance between excitation and inhibition, especially in the brain's critical developmental periods, are considered an integral part of the pathophysiology of autism. However, the precise mechanisms have not yet been established. SH3 and multiple Ankyrin repeat domains 3 (Shank3) deficient mice represent a well-established transgenic model of a neurodevelopmental disorder with autistic symptomatology. In this study, we characterize the consequences of Shank3 deficiency according to (1) expression of specific markers of different neuronal populations in pups and adult mice and (2) social behaviour and anxiety in adult mice. Our research found enhanced expression of serotonin transporter and choline acetyltransferase in the hippocampus and hypothalamus in Shank3-deficient pups. We demonstrated marked brain region differences in expression of excitatory glutamatergic markers in pups and adult Shank3 deficient mice. We also observed reduced expression of inhibitory GABAergic markers and GABA receptor subunits in several brain areas in both pups and adult Shank3 deficient mice. Further analysis of dopaminergic brain areas (nucleus accumbens, ventral tegmental area) revealed lower expression levels of GABAergic markers in adult Shank3 deficient mice. Adult Shank3- deficient mice exhibited excessive repetitive behaviour, a higher level of anxiety, and lower locomotor activity. Our data support the theory of an imbalance between excitatory and inhibitory neurotransmission in conditions of abnormal SHANK3 protein. We therefore suggest that autism-like conditions are accompanied by reduced expression of GABAergic markers in the brain during early development as well as in the adult age, which could be associated with long-lasting behavioural abnormalities.
Collapse
|
18
|
Garrudo FFF, Nogueira DES, Rodrigues CAV, Ferreira FA, Paradiso P, Colaço R, Marques AC, Cabral JMS, Morgado J, Linhardt RJ, Ferreira FC. Electrical stimulation of neural-differentiating iPSCs on novel coaxial electroconductive nanofibers. Biomater Sci 2021; 9:5359-5382. [PMID: 34223566 DOI: 10.1039/d1bm00503k] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neural tissue engineering strategies are paramount to create fully mature neurons, necessary for new therapeutic strategies for neurological diseases or the creation of reliable in vitro models. Scaffolds can provide physical support for these neurons and enable cues for enhancing neural cell differentiation, such as electrical current. Coaxial electrospinning fibers, designed to fulfill neural cell needs, bring together an electroconductive shell layer (PCL-PANI), able to mediate electrical stimulation of cells cultivated on fibers mesh surface, and a soft core layer (PGS), used to finetune fiber diameter (951 ± 465 nm) and mechanical properties (1.3 ± 0.2 MPa). Those dual functional coaxial fibers are electroconductive (0.063 ± 0.029 S cm-1, stable over 21 days) and biodegradable (72% weigh loss in 12 hours upon human lipase accelerated assay). For the first time, the long-term effects of electrical stimulation on induced neural progenitor cells were studied using such fibers. The results show increase in neural maturation (upregulation of MAP2, NEF-H and SYP), up-regulation of glutamatergic marker genes (VGLUT1 - 15-fold) and voltage-sensitive channels (SCN1α - 12-fold, CACNA1C - 32-fold), and a down-regulation of GABAergic marker (GAD67 - 0.09-fold), as detected by qRT-PCR. Therefore, this study suggest a shift from an inhibitory to an excitatory neural cell profile. This work shows that the PGS/PCL-PANI coaxial fibers here developed have potential applications in neural tissue engineering.
Collapse
Affiliation(s)
- Fábio F F Garrudo
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA. and Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal and Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Diogo E S Nogueira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Flávio A Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Patrizia Paradiso
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Rogério Colaço
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Ana C Marques
- CERENA, DEQ, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge Morgado
- Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA.
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
19
|
Leveraging VGLUT3 Functions to Untangle Brain Dysfunctions. Trends Pharmacol Sci 2021; 42:475-490. [PMID: 33775453 DOI: 10.1016/j.tips.2021.03.003] [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: 10/12/2020] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 11/21/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) were long thought to be specific markers of glutamatergic excitatory transmission. The discovery, two decades ago, of the atypical VGLUT3 has thoroughly modified this oversimplified view. VGLUT3 is strategically expressed in discrete populations of glutamatergic, cholinergic, serotonergic, and even GABAergic neurons. Recent reports show the subtle, but critical, implications of VGLUT3-dependent glutamate co-transmission and its roles in the regulation of diverse brain functions and dysfunctions. Progress in the neuropharmacology of VGLUT3 could lead to decisive breakthroughs in the treatment of Parkinson's disease (PD), addiction, eating disorders, anxiety, presbycusis, or pain. This review summarizes recent findings on VGLUT3 and its vesicular underpinnings as well as on possible ways to target this atypical transporter for future therapeutic strategies.
Collapse
|
20
|
Horie S, Kiyokage E, Hayashi S, Inoue K, Sohn J, Hioki H, Furuta T, Toida K. Structural basis for noradrenergic regulation of neural circuits in the mouse olfactory bulb. J Comp Neurol 2021; 529:2189-2208. [PMID: 33616936 DOI: 10.1002/cne.25085] [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: 04/24/2020] [Revised: 11/19/2020] [Accepted: 11/29/2020] [Indexed: 11/09/2022]
Abstract
Olfactory input is processed in the glomerulus of the main olfactory bulb (OB) and relayed to higher centers in the brain by projection neurons. Conversely, centrifugal inputs from other brain regions project to the OB. We have previously analyzed centrifugal inputs into the OB from several brain regions using single-neuron labeling. In this study, we analyzed the centrifugal noradrenergic (NA) fibers derived from the locus coeruleus (LC), because their projection pathways and synaptic connections in the OB have not been clarified in detail. We analyzed the NA centrifugal projections by single-neuron labeling and immunoelectron microscopy. Individual NA neurons labeled by viral infection were three-dimensionally traced using Neurolucida software to visualize the projection pathway from the LC to the OB. Also, centrifugal NA fibers were visualized using an antibody for noradrenaline transporter (NET). NET immunoreactive (-ir) fibers contained many varicosities and synaptic vesicles. Furthermore, electron tomography demonstrated that NET-ir fibers formed asymmetrical synapses of varied morphology. Although these synapses were present at varicosities, the density of synapses was relatively low throughout the OB. The maximal density of synapses was found in the external plexiform layer; about 17% of all observed varicosities contained synapses. These results strongly suggest that NA-containing fibers in the OB release NA from both varicosities and synapses to influence the activities of OB neurons. The present study provides a morphological basis for olfactory modulation by centrifugal NA fibers derived from the LC.
Collapse
Affiliation(s)
- Sawa Horie
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Emi Kiyokage
- Department of Medical Technology, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Shuichi Hayashi
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kanako Inoue
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Jaerin Sohn
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Hiroyuki Hioki
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takahiro Furuta
- Department of Oral Anatomy and Neurobiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Kazunori Toida
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Okayama, Japan.,Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| |
Collapse
|
21
|
Terayama R, Uchibe K. Reorganization of synaptic inputs to spinal dorsal horn neurons in neuropathic pain. Int J Neurosci 2021; 132:1210-1216. [PMID: 33428497 DOI: 10.1080/00207454.2021.1873980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Peripheral nerve injuries produce a variety of negative structural and functional changes in the central terminal sites of damaged axons, as well as the injured primary afferents. Such changes have been shown to be involved in the development of neuropathic pain, which includes abnormal pain sensations such as allodynia and hyperalgesia. Since the spinal dorsal horn is the first central site where signals from peripheral sensory nerves are transmitted and shows a variety of changes after peripheral nerve injury or chronic inflammation of peripheral tissues, it is one of the most important sites contributing to the mechanisms underlying the development of neuropathic pain. The functional disruption of inhibitory interneurons and glial activation in the spinal dorsal horn after peripheral nerve injury cause reorganization of neuronal circuits and changes in the excitability of second-order neurons. These events are involved in the development or maintenance of neuropathic pain. Here, we describe the interactions of primary afferents, interneurons, and glial cells that may cause reorganization of synaptic inputs to spinal dorsal horn neurons after peripheral nerve injury.
Collapse
Affiliation(s)
- Ryuji Terayama
- Department of Maxillofacial Anatomy and Neuroscience, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kenta Uchibe
- Department of Maxillofacial Anatomy and Neuroscience, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
22
|
Zheng RZ, Xing J, Huang Q, Yang XT, Zhao CY, Li XY. Integration of single-cell and bulk RNA sequencing data reveals key cell types and regulators in traumatic brain injury. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:1201-1214. [PMID: 33757183 DOI: 10.3934/mbe.2021065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability and mortality worldwide, whose symptoms ranging from mild to severe, even life-threatening. However, specific cell types and key regulators involved in traumatic brain injury have not been well elucidated. In this study, utilizing single-cell RNA-seq (scRNA-seq) data from mice with TBI, we have successfully identified and characterized 13 cell populations including astrocytes, oligodendrocyte, newly formed oligodendrocytes, microglia, two types of endothelial cells, five types of excitatory and two types of inhibitory neurons. Differential expression analysis and gene set enrichment analysis (GSEA) revealed the upregulation of microglia and endothelial markers, along with the downregulation of markers of excitatory neurons in TBI. The cell-cell communication analysis revealed that microglia and endothelial cell might interact through the interaction of Icam1-Il2rg and C1qa-Cd93, and microglia might also communicate with each other via Icam1-Itagm. The autocrine ligand-receptor in microglia might result in activation of TYROBP causal network via Icam1-Itgam. The cell-cell contact between microglia and endothelial cell might activate integrin signaling pathways. Moreover, we also found that genes involved in microglia activation were highly downregulated in Tyrobp/Dap12-deficient microglia, indicating that the upregulation of Tyrobp and TYROBP causal network in microglia might be a candidate therapeutic target in TBI. In contrast, the excitatory neurons were involved in maintaining normal brain function, and their inactivation might cause dysfunction of nervous system in TBI patients. In conclusion, the present study has discerned major cell types such as microglia, endothelial cells and excitatory neurons, and revealed key regulator such as TYROBP, C1QA, and CD93 in TBI, which shall improve our understanding of the pathogenesis of TBI.
Collapse
Affiliation(s)
- Rui-Zhe Zheng
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jin Xing
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Qiong Huang
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xi-Tao Yang
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chang-Yi Zhao
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xin-Yuan Li
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
23
|
Moore C, Xu M, Bohlen JK, Meshul CK. Differential ultrastructural alterations in the Vglut2 glutamatergic input to the substantia nigra pars compacta/pars reticulata following nigrostriatal dopamine loss in a progressive mouse model of Parkinson’s disease. Eur J Neurosci 2020; 53:2061-2077. [DOI: 10.1111/ejn.14894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Cynthia Moore
- Research ServicesVA Medical Center/Portland Portland OR USA
| | - Mo Xu
- Research ServicesVA Medical Center/Portland Portland OR USA
| | | | - Charles K. Meshul
- Research ServicesVA Medical Center/Portland Portland OR USA
- Department of Behavioral Neuroscience and Pathology Oregon Heath & Science University Portland OR USA
| |
Collapse
|
24
|
Excitatory VTA to DH projections provide a valence signal to memory circuits. Nat Commun 2020; 11:1466. [PMID: 32193428 PMCID: PMC7081331 DOI: 10.1038/s41467-020-15035-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 02/11/2020] [Indexed: 12/21/2022] Open
Abstract
The positive or negative value (valence) of past experiences is normally integrated into neuronal circuits that encode episodic memories and plays an important role in guiding behavior. Here, we show, using mouse behavioral models, that glutamatergic afferents from the ventral tegmental area to the dorsal hippocampus (VTA→DH) signal negative valence to memory circuits, leading to the formation of fear-inducing context memories and to context-specific reinstatement of fear. To a lesser extent, these projections also contributed to opioid-induced place preference, suggesting a role in signaling positive valence as well, and thus a lack of dedicated polarity. Manipulations of VTA terminal activity were more effective in females and paralleled by sex differences in glutamatergic signaling. By prioritizing retrieval of negative and positive over neutral memories, the VTA→DH circuit can facilitate the selection of adaptive behaviors when current and past experiences are valence congruent. The neuronal pathway that signals the positive or negative value of memories is not well understood. Here, the authors report that an excitatory projection from the ventral tegmental area to the dorsal hippocampus carries the valence information, contributing, especially in females, to the recurrence of fear and to drug seeking behavior.
Collapse
|
25
|
Sakaguchi S, Shintani S, Kamio K, Sekiya A, Kato S, Muroi Y, Horiuchi M, Furuoka H. Selective neuronal vulnerability is involved in cerebellar lesions of Guinea pigs infected with bovine spongiform encephalopathy (BSE) prions: Immunohistochemical and electron microscopic investigations. Neuropathology 2019; 40:167-179. [PMID: 31797465 DOI: 10.1111/neup.12613] [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: 08/14/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 11/30/2022]
Abstract
The cerebellar lesions of bovine spongiform encephalopathy (BSE)-infected guinea pigs were characterized as severe atrophy of the cerebellar cortex associated with the loss of granule cells, decrease in the width of the molecular layer, and intense protease-resistant prion protein (PrPSc ) accumulations that are similar to cerebellar lesions in kuru and the VV2 type of sporadic Creutzfeldt-Jakob disease. The aim of this study is to assess the relationships between the distribution and localization of PrPSc and synapses expressing neurotransmitter transporters in order to reveal the pathogenesis of the disease. We used cell-type-specific immunohistochemical makers recognizing glutamatergic and γ-aminobutylic acid (GABA)ergic terminals to identify terminals impaired with PrPSc accumulations. The distribution of PrPSc accumulations and immunoreactivity of synaptic vesicles were studied throughout the neuroanatomical pathways in cerebellar lesions. Time course study demonstrated that PrPSc accumulation showed a tendency to spread from granular layer to molecular layer. The immunoreactivity of vesicular glutamate transporter 1 (VGluT1) was localized in axon terminals of cerebellar granule cells, and decreased in association with the severity of PrPSc accumulations and loss of granule cells. Immunoreactivities of vesicular glutamate transporter 2 (VGluT2) and vesicular GABA transporter (VGAT) that exist in axon terminals of inferior olivary neurons and GABAergic synapses of Purkinje cells, respectively, were preserved well in these lesions. In brainstem, VGluT1 immunoreactivity decreased selectively in pontine nuclei that are a component of the pontocerebellar pathway, although other neurotransmitter immunoreactivities were preserved well. Our findings suggest that the selective loss of VGluT1-immunoreactive synapses subsequent to PrPSc accumulations can contribute to the pathogenesis of cerebellar lesions of BSE-infected guinea pigs.
Collapse
Affiliation(s)
- Shoichi Sakaguchi
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Sayo Shintani
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Kyohei Kamio
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Akio Sekiya
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Satomi Kato
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Yoshikage Muroi
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Motohiro Horiuchi
- Laboratory of Veterinary Hygiene, Faculty of Veterinary Medicine, Graduate School of Infectious Diseases, Hokkaido University, Sapporo, Japan
| | - Hidefumi Furuoka
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| |
Collapse
|
26
|
The solute carrier transporters and the brain: Physiological and pharmacological implications. Asian J Pharm Sci 2019; 15:131-144. [PMID: 32373195 PMCID: PMC7193445 DOI: 10.1016/j.ajps.2019.09.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/17/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023] Open
Abstract
Solute carriers (SLCs) are the largest family of transmembrane transporters that determine the exchange of various substances, including nutrients, ions, metabolites, and drugs across biological membranes. To date, the presence of about 287 SLC genes have been identified in the brain, among which mutations or the resultant dysfunctions of 71 SLC genes have been reported to be correlated with human brain disorders. Although increasing interest in SLCs have focused on drug development, SLCs are currently still under-explored as drug targets, especially in the brain. We summarize the main substrates and functions of SLCs that are expressed in the brain, with an emphasis on selected SLCs that are important physiologically, pathologically, and pharmacologically in the blood-brain barrier, astrocytes, and neurons. Evidence suggests that a fraction of SLCs are regulated along with the occurrences of brain disorders, among which epilepsy, neurodegenerative diseases, and autism are representative. Given the review of SLCs involved in the onset and procession of brain disorders, we hope these SLCs will be screened as promising drug targets to improve drug delivery to the brain.
Collapse
|
27
|
The functional microscopic neuroanatomy of the human subthalamic nucleus. Brain Struct Funct 2019; 224:3213-3227. [PMID: 31562531 PMCID: PMC6875153 DOI: 10.1007/s00429-019-01960-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/14/2019] [Indexed: 01/19/2023]
Abstract
The subthalamic nucleus (STN) is successfully used as a surgical target for deep brain stimulation in the treatment of movement disorders. Interestingly, the internal structure of the STN is still incompletely understood. The objective of the present study was to investigate three-dimensional (3D) immunoreactivity patterns for 12 individual protein markers for GABA-ergic, serotonergic, dopaminergic as well as glutamatergic signaling. We analyzed the immunoreactivity using optical densities and created a 3D reconstruction of seven postmortem human STNs. Quantitative modeling of the reconstructed 3D immunoreactivity patterns revealed that the applied protein markers show a gradient distribution in the STN. These gradients were predominantly organized along the ventromedial to dorsolateral axis of the STN. The results are of particular interest in view of the theoretical underpinning for surgical targeting, which is based on a tripartite distribution of cognitive, limbic and motor function in the STN.
Collapse
|
28
|
Yamawaki N, Corcoran KA, Guedea AL, Shepherd GMG, Radulovic J. Differential Contributions of Glutamatergic Hippocampal→Retrosplenial Cortical Projections to the Formation and Persistence of Context Memories. Cereb Cortex 2019; 29:2728-2736. [PMID: 29878069 PMCID: PMC6519694 DOI: 10.1093/cercor/bhy142] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/02/2018] [Indexed: 12/20/2022] Open
Abstract
Learning to associate stressful events with specific environmental contexts depends on excitatory transmission in the hippocampus, but how this information is transmitted to the neocortex for lasting memory storage is unclear. We identified dorsal hippocampal (DH) projections to the retrosplenial cortex (RSC), which arise mainly from the subiculum and contain either the vesicular glutamate transporter 1 (vGlut1) or vGlut2. Both vGlut1+ and vGlut2+ axons strongly excite and disynaptically inhibit RSC pyramidal neurons in superficial layers, but vGlut2+ axons trigger greater inhibition that spreads to deep layers, indicating that these pathways engage RSC circuits via partially redundant, partially differentiated cellular mechanisms. Using contextual fear conditioning in mice to model contextual associative memories, together with chemogenetic axonal silencing, we found that vGlut1+ projections are principally involved in processing recent context memories whereas vGlut2+ projections contribute to their long-lasting storage. Thus, within the DH→RSC pathway, engagement of vGlut1+ and vGlut2+ circuits differentially contribute to the formation and persistence of fear-inducing context memories.
Collapse
Affiliation(s)
- Naoki Yamawaki
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Kevin A Corcoran
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Anita L Guedea
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Gordon M G Shepherd
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Jelena Radulovic
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
29
|
Dorgans K, Demais V, Bailly Y, Poulain B, Isope P, Doussau F. Short-term plasticity at cerebellar granule cell to molecular layer interneuron synapses expands information processing. eLife 2019; 8:41586. [PMID: 31081751 PMCID: PMC6533085 DOI: 10.7554/elife.41586] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 05/11/2019] [Indexed: 12/14/2022] Open
Abstract
Information processing by cerebellar molecular layer interneurons (MLIs) plays a crucial role in motor behavior. MLI recruitment is tightly controlled by the profile of short-term plasticity (STP) at granule cell (GC)-MLI synapses. While GCs are the most numerous neurons in the brain, STP diversity at GC-MLI synapses is poorly documented. Here, we studied how single MLIs are recruited by their distinct GC inputs during burst firing. Using slice recordings at individual GC-MLI synapses of mice, we revealed four classes of connections segregated by their STP profile. Each class differentially drives MLI recruitment. We show that GC synaptic diversity is underlain by heterogeneous expression of synapsin II, a key actor of STP and that GC terminals devoid of synapsin II are associated with slow MLI recruitment. Our study reveals that molecular, structural and functional diversity across GC terminals provides a mechanism to expand the coding range of MLIs.
Collapse
Affiliation(s)
- Kevin Dorgans
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, Université de Strasbourg, Strasbourg, France
| | - Valérie Demais
- Plateforme Imagerie in vitro, CNRS UPS 3156, Strasbourg, France
| | - Yannick Bailly
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, Université de Strasbourg, Strasbourg, France.,Plateforme Imagerie in vitro, CNRS UPS 3156, Strasbourg, France
| | - Bernard Poulain
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, Université de Strasbourg, Strasbourg, France
| | - Philippe Isope
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, Université de Strasbourg, Strasbourg, France
| | - Frédéric Doussau
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
30
|
Kourakis MJ, Borba C, Zhang A, Newman-Smith E, Salas P, Manjunath B, Smith WC. Parallel visual circuitry in a basal chordate. eLife 2019; 8:44753. [PMID: 30998184 PMCID: PMC6499539 DOI: 10.7554/elife.44753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/11/2019] [Indexed: 12/28/2022] Open
Abstract
A common CNS architecture is observed in all chordates, from vertebrates to basal chordates like the ascidian Ciona. Ciona stands apart among chordates in having a complete larval connectome. Starting with visuomotor circuits predicted by the Ciona connectome, we used expression maps of neurotransmitter use with behavioral assays to identify two parallel visuomotor circuits that are responsive to different components of visual stimuli. The first circuit is characterized by glutamatergic photoreceptors and responds to the direction of light. These photoreceptors project to cholinergic motor neurons, via two tiers of cholinergic interneurons. The second circuit responds to changes in ambient light and mediates an escape response. This circuit uses GABAergic photoreceptors which project to GABAergic interneurons, and then to cholinergic interneurons. Our observations on the behavior of larvae either treated with a GABA receptor antagonist or carrying a mutation that eliminates photoreceptors indicate the second circuit is disinhibitory.
Collapse
Affiliation(s)
- Matthew J Kourakis
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - Cezar Borba
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - Angela Zhang
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Erin Newman-Smith
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States.,Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - Priscilla Salas
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| | - B Manjunath
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - William C Smith
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States.,Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, United States
| |
Collapse
|
31
|
Jones GL, Wittmann G, Yokosawa EB, Yu H, Mercer AJ, Lechan RM, Low MJ. Selective Restoration of Pomc Expression in Glutamatergic POMC Neurons: Evidence for a Dynamic Hypothalamic Neurotransmitter Network. eNeuro 2019; 6:ENEURO.0400-18.2019. [PMID: 30957016 PMCID: PMC6449166 DOI: 10.1523/eneuro.0400-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 01/19/2023] Open
Abstract
Hypothalamic POMC deficiency leads to obesity and metabolic deficiencies, largely due to the loss of melanocortin peptides. However, POMC neurons in the arcuate nucleus (ARC) are comprised of glutamatergic and GABAergic subpopulations. The developmental program, relative proportion and function of these two subpopulations are unresolved. To test whether glutamatergic POMC neurons serve a distinct role in maintaining energy homeostasis, we activated Pomc expression Cre- dependently in Vglut2-expressing neurons of mice with conditionally silenced Pomc alleles. The Vglut2-Pomc restored mice had normal ARC Pomc mRNA levels, POMC immunoreactivity, as well as body weight and body composition at age 12 weeks. Unexpectedly, the cumulative total of Vglut2+ glutamatergic- and Gad67+ GABAergic-Pomc neurons detected by in situ hybridization (ISH) exceeded 100% in both Vglut2- Pomc restored and control mice, indicating that a subpopulation of Pomc neurons must express both neuronal markers. Consistent with this hypothesis, triple ISH of C57BL/6J hypothalami revealed that 35% of ARC Pomc neurons were selectively Gad67+, 21% were selectively Vglut2+, and 38% expressed both Gad67 and Vglut2. The single Gad67+ and Vglut2+Pomc neurons were most prevalent in the rostral ARC, while the Vglut2/Gad67+ dual-phenotype cells predominated in the caudal ARC. A lineage trace using Ai9-tdTomato reporter mice to label fluorescently all Vglut2-expressing neurons showed equal numbers of tdTomato+ and tdTomato- POMC immunoreactive neurons. Together, these data suggest that POMC neurons exhibit developmental plasticity in their expression of glutamatergic and GABAergic markers, enabling re-establishment of normal energy homeostasis in the Vglut2-Pomc restored mice.
Collapse
Affiliation(s)
- Graham L. Jones
- Neuroscience Graduate Program
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Gábor Wittmann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, Boston, MA 02111
| | - Eva B. Yokosawa
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Hui Yu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Aaron J. Mercer
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Ronald M. Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, Boston, MA 02111
| | - Malcolm J. Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105
| |
Collapse
|
32
|
Deng B, Li Q, Liu X, Cao Y, Li B, Qian Y, Xu R, Mao R, Zhou E, Zhang W, Huang J, Rao Y. Chemoconnectomics: Mapping Chemical Transmission in Drosophila. Neuron 2019; 101:876-893.e4. [PMID: 30799021 DOI: 10.1016/j.neuron.2019.01.045] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/02/2018] [Accepted: 01/17/2019] [Indexed: 12/27/2022]
Abstract
We define the chemoconnectome (CCT) as the entire set of neurotransmitters, neuromodulators, neuropeptides, and their receptors underlying chemotransmission in an animal. We have generated knockout lines of Drosophila CCT genes for functional investigations and knockin lines containing Gal4 and other tools for examining gene expression and manipulating neuronal activities, with a versatile platform allowing genetic intersections and logic gates. CCT reveals the coexistence of specific transmitters but mutual exclusion of the major inhibitory and excitatory transmitters in the same neurons. One neuropeptide and five receptors were detected in glia, with octopamine β2 receptor functioning in glia. A pilot screen implicated 41 genes in sleep regulation, with the dopamine receptor Dop2R functioning in neurons expressing the peptides Dilp2 and SIFa. Thus, CCT is a novel concept, chemoconnectomics a new approach, and CCT tool lines a powerful resource for systematic investigations of chemical-transmission-mediated neural signaling circuits underlying behavior and cognition.
Collapse
Affiliation(s)
- Bowen Deng
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Qi Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Xinxing Liu
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yue Cao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Bingfeng Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yongjun Qian
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Rui Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Renbo Mao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Enxing Zhou
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Wenxia Zhang
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Juan Huang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China.
| |
Collapse
|
33
|
Trudeau LE, El Mestikawy S. Glutamate Cotransmission in Cholinergic, GABAergic and Monoamine Systems: Contrasts and Commonalities. Front Neural Circuits 2018; 12:113. [PMID: 30618649 PMCID: PMC6305298 DOI: 10.3389/fncir.2018.00113] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/03/2018] [Indexed: 11/13/2022] Open
Abstract
Multiple discoveries made since the identification of vesicular glutamate transporters (VGLUTs) two decades ago revealed that many neuronal populations in the brain use glutamate in addition to their "primary" neurotransmitter. Such a mode of cotransmission has been detected in dopamine (DA), acetylcholine (ACh), serotonin (5-HT), norepinephrine (NE) and surprisingly even in GABA neurons. Interestingly, work performed by multiple groups during the past decade suggests that the use of glutamate as a cotransmitter takes different forms in these different populations of neurons. In the present review, we will provide an overview of glutamate cotransmission in these different classes of neurons, highlighting puzzling differences in: (1) the proportion of such neurons expressing a VGLUT in different brain regions and at different stages of development; (2) the sub-cellular localization of the VGLUT; (3) the localization of the VGLUT in relation to the neurons' other vesicular transporter; and (4) the functional role of glutamate cotransmission.
Collapse
Affiliation(s)
- Louis-Eric Trudeau
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Salah El Mestikawy
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Sorbonne Universités, Université Pierre et Marie Curie UM 119-CNRS UMR 8246-INSERM U1130, Neurosciences Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS), Paris, France
| |
Collapse
|
34
|
β-actin regulates a heterochromatin landscape essential for optimal induction of neuronal programs during direct reprograming. PLoS Genet 2018; 14:e1007846. [PMID: 30557298 PMCID: PMC6312353 DOI: 10.1371/journal.pgen.1007846] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 12/31/2018] [Accepted: 11/21/2018] [Indexed: 02/02/2023] Open
Abstract
During neuronal development, β-actin serves an important role in growth cone mediated axon guidance. Consistent with this notion, in vivo ablation of the β-actin gene leads to abnormalities in the nervous system. However, whether β-actin is involved in the regulation of neuronal gene programs is not known. In this study, we directly reprogramed β-actin+/+ WT, β-actin+/- HET and β-actin-/- KO mouse embryonic fibroblast (MEFs) into chemically induced neurons (CiNeurons). Using RNA-seq analysis, we profiled the transcriptome changes among the CiNeurons. We discovered that induction of neuronal gene programs was impaired in KO CiNeurons in comparison to WT ones, whereas HET CiNeurons showed an intermediate levels of induction. ChIP-seq analysis of heterochromatin markers demonstrated that the impaired expression of neuronal gene programs correlated with the elevated H3K9 and H3K27 methylation levels at gene loci in β-actin deficient MEFs, which is linked to the loss of chromatin association of the BAF complex ATPase subunit Brg1. Together, our study shows that heterochromatin alteration in β-actin null MEFs impedes the induction of neuronal gene programs during direct reprograming. These findings are in line with the notion that H3K9Me3-based heterochromatin forms a major epigenetic barrier during cell fate change. Although β-actin plays an important role in growth cone mediated axon guidance in neurons, the potential role of β-actin in controlling neuron differentiation remains unknown. Here, we converted β-actin+/+ WT, β-actin+/- HET and β-actin-/- KO mouse embryonic fibroblast (MEFs) into chemically induced neurons (CiNeurons) by direct reprograming. We found that the up-regulation of neuronal programs was impaired in β-actin-/- CiNeurons in comparison to WT ones. β-actin+/- HET CiNeurons showed an intermediate level of neuronal program expression, suggesting that β-actin dosage plays an important role during direct neuronal reprograming. Importantly, the impaired up-regulation of neuron-related genes was associated with the elevated H3K9 and H3K27 methylation levels at gene loci in KO MEFs. These epigenetic changes were accompanied by the impaired chromatin association of Brg1-containing chromatin remodeling BAF complex in β-actin null cells. Together our study demonstrates that β-actin is required for the optimal induction of neuronal gene programs during direct reprograming by presetting a favorable chromatin status.
Collapse
|
35
|
Moore AM, Abbott G, Mair J, Prescott M, Campbell RE. Mapping GABA and glutamate inputs to gonadotrophin-releasing hormone neurones in male and female mice. J Neuroendocrinol 2018; 30:e12657. [PMID: 30415474 DOI: 10.1111/jne.12657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 11/28/2022]
Abstract
Gonadotrophin-releasing hormone (GnRH) neurone function is dependent upon gonadal steroid hormone feedback, which is communicated in large part through an afferent neuronal network. The classical neurotransmitters GABA and glutamate are important regulators of GnRH neurone activity and are implicated in mediating feedback signals. In the present study, we aimed to determine whether GABAergic or glutamatergic input to GnRH neurones differs between males and females and/or exhibits morphological plasticity in response to steroid hormone feedback in females. Tissue collected from GnRH-green fluorescent protein (GFP) male and female mice in dioestrus underwent immunofluorescence labelling of GFP and either the vesicular GABA transporter (VGAT) or the vesicular glutamate transporter 2 (VGLUT2). No differences in the densities or absolute numbers of VGAT-immunoreactive (-IR) or VGLUT2-IR puncta apposed to GnRH neurones were identified between males and females. The most significant input from either neurotransmitter was to the proximal dendritic region and 80% of VGAT-IR puncta apposed to GnRH neurones co-localised with synaptophysin. Putative inputs were also assessed in ovariectomised (OVX) female mice treated with negative (OVX+E) or positive (OVX+E+E) feedback levels of oestrogen, and OVX+E+E mice were killed during the expected GnRH/luteinising hormone surge. No differences in VGLUT2-IR contacts to GnRH neurones were identified between animals under the negative-feedback influence of oestrogen (OVX+E) or the positive influence of oestrogen (OVX+E+E), regardless of cFos activation status. By contrast, a significant elevation in putative GABAergic inputs to GnRH neurones at the time of the preovulatory surge was found in the cFos-negative subset of GnRH neurones, both at the level of the soma and at the proximal dendrite. Taken together, these data suggest that, although GABAergic and glutamatergic innervation of GnRH neurones is not sexually differentiated, cyclic fluctuations in steroid hormone feedback over the female oestrous cycle result in plastic changes in GABAergic inputs to a subpopulation of GnRH neurones.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Georgina Abbott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jonathan Mair
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
36
|
Tjaden J, Pieczora L, Wach F, Theiss C, Theis V. Cultivation of Purified Primary Purkinje Cells from Rat Cerebella. Cell Mol Neurobiol 2018; 38:1399-1412. [PMID: 30066224 DOI: 10.1007/s10571-018-0606-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/24/2018] [Indexed: 10/28/2022]
Abstract
Primary neurons are difficult to cultivate because they are often part of a complex tissue, and synaptically connected to numerous other cell types. These circumstances often prevent us from unveiling molecular and metabolic mechanisms of distinct cells, as functional signals or assays cannot clearly be correlated with them due to interfering signals from other parts of the culture. We therefore present an up-to-date method for obtaining a highly purified neuronal culture of Purkinje cells. In the past, Purkinje cells were successfully isolated from young mouse cerebella, but this protocol was never adapted to other mammals. We therefore provide an updated and adjusted protocol for Purkinje cell isolation from rat instead of mouse cerebella. To purify Purkinje cells, we obtained perinatal rat cerebella, dissociated them and performed a Percoll gradient centrifugation to segregate the smaller and larger cell fractions. In a second step, we performed an immunopanning procedure to enrich only Purkinje cells from the large cell fraction. Based on former protocols, we used a different antibody for the immunopanning procedure and adjusted several aspects from the initial protocol to improve the yield and vitality of Purkinje cells. We provide RT-qPCR-based purity data obtained with this protocol and show the behaviour and the growth of these purified Purkinje cells. We provide a highly reproducible purification protocol for Purkinje cell cultures of high purity that allows functional analysis and downstream assays on living rat Purkinje cells and further morphological growth analysis in future.
Collapse
Affiliation(s)
- Jonas Tjaden
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Lukas Pieczora
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Frederique Wach
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| |
Collapse
|
37
|
Azeez IA, Del Gallo F, Cristino L, Bentivoglio M. Daily Fluctuation of Orexin Neuron Activity and Wiring: The Challenge of "Chronoconnectivity". Front Pharmacol 2018; 9:1061. [PMID: 30319410 PMCID: PMC6167434 DOI: 10.3389/fphar.2018.01061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
In the heterogeneous hub represented by the lateral hypothalamus, neurons containing the orexin/hypocretin peptides play a key role in vigilance state transitions and wakefulness stability, energy homeostasis, and other functions relevant for motivated behaviors. Orexin neurons, which project widely to the neuraxis, are innervated by multiple extra- and intra-hypothalamic sources. A key property of the adaptive capacity of orexin neurons is represented by daily variations of activity, which is highest in the period of the animal’s activity and wakefulness. These sets of data are here reviewed. They concern the discharge profile during the sleep/wake cycle, spontaneous Fos induction, peptide synthesis and release reflected by immunostaining intensity and peptide levels in the cerebrospinal fluid as well as postsynaptic effects. At the synaptic level, adaptive capacity of orexin neurons subserved by remodeling of excitatory and inhibitory inputs has been shown in response to changes in the nutritional status and prolonged wakefulness. The present review wishes to highlight that synaptic plasticity in the wiring of orexin neurons also occurs in unperturbed conditions and could account for diurnal variations of orexin neuron activity. Data in zebrafish larvae have shown rhythmic changes in the density of inhibitory innervation of orexin dendrites in relation to vigilance states. Recent findings in mice have indicated a diurnal reorganization of the excitatory/inhibitory balance in the perisomatic innervation of orexin neurons. Taken together these sets of data point to “chronoconnectivity,” i.e., a synaptic rearrangement of inputs to orexin neurons over the course of the day in relation to sleep and wake states. This opens questions on the underlying circadian and homeostatic regulation and on the involved players at synaptic level, which could implicate dual transmitters, cytoskeletal rearrangements, hormonal regulation, as well as surrounding glial cells and extracellular matrix. Furthermore, the question arises of a “chronoconnectivity” in the wiring of other neuronal cell groups of the sleep-wake-regulatory network, many of which are characterized by variations of their firing rate during vigilance states.
Collapse
Affiliation(s)
- Idris A Azeez
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Del Gallo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience, Verona Unit, Verona, Italy
| |
Collapse
|
38
|
Roccaro-Waldmeyer DM, Girard F, Milani D, Vannoni E, Prétôt L, Wolfer DP, Celio MR. Eliminating the VGlut2-Dependent Glutamatergic Transmission of Parvalbumin-Expressing Neurons Leads to Deficits in Locomotion and Vocalization, Decreased Pain Sensitivity, and Increased Dominance. Front Behav Neurosci 2018; 12:146. [PMID: 30072881 PMCID: PMC6058961 DOI: 10.3389/fnbeh.2018.00146] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/26/2018] [Indexed: 11/13/2022] Open
Abstract
The calcium-binding protein parvalbumin (PV) is a recognized marker of short-axon GABA-ergic neurons in the cortex and the hippocampus. However in addition, PV is expressed by excitatory, glutamatergic neurons in various areas of the brain and spinal cord. Depending on the location of these neurons, loading of their synaptic vesicles with glutamate is mediated by either of three vesicular glutamate transporters (VGlut): VGlut1, VGlut2, or VGlut3. Driven by our interest in one of these glutamatergic/PV-expressing cell clusters-the lateral hypothalamic parvafox nucleus-we investigated the functions of this population of neurons by the selective deletion of VGlut2 expression in PV-expressing cells according to the Cre/Lox-approach. PV-Cre;VGlut2-Lox mutant mice are phenotypically characterized by deficits in locomotion and vocalization, by a decreased thermal nociception, and by an increased social dominance. We conducted a search of the Allen Brain Atlas for regions that might co-express the genes encoding PV and VGlut2, and that might thus contribute to the manifestation of the observed phenotypes. Our survey revealed several structures that could contribute to the deficits in locomotion and vocalization, such as the red, the subthalamic and the deep cerebellar nuclei. It also disclosed that a shift in the balance of afferental glutamatergic neurotransmission to the periaqueductal gray matter might be accountable for the decrease in sensitivity to pain and for the increase in social dominance. As a whole, this study broadens the state of knowledge about PV-expressing excitatory neurons.
Collapse
Affiliation(s)
- Diana M Roccaro-Waldmeyer
- Anatomy and Programme in Neuroscience, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Franck Girard
- Anatomy and Programme in Neuroscience, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Daniele Milani
- Anatomy and Programme in Neuroscience, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Elisabetta Vannoni
- Division of Functional Neuroanatomy, Institute of Anatomy, Department of Medicine, University of Zurich, Zurich, Switzerland
| | - Laurent Prétôt
- Anatomy and Programme in Neuroscience, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - David P Wolfer
- Division of Functional Neuroanatomy, Institute of Anatomy, Department of Medicine, University of Zurich, Zurich, Switzerland.,Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Marco R Celio
- Anatomy and Programme in Neuroscience, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
39
|
Rau AR, Hughes AR, Hentges ST. Various transgenic mouse lines to study proopiomelanocortin cells in the brain stem label disparate populations of GABAergic and glutamatergic neurons. Am J Physiol Regul Integr Comp Physiol 2018; 315:R144-R152. [PMID: 29590552 PMCID: PMC6087889 DOI: 10.1152/ajpregu.00047.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 12/31/2022]
Abstract
Products of the proopiomelanocortin (POMC) prohormone regulate aspects of analgesia, reward, and energy balance; thus, the neurons that produce POMC in the hypothalamus have received considerable attention. However, there are also cells in the nucleus of the solitary tract (NTS) that transcribe Pomc, although low levels of Pomc mRNA and relative lack of POMC peptide products in the adult mouse NTS have hindered the study of these cells. Therefore, studies of NTS POMC cells have largely relied on transgenic mouse lines. Here, we set out to determine the amino acid (AA) transmitter phenotype of NTS POMC neurons by using Pomc-Gfp transgenic mice to identify POMC cells. We found that cells expressing the green fluorescent protein (GFP) represent a mix of GABAergic and glutamatergic cells as indicated by Gad2 and vesicular Glut2 ( vGlut2) mRNA expression, respectively. We then examined the AA phenotype of POMC cells labeled by a Pomc-Cre transgene and found that these are also a mix of GABAergic and glutamatergic cells. However, the NTS cells labeled by the Gfp- and Cre-containing transgenes represented distinct populations of cells in three different Pomc-Cre mouse lines. Consistent with previous work, we were unable to reliably detect Pomc mRNA in the NTS despite clear expression in the hypothalamus. Thus, it was not possible to determine which transgenic tool most accurately identifies NTS cells that may express Pomc or release POMC peptides, although the results indicate the transgenic tools for study of these NTS neurons can label disparate populations of cells with varied AA phenotypes.
Collapse
Affiliation(s)
- Andrew R Rau
- Department of Biomedical Sciences, Colorado State University , Fort Collins, Colorado
| | - Alexander R Hughes
- Department of Biomedical Sciences, Colorado State University , Fort Collins, Colorado
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University , Fort Collins, Colorado
| |
Collapse
|
40
|
Soteros BM, Cong Q, Palmer CR, Sia GM. Sociability and synapse subtype-specific defects in mice lacking SRPX2, a language-associated gene. PLoS One 2018; 13:e0199399. [PMID: 29920554 PMCID: PMC6007900 DOI: 10.1371/journal.pone.0199399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/06/2018] [Indexed: 01/05/2023] Open
Abstract
The FoxP2 transcription factor and its target genes have been implicated in developmental brain diseases with a prominent language component, such as developmental verbal dyspraxia and specific language impairment. How FoxP2 affects neural circuitry development remains poorly understood. The sushi domain protein SRPX2 is a target of FoxP2, and mutations in SRPX2 are associated with language defects in humans. We have previously shown that SRPX2 is a synaptogenic protein that increases excitatory synapse density. Here we provide the first characterization of mice lacking the SRPX2 gene, and show that these mice exhibit defects in both neural circuitry and communication and social behaviors. Specifically, we show that mice lacking SRPX2 show a specific reduction in excitatory VGlut2 synapses in the cerebral cortex, while VGlut1 and inhibitory synapses were largely unaffected. SRPX2 KO mice also exhibit an abnormal ultrasonic vocalization ontogenetic profile in neonatal pups, and reduced preference for social novelty. These data demonstrate a functional role for SRPX2 during brain development, and further implicate FoxP2 and its targets in regulating the development of vocalization and social circuits.
Collapse
Affiliation(s)
- Breeanne M. Soteros
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Qifei Cong
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Christian R. Palmer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Gek-Ming Sia
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- * E-mail:
| |
Collapse
|
41
|
Ogundele OM, Pardo J, Francis J, Goya RG, Lee CC. A Putative Mechanism of Age-Related Synaptic Dysfunction Based on the Impact of IGF-1 Receptor Signaling on Synaptic CaMKIIα Phosphorylation. Front Neuroanat 2018; 12:35. [PMID: 29867375 PMCID: PMC5960681 DOI: 10.3389/fnana.2018.00035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 04/18/2018] [Indexed: 01/13/2023] Open
Abstract
Insulin-like growth factor 1 receptor (IGF-1R) signaling regulates the activity and phosphorylation of downstream kinases linked to inflammation, neurodevelopment, aging and synaptic function. In addition to the control of Ca2+ currents, IGF-1R signaling modulates the activity of calcium-calmodulin-dependent kinase 2 alpha (CaMKIIα) and mitogen activated protein kinase (MAPK/ErK) through multiple signaling pathways. These proteins (CaMKIIα and MAPK) regulate Ca2+ movement and long-term potentiation (LTP). Since IGF-1R controls the synaptic activity of Ca2+, CaMKIIα and MAPK signaling, the possible mechanism through which an age-dependent change in IGF-1R can alter the synaptic expression and phosphorylation of these proteins in aging needs to be investigated. In this study, we evaluated the relationship between an age-dependent change in brain IGF-1R and phosphorylation of CaMKIIα/MAPK. Furthermore, we elucidated possible mechanisms through which dysregulated CaMKIIα/MAPK interaction may be linked to a change in neurotransmitter processing and synaptic function. Male C57BL/6 VGAT-Venus mice at postnatal days 80 (P80), 365 and 730 were used to study age-related neural changes in two brain regions associated with cognitive function: hippocampus and prefrontal cortex (PFC). By means of high throughput confocal imaging and quantitative immunoblotting, we evaluated the distribution and expression of IGF-1, IGF-1R, CaMKIIα, p-CaMKIIα, MAPK and p-MAPK in whole brain lysate, hippocampus and cortex. Furthermore, we compared protein expression patterns and regional changes at P80, P365 and P730. Ultimately, we determined the relative phosphorylation pattern of CaMKIIα and MAPK through quantification of neural p-CaMKIIα and p-MAPK/ErK, and IGF-1R expression for P80, P365 and P730 brain samples. In addition to a change in synaptic function, our results show a decrease in neural IGF-1/IGF-1R expression in whole brain, hippocampus and cortex of aged mice. This was associated with a significant upregulation of phosphorylated neural MAPK (p-MAPK) and decrease in total brain CaMKIIα (i.e., CaMKIIα and p-CaMKIIα) in the aged brain. Taken together, we showed that brain aging is associated with a change in neural IGF-1/IGF-1R expression and may be linked to a change in phosphorylation of synaptic kinases (CaMKIIα and MAPK) that are involved in the modulation of LTP.
Collapse
Affiliation(s)
- Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Joaquin Pardo
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Rodolfo G. Goya
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
42
|
Luo YJ, Li YD, Wang L, Yang SR, Yuan XS, Wang J, Cherasse Y, Lazarus M, Chen JF, Qu WM, Huang ZL. Nucleus accumbens controls wakefulness by a subpopulation of neurons expressing dopamine D 1 receptors. Nat Commun 2018; 9:1576. [PMID: 29679009 PMCID: PMC5910424 DOI: 10.1038/s41467-018-03889-3] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Nucleus accumbens (NAc) is involved in behaviors that depend on heightened wakefulness, but its impact on arousal remains unclear. Here, we demonstrate that NAc dopamine D1 receptor (D1R)-expressing neurons are essential for behavioral arousal. Using in vivo fiber photometry in mice, we find arousal-dependent increases in population activity of NAc D1R neurons. Optogenetic activation of NAc D1R neurons induces immediate transitions from non-rapid eye movement sleep to wakefulness, and chemogenetic stimulation prolongs arousal, with decreased food intake. Patch-clamp, tracing, immunohistochemistry, and electron microscopy reveal that NAc D1R neurons project to the midbrain and lateral hypothalamus, and might disinhibit midbrain dopamine neurons and lateral hypothalamus orexin neurons. Photoactivation of terminals in the midbrain and lateral hypothalamus is sufficient to induce wakefulness. Silencing of NAc D1R neurons suppresses arousal, with increased nest-building behaviors. Collectively, our data indicate that NAc D1R neuron circuits are essential for the induction and maintenance of wakefulness. The nucleus accumbens regulates many behaviours that depend on arousal. Here the authors show that dopamine D1 receptor neurons in the nucleus accumbens can directly regulate wakefulness.
Collapse
Affiliation(s)
- Yan-Jia Luo
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.,Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences,, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine,, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 201108, China
| | - Ya-Dong Li
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Lu Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.,Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences,, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine,, Fudan University, Shanghai, 200032, China
| | - Su-Rong Yang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.,Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences,, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine,, Fudan University, Shanghai, 200032, China
| | - Xiang-Shan Yuan
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Juan Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Jiang-Fan Chen
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Wei-Min Qu
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China. .,Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences,, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine,, Fudan University, Shanghai, 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences; Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China. .,Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences,, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine,, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
Preobraschenski J, Cheret C, Ganzella M, Zander JF, Richter K, Schenck S, Jahn R, Ahnert-Hilger G. Dual and Direction-Selective Mechanisms of Phosphate Transport by the Vesicular Glutamate Transporter. Cell Rep 2018; 23:535-545. [DOI: 10.1016/j.celrep.2018.03.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/13/2018] [Accepted: 03/14/2018] [Indexed: 11/30/2022] Open
|
44
|
Garrett L, Ung M, Heermann T, Niedermeier KM, Hölter S. Analysis of Neuropsychiatric Disease‐Related Functional Neuroanatomical Markers in Mice. ACTA ACUST UNITED AC 2018; 8:79-128. [DOI: 10.1002/cpmo.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lillian Garrett
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
- German Mouse Clinic, Helmholtz Zentrum München; German Research Centre for Environmental Health Neuherberg Germany
| | - Marie‐Claire Ung
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Tamara Heermann
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Kristina M. Niedermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Sabine Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
- German Mouse Clinic, Helmholtz Zentrum München; German Research Centre for Environmental Health Neuherberg Germany
| |
Collapse
|
45
|
Johnson CS, Bains JS, Watts AG. Neurotransmitter diversity in pre-synaptic terminals located in the parvicellular neuroendocrine paraventricular nucleus of the rat and mouse hypothalamus. J Comp Neurol 2018; 526:1287-1306. [PMID: 29424419 DOI: 10.1002/cne.24407] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 02/02/2023]
Abstract
Virtually all rodent neuroendocrine corticotropin-releasing-hormone (CRH) neurons are in the dorsal medial parvicellular (mpd) part of the paraventricular nucleus of the hypothalamus (PVH). They form the final common pathway for adrenocortical stress responses. Their activity is controlled by sets of GABA-, glutamate-, and catecholamine-containing inputs arranged in an interactive pre-motor network. Defining the nature and arrangement of these inputs can help clarify how stressor type and intensity information is conveyed to neuroendocrine neurons. Here we use immunohistochemistry with high-resolution 3-dimensional image analyses to examine the arrangement of single- and co-occurring GABA, glutamate, and catecholamine markers in synaptophysin-defined pre-synaptic terminals in the PVHmpd of unstressed rats and Crh-IRES-Cre;Ai14 transgenic mice: respectively, vesicular glutamate transporter 2 (VGluT2), vesicular GABA transporter (VGAT), dopamine β-hydroxylase (DBH), and phenylethanolamine n-methyltransferase (PNMT). Just over half of all PVHmpd pre-synaptic terminals contain VGAT, with slightly less containing VGluT2. The vast majority of terminal appositions with mouse CRH neurons occur non-somatically. However, there are significantly more somatic VGAT than VGluT2 appositions. In the rat PVHmpd, about five times as many pre-synaptic terminals contain PNMT than DBH only. However, because epinephrine release has never been detected in the PVH, PNMT terminals may functionally be noradrenergic not adrenergic. PNMT and VGluT2 co-occur in some pre-synaptic terminals indicating the potential for co-transmission of glutamate and norepinephrine. Collectively, these results provide a structural basis for how GABA/glutamate/catecholamine interactions enable adrenocortical responses to fast-onset interosensory stimuli, and more broadly, how combinations of PVH neurotransmitters and neuromodulators interact dynamically to control adrenocortical activity.
Collapse
Affiliation(s)
- Caroline S Johnson
- The Department of Biological Sciences, USC Dornsife College of Letters, Arts, and Sciences, and Neuroscience, Graduate Program, University of Southern California, Los Angeles, California
| | - Jaideep S Bains
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Alberta, Canada
| | - Alan G Watts
- The Department of Biological Sciences, USC Dornsife College of Letters, Arts, and Sciences, and Neuroscience, Graduate Program, University of Southern California, Los Angeles, California
| |
Collapse
|
46
|
Richter K, Schmutz I, Darna M, Zander JF, Chavan R, Albrecht U, Ahnert-Hilger G. VGLUT1 Binding to Endophilin or Intersectin1 and Dynamin Phosphorylation in a Diurnal Context. Neuroscience 2018; 371:29-37. [DOI: 10.1016/j.neuroscience.2017.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
|
47
|
Wagner F, Weiss T, Veh RW. Electrophysiological properties of neurons and synapses in the lateral habenular complex (LHb). Pharmacol Biochem Behav 2017; 162:38-45. [DOI: 10.1016/j.pbb.2017.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/28/2017] [Accepted: 07/18/2017] [Indexed: 11/26/2022]
|
48
|
Palazzolo G, Moroni M, Soloperto A, Aletti G, Naldi G, Vassalli M, Nieus T, Difato F. Fast wide-volume functional imaging of engineered in vitro brain tissues. Sci Rep 2017; 7:8499. [PMID: 28819205 PMCID: PMC5561227 DOI: 10.1038/s41598-017-08979-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
The need for in vitro models that mimic the human brain to replace animal testing and allow high-throughput screening has driven scientists to develop new tools that reproduce tissue-like features on a chip. Three-dimensional (3D) in vitro cultures are emerging as an unmatched platform that preserves the complexity of cell-to-cell connections within a tissue, improves cell survival, and boosts neuronal differentiation. In this context, new and flexible imaging approaches are required to monitor the functional states of 3D networks. Herein, we propose an experimental model based on 3D neuronal networks in an alginate hydrogel, a tunable wide-volume imaging approach, and an efficient denoising algorithm to resolve, down to single cell resolution, the 3D activity of hundreds of neurons expressing the calcium sensor GCaMP6s. Furthermore, we implemented a 3D co-culture system mimicking the contiguous interfaces of distinct brain tissues such as the cortical-hippocampal interface. The analysis of the network activity of single and layered neuronal co-cultures revealed cell-type-specific activities and an organization of neuronal subpopulations that changed in the two culture configurations. Overall, our experimental platform represents a simple, powerful and cost-effective platform for developing and monitoring living 3D layered brain tissue on chip structures with high resolution and high throughput.
Collapse
Affiliation(s)
- G Palazzolo
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - M Moroni
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.,Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - A Soloperto
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - G Aletti
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - G Naldi
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - M Vassalli
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - T Nieus
- Department of Biomedical and Clinical Sciences "L. Sacco", Università degli Studi di Milano, Milano, Italy.
| | - F Difato
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.
| |
Collapse
|
49
|
Fattorini G, Ciriachi C, Conti F. Few, Activity-Dependent, and Ubiquitous VGLUT1/VGAT Terminals in Rat and Mouse Brain. Front Cell Neurosci 2017; 11:229. [PMID: 28848395 PMCID: PMC5550726 DOI: 10.3389/fncel.2017.00229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/20/2017] [Indexed: 11/13/2022] Open
Abstract
In the neocortex of adult rats VGLUT1 and VGAT co-localize in axon terminals which form both symmetric and asymmetric synapses. They are expressed in the same synaptic vesicles which participate in the exo-endocytotic cycle. Virtually nothing, however, is known on whether VGLUT1/VGAT co-localization occurs in other brain regions. We therefore mapped the distribution of terminals co-expressing VGLUT1/VGAT in the striatum, hippocampus, thalamus, and cerebellar and cerebral cortices of rats and mice. Confocal microscopy analysis revealed that, in both rat and mouse brain, VGLUT1/VGAT+ terminals were present in all brain regions studied, and that their percentage was low and comparable in both species. These results provide the first demonstration that co-expression of VGLUT1 and VGAT is a widespread phenomenon. Since VGLUT1/VGAT+ axon terminals are regulated in an activity-dependent manner and co-release glutamate and GABA, we hypothesize that, though not numerous, they can contribute to regulating excitation/inhibition balance in physiological conditions, thereby playing a role in several neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle MarcheAncona, Italy.,Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani - Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy
| | - Chiara Ciriachi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle MarcheAncona, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle MarcheAncona, Italy.,Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani - Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy.,Fondazione di Medicina Molecolare, Università Politecnica delle MarcheAncona, Italy
| |
Collapse
|
50
|
Budzillo A, Duffy A, Miller KE, Fairhall AL, Perkel DJ. Dopaminergic modulation of basal ganglia output through coupled excitation-inhibition. Proc Natl Acad Sci U S A 2017; 114:5713-5718. [PMID: 28507134 PMCID: PMC5465888 DOI: 10.1073/pnas.1611146114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Learning and maintenance of skilled movements require exploration of motor space and selection of appropriate actions. Vocal learning and social context-dependent plasticity in songbirds depend on a basal ganglia circuit, which actively generates vocal variability. Dopamine in the basal ganglia reduces trial-to-trial neural variability when the bird engages in courtship song. Here, we present evidence for a unique, tonically active, excitatory interneuron in the songbird basal ganglia that makes strong synaptic connections onto output pallidal neurons, often linked in time with inhibitory events. Dopamine receptor activity modulates the coupling of these excitatory and inhibitory events in vitro, which results in a dynamic change in the synchrony of a modeled population of basal ganglia output neurons receiving excitatory and inhibitory inputs. The excitatory interneuron thus serves as one biophysical mechanism for the introduction or modulation of neural variability in this circuit.
Collapse
Affiliation(s)
- Agata Budzillo
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA 98195
| | - Alison Duffy
- Department of Physics, University of Washington, Seattle, WA 98195
| | - Kimberly E Miller
- Department of Otolaryngology, University of Washington, Seattle, WA 98195
| | - Adrienne L Fairhall
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
- University of Washington Institute for Neuroengineering, University of Washington, Seattle, WA 98195
- Center for Sensorimotor Neural Engineering, University of Washington, Seattle, WA 98195
| | - David J Perkel
- Department of Otolaryngology, University of Washington, Seattle, WA 98195;
- University of Washington Institute for Neuroengineering, University of Washington, Seattle, WA 98195
- Department of Biology, University of Washington, Seattle, WA 98195
| |
Collapse
|