1
|
Sun W, Wu H, Peng Y, Zheng X, Li J, Zeng D, Tang P, Zhao M, Feng H, Li H, Liang Y, Su J, Chen X, Hökfelt T, He J. Heterosynaptic plasticity of the visuo-auditory projection requires cholecystokinin released from entorhinal cortex afferents. eLife 2024; 13:e83356. [PMID: 38436304 PMCID: PMC10954309 DOI: 10.7554/elife.83356] [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: 09/09/2022] [Accepted: 03/03/2024] [Indexed: 03/05/2024] Open
Abstract
The entorhinal cortex is involved in establishing enduring visuo-auditory associative memory in the neocortex. Here we explored the mechanisms underlying this synaptic plasticity related to projections from the visual and entorhinal cortices to the auditory cortex in mice using optogenetics of dual pathways. High-frequency laser stimulation (HFS laser) of the visuo-auditory projection did not induce long-term potentiation. However, after pairing with sound stimulus, the visuo-auditory inputs were potentiated following either infusion of cholecystokinin (CCK) or HFS laser of the entorhino-auditory CCK-expressing projection. Combining retrograde tracing and RNAscope in situ hybridization, we show that Cck expression is higher in entorhinal cortex neurons projecting to the auditory cortex than in those originating from the visual cortex. In the presence of CCK, potentiation in the neocortex occurred when the presynaptic input arrived 200 ms before postsynaptic firing, even after just five trials of pairing. Behaviorally, inactivation of the CCK+ projection from the entorhinal cortex to the auditory cortex blocked the formation of visuo-auditory associative memory. Our results indicate that neocortical visuo-auditory association is formed through heterosynaptic plasticity, which depends on release of CCK in the neocortex mostly from entorhinal afferents.
Collapse
Affiliation(s)
- Wenjian Sun
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Haohao Wu
- Department of Neuroscience, Karolinska InstitutetStockholmSweden
| | - Yujie Peng
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Xuejiao Zheng
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Jing Li
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Dingxuan Zeng
- Department of Neuroscience, City University of Hong KongHong KongChina
| | - Peng Tang
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Ming Zhao
- Department of Neuroscience, Karolinska InstitutetStockholmSweden
| | - Hemin Feng
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Hao Li
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Ye Liang
- Department of Neuroscience, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Junfeng Su
- Department of Neuroscience, City University of Hong KongHong KongChina
| | - Xi Chen
- Department of Neuroscience, City University of Hong KongHong KongChina
- City University of Hong Kong Shenzhen Research InstituteShenzhenChina
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska InstitutetStockholmSweden
- Institute of Advanced Study, City University of Hong KongHong KongChina
| | - Jufang He
- Department of Neuroscience, City University of Hong KongHong KongChina
- City University of Hong Kong Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
2
|
Ogyu K, Matsushita K, Honda S, Wada M, Tamura S, Takenouchi K, Tobari Y, Kusudo K, Kato H, Koizumi T, Arai N, Koreki A, Matsui M, Uchida H, Fujii S, Onaya M, Hirano Y, Mimura M, Nakajima S, Noda Y. Decrease in gamma-band auditory steady-state response in patients with treatment-resistant schizophrenia. Schizophr Res 2023; 252:129-137. [PMID: 36641960 DOI: 10.1016/j.schres.2023.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/26/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND Thirty percent of patients with schizophrenia do not respond to non-clozapine antipsychotics and are termed treatment-resistant schizophrenia (TRS). The 40-Hz auditory steady-state response (ASSR) is a well-known to be reduced in patients with schizophrenia compared to healthy controls (HCs), suggesting impaired gamma oscillation in schizophrenia. Given no ASSR study on TRS, we aimed to examine the neurophysiological basis of TRS employing 40-Hz ASSR paradigm. METHOD We compared ASSR measures among HCs, patients with non-TRS, and patients with TRS. TRS criteria were defined by a score of 4 or higher on two items of the Positive and Negative Syndrome Scale (PANSS) positive symptoms despite standard antipsychotic treatment. Participants were examined for ASSR with 40-Hz click-train stimulus, and then time-frequency analysis was performed to calculate evoked power and phase-locking factor (PLF) of 40-Hz ASSR. RESULTS A total of 79 participants were included: 27 patients with TRS (PANSS = 92.6 ± 15.8); 27 patients with non-TRS (PANSS = 63.3 ± 14.7); and 25 HCs. Evoked power in 40-Hz ASSR was lower in the TRS group than in the HC group (F2,79 = 8.37, p = 0.015; TRS vs. HCs: p = 0.012, d = 1.1) while no differences in PLF were found between the groups. CONCLUSION These results suggest that glutamatergic and GABAergic neurophysiological dysfunctions are involved in the pathophysiology of TRS. Our findings warrant more comprehensive and longitudinal studies for deep phenotyping of TRS.
Collapse
Affiliation(s)
- Kamiyu Ogyu
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba 266-0007, Japan
| | - Karin Matsushita
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masataka Wada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shunsuke Tamura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazumasa Takenouchi
- Department of Clinical Laboratory Medicine, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba 266-0007, Japan
| | - Yui Tobari
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan; Faculty of Environment and Information Studies, Keio University, Kanagawa, Kanagawa 252-0882, Japan
| | - Keisuke Kusudo
- Department of Psychiatry, National Hospital Organization Chiba Medical Center, Chiba 260-8606, Japan
| | - Hideo Kato
- Department of Epileptology, National Center of Neurology and Psychiatry Hospital, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Teruki Koizumi
- Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba 266-0007, Japan
| | - Naohiro Arai
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Akihiro Koreki
- Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba 266-0007, Japan
| | - Mie Matsui
- Department of Clinical Cognitive Neuroscience, Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 920-1164, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinya Fujii
- Faculty of Environment and Information Studies, Keio University, Kanagawa, Kanagawa 252-0882, Japan
| | - Mitsumoto Onaya
- Department of Psychiatry, National Hospital Organization Shimofusa Psychiatric Medical Center, Chiba 266-0007, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan; Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada.
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
3
|
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
|
4
|
Fish KN, Joffe ME. Targeting prefrontal cortex GABAergic microcircuits for the treatment of alcohol use disorder. Front Synaptic Neurosci 2022; 14:936911. [PMID: 36105666 PMCID: PMC9465392 DOI: 10.3389/fnsyn.2022.936911] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Developing novel treatments for alcohol use disorders (AUDs) is of paramount importance for improving patient outcomes and alleviating the suffering related to the disease. A better understanding of the molecular and neurocircuit mechanisms through which alcohol alters brain function will be instrumental in the rational development of new efficacious treatments. Clinical studies have consistently associated the prefrontal cortex (PFC) function with symptoms of AUDs. Population-level analyses have linked the PFC structure and function with heavy drinking and/or AUD diagnosis. Thus, targeting specific PFC cell types and neural circuits holds promise for the development of new treatments. Here, we overview the tremendous diversity in the form and function of inhibitory neuron subtypes within PFC and describe their therapeutic potential. We then summarize AUD population genetics studies, clinical neurophysiology findings, and translational neuroscience discoveries. This study collectively suggests that changes in fast transmission through PFC inhibitory microcircuits are a central component of the neurobiological effects of ethanol and the core symptoms of AUDs. Finally, we submit that there is a significant and timely need to examine sex as a biological variable and human postmortem brain tissue to maximize the efforts in translating findings to new clinical treatments.
Collapse
Affiliation(s)
| | - Max E. Joffe
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
5
|
Eiden LE, Hernández VS, Jiang SZ, Zhang L. Neuropeptides and small-molecule amine transmitters: cooperative signaling in the nervous system. Cell Mol Life Sci 2022; 79:492. [PMID: 35997826 PMCID: PMC11072502 DOI: 10.1007/s00018-022-04451-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
Neuropeptides are expressed in cell-specific patterns throughout mammalian brain. Neuropeptide gene expression has been useful for clustering neurons by phenotype, based on single-cell transcriptomics, and for defining specific functional circuits throughout the brain. How neuropeptides function as first messengers in inter-neuronal communication, in cooperation with classical small-molecule amine transmitters (SMATs) is a current topic of systems neurobiology. Questions include how neuropeptides and SMATs cooperate in neurotransmission at the molecular, cellular and circuit levels; whether neuropeptides and SMATs always co-exist in neurons; where neuropeptides and SMATs are stored in the neuron, released from the neuron and acting, and at which receptors, after release; and how neuropeptides affect 'classical' transmitter function, both directly upon co-release, and indirectly, via long-term regulation of gene transcription and neuronal plasticity. Here, we review an extensive body of data about the distribution of neuropeptides and their receptors, their actions after neuronal release, and their function based on pharmacological and genetic loss- and gain-of-function experiments, that addresses these questions, fundamental to understanding brain function, and development of neuropeptide-based, and potentially combinatorial peptide/SMAT-based, neurotherapeutics.
Collapse
Affiliation(s)
- Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA.
| | - Vito S Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Sunny Z Jiang
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
6
|
Abstract
The neuropeptide system encompasses the most diverse family of neurotransmitters, but their expression, cellular localization, and functional role in the human brain have received limited attention. Here, we study human postmortem samples from prefrontal cortex (PFC), a key brain region, and employ RNA sequencing and RNAscope methods integrated with published single-cell data. Our aim is to characterize the distribution of peptides and their receptors in 17 PFC subregions and to explore their role in chemical signaling. The results suggest that the well-established anatomical and functional heterogeneity of human PFC is also reflected in the expression pattern of the neuropeptides. Our findings support ongoing efforts from academia and pharmaceutical companies to explore the potential of neuropeptide receptors as targets for drug development. Human prefrontal cortex (hPFC) is a complex brain region involved in cognitive and emotional processes and several psychiatric disorders. Here, we present an overview of the distribution of the peptidergic systems in 17 subregions of hPFC and three reference cortices obtained by microdissection and based on RNA sequencing and RNAscope methods integrated with published single-cell transcriptomics data. We detected expression of 60 neuropeptides and 60 neuropeptide receptors in at least one of the hPFC subregions. The results reveal that the peptidergic landscape in PFC consists of closely located and functionally different subregions with unique peptide/transmitter–related profiles. Neuropeptide-rich PFC subregions were identified, encompassing regions from anterior cingulate cortex/orbitofrontal gyrus. Furthermore, marked differences in gene expression exist between different PFC regions (>5-fold; cocaine and amphetamine–regulated transcript peptide) as well as between PFC regions and reference regions, for example, for somatostatin and several receptors. We suggest that the present approach allows definition of, still hypothetical, microcircuits exemplified by glutamatergic neurons expressing a peptide cotransmitter either as an agonist (hypocretin/orexin) or antagonist (galanin). Specific neuropeptide receptors have been identified as possible targets for neuronal afferents and, interestingly, peripheral blood-borne peptide hormones (leptin, adiponectin, gastric inhibitory peptide, glucagon-like peptides, and peptide YY). Together with other recent publications, our results support the view that neuropeptide systems may play an important role in hPFC and underpin the concept that neuropeptide signaling helps stabilize circuit connectivity and fine-tune/modulate PFC functions executed during health and disease.
Collapse
|
7
|
Casello SM, Flores RJ, Yarur HE, Wang H, Awanyai M, Arenivar MA, Jaime-Lara RB, Bravo-Rivera H, Tejeda HA. Neuropeptide System Regulation of Prefrontal Cortex Circuitry: Implications for Neuropsychiatric Disorders. Front Neural Circuits 2022; 16:796443. [PMID: 35800635 PMCID: PMC9255232 DOI: 10.3389/fncir.2022.796443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
Neuropeptides, a diverse class of signaling molecules in the nervous system, modulate various biological effects including membrane excitability, synaptic transmission and synaptogenesis, gene expression, and glial cell architecture and function. To date, most of what is known about neuropeptide action is limited to subcortical brain structures and tissue outside of the central nervous system. Thus, there is a knowledge gap in our understanding of neuropeptide function within cortical circuits. In this review, we provide a comprehensive overview of various families of neuropeptides and their cognate receptors that are expressed in the prefrontal cortex (PFC). Specifically, we highlight dynorphin, enkephalin, corticotropin-releasing factor, cholecystokinin, somatostatin, neuropeptide Y, and vasoactive intestinal peptide. Further, we review the implication of neuropeptide signaling in prefrontal cortical circuit function and use as potential therapeutic targets. Together, this review summarizes established knowledge and highlights unknowns of neuropeptide modulation of neural function underlying various biological effects while offering insights for future research. An increased emphasis in this area of study is necessary to elucidate basic principles of the diverse signaling molecules used in cortical circuits beyond fast excitatory and inhibitory transmitters as well as consider components of neuropeptide action in the PFC as a potential therapeutic target for neurological disorders. Therefore, this review not only sheds light on the importance of cortical neuropeptide studies, but also provides a comprehensive overview of neuropeptide action in the PFC to serve as a roadmap for future studies in this field.
Collapse
Affiliation(s)
- Sanne M. Casello
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rodolfo J. Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Monique Awanyai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Miguel A. Arenivar
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rosario B. Jaime-Lara
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Hector Bravo-Rivera
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Hugo A. Tejeda,
| |
Collapse
|
8
|
Somatostatin and Somatostatin-Containing Interneurons—From Plasticity to Pathology. Biomolecules 2022; 12:biom12020312. [PMID: 35204812 PMCID: PMC8869243 DOI: 10.3390/biom12020312] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/27/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the obvious differences in the pathophysiology of distinct neuropsychiatric diseases or neurodegenerative disorders, some of them share some general but pivotal mechanisms, one of which is the disruption of excitation/inhibition balance. Such an imbalance can be generated by changes in the inhibitory system, very often mediated by somatostatin-containing interneurons (SOM-INs). In physiology, this group of inhibitory interneurons, as well as somatostatin itself, profoundly shapes the brain activity, thus influencing the behavior and plasticity; however, the changes in the number, density and activity of SOM-INs or levels of somatostatin are found throughout many neuropsychiatric and neurological conditions, both in patients and animal models. Here, we (1) briefly describe the brain somatostatinergic system, characterizing the neuropeptide somatostatin itself, its receptors and functions, as well the physiology and circuitry of SOM-INs; and (2) summarize the effects of the activity of somatostatin and SOM-INs in both physiological brain processes and pathological brain conditions, focusing primarily on learning-induced plasticity and encompassing selected neuropsychological and neurodegenerative disorders, respectively. The presented data indicate the somatostatinergic-system-mediated inhibition as a substantial factor in the mechanisms of neuroplasticity, often disrupted in a plethora of brain pathologies.
Collapse
|
9
|
Kajita Y, Mushiake H. Heterogeneous GAD65 Expression in Subtypes of GABAergic Neurons Across Layers of the Cerebral Cortex and Hippocampus. Front Behav Neurosci 2021; 15:750869. [PMID: 34803625 PMCID: PMC8595203 DOI: 10.3389/fnbeh.2021.750869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Gamma-aminobutyric acid (GABA), a major inhibitory transmitter in the central nervous system, is synthesized via either of two enzyme isoforms, GAD65 or GAD67. GAD65 is synthesized in the soma but functions at synaptic terminals in an activity-dependent manner, playing a distinct role in excitatory-inhibitory balance. However, the extent to which each GABAergic subtype expresses GAD65 in the resting state remains unclear. In this study, we compared GAD65 expression among six GABAergic subtypes: NPY+, nNOS+, PV+, SOM+, CR+, and CCK+. According to the results, the GABAergic subtypes were classified into two groups per region based on GAD65 expression levels: high-expression (NPY+ and nNOS+) and low-expression groups (PV+, SOM+, CR+, and CCK+) in the cerebral cortex and high-expression (NPY+, nNOS+, and CCK+) and low-expression groups (PV+, SOM+, and CR+) in the hippocampus. Moreover, these expression patterns revealed a distinct laminar distribution in the cerebral cortex and hippocampus. To investigate the extent of GAD65 transport from the soma to synaptic terminals, we examined GAD65 expression in colchicine-treated rats in which GAD65 was synthesized in the soma but not transported to terminals. We found a significant positive correlation in GAD65 expression across subtypes between colchicine-treated and control rats. In summary, each GABAergic subtype exhibits a distinct GAD65 expression pattern across layers of the cerebral cortex and hippocampus. In addition, the level of GAD65 expression in the soma can be used as a proxy for the amount of GAD65 in the cytoplasm. These findings suggest that exploration of the distinct profiles of GAD65 expression among GABAergic subtypes could clarify the roles that GABAergic subtypes play in maintaining the excitatory-inhibitory balance.
Collapse
Affiliation(s)
- Yuki Kajita
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hajime Mushiake
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
10
|
Patthy Á, Murai J, Hanics J, Pintér A, Zahola P, Hökfelt TGM, Harkany T, Alpár A. Neuropathology of the Brainstem to Mechanistically Understand and to Treat Alzheimer's Disease. J Clin Med 2021; 10:jcm10081555. [PMID: 33917176 PMCID: PMC8067882 DOI: 10.3390/jcm10081555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being “en passant”, can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Murai
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Hanics
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Péter Zahola
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
11
|
Lee MJ, Lee WT, Jeon CJ. Organization of Neuropeptide Y-Immunoreactive Cells in the Mongolian gerbil ( Meriones unguiculatus) Visual Cortex. Cells 2021; 10:cells10020311. [PMID: 33546356 PMCID: PMC7913502 DOI: 10.3390/cells10020311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/03/2023] Open
Abstract
Neuropeptide Y (NPY) is found throughout the central nervous system where it appears to be involved in the regulation of a wide range of physiological effects. The Mongolian gerbil, a member of the rodent family Muridae, is a diurnal animal and has been widely used in various aspects of biomedical research. This study was conducted to investigate the organization of NPY-immunoreactive (IR) neurons in the gerbil visual cortex using NPY immunocytochemistry. The highest density of NPY-IR neurons was located in layer V (50.58%). The major type of NPY-IR neuron was a multipolar round/oval cell type (44.57%). Double-color immunofluorescence revealed that 89.55% and 89.95% of NPY-IR neurons contained gamma-aminobutyric acid (GABA) or somatostatin, respectively. Several processes of the NPY-IR neurons surrounded GABAergic interneurons. Although 30.81% of the NPY-IR neurons contained calretinin, NPY and calbindin-D28K-IR neurons were co-expressed rarely (3.75%) and NPY did not co-express parvalbumin. Triple-color immunofluorescence with anti-GluR2 or CaMKII antibodies suggested that some non-GABAergic NPY-IR neurons may make excitatory synaptic contacts. This study indicates that NPY-IR neurons have a notable architecture and are unique subpopulations of the interneurons of the gerbil visual cortex, which could provide additional valuable data for elucidating the role of NPY in the visual process in diurnal animals.
Collapse
|
12
|
Brockway DF, Crowley NA. Turning the 'Tides on Neuropsychiatric Diseases: The Role of Peptides in the Prefrontal Cortex. Front Behav Neurosci 2020; 14:588400. [PMID: 33192369 PMCID: PMC7606924 DOI: 10.3389/fnbeh.2020.588400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Recent advancements in technology have enabled researchers to probe the brain with the greater region, cell, and receptor specificity. These developments have allowed for a more thorough understanding of how regulation of the neurophysiology within a region is essential for maintaining healthy brain function. Stress has been shown to alter the prefrontal cortex (PFC) functioning, and evidence links functional impairments in PFC brain activity with neuropsychiatric disorders. Moreover, a growing body of literature highlights the importance of neuropeptides in the PFC to modulate neural signaling and to influence behavior. The converging evidence outlined in this review indicates that neuropeptides in the PFC are specifically impacted by stress, and are found to be dysregulated in numerous stress-related neuropsychiatric disorders including substance use disorder, major depressive disorder (MDD), posttraumatic stress disorder, and schizophrenia. This review explores how neuropeptides in the PFC function to regulate the neural activity, and how genetic and environmental factors, such as stress, lead to dysregulation in neuropeptide systems, which may ultimately contribute to the pathology of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Dakota F Brockway
- Neuroscience Curriculum, Pennsylvania State University, University Park, PA, United States
| | - Nicole A Crowley
- Neuroscience Curriculum, Pennsylvania State University, University Park, PA, United States.,The Department of Biology, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
13
|
Hou X, Rong C, Wang F, Liu X, Sun Y, Zhang HT. GABAergic System in Stress: Implications of GABAergic Neuron Subpopulations and the Gut-Vagus-Brain Pathway. Neural Plast 2020; 2020:8858415. [PMID: 32802040 PMCID: PMC7416252 DOI: 10.1155/2020/8858415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Stress can cause a variety of central nervous system disorders, which are critically mediated by the γ-aminobutyric acid (GABA) system in various brain structures. GABAergic neurons have different subsets, some of which coexpress certain neuropeptides that can be found in the digestive system. Accumulating evidence demonstrates that the gut-brain axis, which is primarily regulated by the vagus nerve, is involved in stress, suggesting a communication between the "gut-vagus-brain" pathway and the GABAergic neuronal system. Here, we first summarize the evidence that the GABAergic system plays an essential role in stress responses. In addition, we review the effects of stress on different brain regions and GABAergic neuron subpopulations, including somatostatin, parvalbumin, ionotropic serotonin receptor 5-HT3a, cholecystokinin, neuropeptide Y, and vasoactive intestinal peptide, with regard to signaling events, behavioral changes, and pathobiology of neuropsychiatric diseases. Finally, we discuss the gut-brain bidirectional communications and the connection of the GABAergic system and the gut-vagus-brain pathway.
Collapse
Affiliation(s)
- Xueqin Hou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271016, China
| | - Cuiping Rong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Fugang Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271016, China
| | - Xiaoqian Liu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271016, China
| | - Yi Sun
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong 271016, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| |
Collapse
|
14
|
Franco LO, Carvalho MJ, Costa J, Ferreira PA, Guedes JR, Sousa R, Edfawy M, Seabra CM, Cardoso AL, Peça J. Social subordination induced by early life adversity rewires inhibitory control of the prefrontal cortex via enhanced Npy1r signaling. Neuropsychopharmacology 2020; 45:1438-1447. [PMID: 32492699 PMCID: PMC7360628 DOI: 10.1038/s41386-020-0727-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/16/2020] [Accepted: 05/26/2020] [Indexed: 01/06/2023]
Abstract
Social hierarchies are present in most mammalian species. In nature, hierarchies offer a tradeoff between reduction of in-group fighting between males, at the expense of an asymmetric sharing of resources. Early life experiences and stress are known to influence the rank an individual attains in adulthood, but the associated cellular and synaptic alterations are poorly understood. Using a maternal separation protocol, we show that care-deprived mice display a long-lasting submissive phenotype, increased social recognition, and enhanced explorative behavior. These alterations are consistent with an adaptation that favors exploration rather than confrontation within a group setting. At the neuronal level, these animals display dendritic atrophy and enhanced inhibitory synaptic inputs in medial prefrontal cortex (mPFC) neurons. To determine what could underlie this synaptic modification, we first assessed global gene expression changes via RNAseq, and next focused on a smaller subset of putatively altered synaptic receptors that could explain the changes in synaptic inhibition. Using different cohorts of maternally deprived mice, we validated a significant increase in the expression of Npy1r, a receptor known to play a role in maternal care, anxiety, foraging, and regulation of group behavior. Using electrophysiological recordings in adult mice while blocking NPY1R signaling, we determined that this receptor plays a key role in enhancing GABAergic currents in mice that experience maternal deprivation. Taken together, our work highlights the potential of regulating NPY1R in social anxiety disorders and the alterations induced in brain circuitry as a consequence of early life stress and adversity.
Collapse
Affiliation(s)
- Lara O. Franco
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cPhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Mário J. Carvalho
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,MIT-Portugal Bioengineering Systems Doctoral Program, Coimbra, Portugal
| | - Jéssica Costa
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cPhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Pedro A. Ferreira
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Joana R. Guedes
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Renato Sousa
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Mohamed Edfawy
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Catarina M. Seabra
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Ana L. Cardoso
- 0000 0000 9511 4342grid.8051.cCNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - João Peça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal. .,Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
15
|
Pernia M, Díaz I, Colmenárez-Raga AC, Rivadulla C, Cudeiro J, Plaza I, Merchán MA. Cross-modal reaction of auditory and visual cortices after long-term bilateral hearing deprivation in the rat. Brain Struct Funct 2020; 225:129-148. [PMID: 31781971 PMCID: PMC6957565 DOI: 10.1007/s00429-019-01991-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 11/21/2019] [Indexed: 12/26/2022]
Abstract
Visual cortex (VC) over-activation analysed by evoked responses has been demonstrated in congenital deafness and after long-term acquired hearing loss in humans. However, permanent hearing deprivation has not yet been explored in animal models. Thus, the present study aimed to examine functional and molecular changes underlying the visual and auditory cross-modal reaction. For such purpose, we analysed cortical visual evoked potentials (VEPs) and the gene expression (RT-qPCR) of a set of markers for neuronal activation (c-Fos) and activity-dependent homeostatic compensation (Arc/Arg3.1). To determine the state of excitation and inhibition, we performed RT-qPCR and quantitative immunocytochemistry for excitatory (receptor subunits GluA2/3) and inhibitory (GABAA-α1, GABAB-R2, GAD65/67 and parvalbumin-PV) markers. VC over-activation was demonstrated by a significant increase in VEPs wave N1 and by up-regulation of the activity-dependent early genes c-Fos and Arc/Arg3.1 (thus confirming, by RT-qPCR, our previously published immunocytochemical results). GluA2 gene and protein expression were significantly increased in the auditory cortex (AC), particularly in layers 2/3 pyramidal neurons, but inhibitory markers (GAD65/67 and PV-GABA interneurons) were also significantly upregulated in the AC, indicating a concurrent increase in inhibition. Therefore, after permanent hearing loss in the rat, the VC is not only over-activated but also potentially balanced by homeostatic regulation, while excitatory and inhibitory markers remain imbalanced in the AC, most likely resulting from changes in horizontal intermodal regulation.
Collapse
Affiliation(s)
- M Pernia
- Instituto de Neurociencias of Castilla y León-INCyL, Universidad de Salamanca, Salamanca, Spain
| | - I Díaz
- Instituto de Neurociencias of Castilla y León-INCyL, Universidad de Salamanca, Salamanca, Spain
| | - A C Colmenárez-Raga
- Instituto de Neurociencias of Castilla y León-INCyL, Universidad de Salamanca, Salamanca, Spain
| | - C Rivadulla
- Centro de Investigaciones Científicas Avanzadas (CICA), Facultad de Ciencias de la Salud, Universidad de A Coruña and Instituto de Investigaciones Biomédicas de A Coruña (INIBIC), A Coruña, Spain
| | - J Cudeiro
- Centro de Investigaciones Científicas Avanzadas (CICA), Facultad de Ciencias de la Salud, Universidad de A Coruña and Instituto de Investigaciones Biomédicas de A Coruña (INIBIC), A Coruña, Spain
| | - I Plaza
- Instituto de Neurociencias of Castilla y León-INCyL, Universidad de Salamanca, Salamanca, Spain
| | - M A Merchán
- Instituto de Neurociencias of Castilla y León-INCyL, Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
16
|
Vázquez-León P, Campos-Rodríguez C, Gonzalez-Pliego C, Miranda-Páez A. Differential effects of cholecystokinin (CCK-8) microinjection into the ventrolateral and dorsolateral periaqueductal gray on anxiety models in Wistar rats. Horm Behav 2018; 106:105-111. [PMID: 30342011 DOI: 10.1016/j.yhbeh.2018.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 01/27/2023]
Abstract
Cholecystokinin (CCK) is one of the main neurohormone peptide systems in the brain, and a major anxiogenic mediator. The periaqueductal gray (PAG) is a key midbrain structure for defensive behaviors, which could include anxiety, fear, or even panic. The CCK system has wide distribution in the PAG, where the dorsolateral region (DL) participates in active defensive behavior and the ventrolateral region (VL) in passive defensive behavior. The aim of this study was to assess the effect of CCK-8 microinjection into DL-PAG or VL-PAG on anxiety-like behavior through two tests: elevated plus maze (EPM) and defensive burying behavior (DBB). CCK-8 (0.5 and 1.0 μg/0.5 μL) presently microinjected into the DL-PAG produced an anxiogenic-like effect on the EPM evidenced by decreasing the time spent/number of entries in open arms compared to vehicle group. Additionally, the latency to burying decreased and burying time increased on the DBB test. Contrarily, CCK-8 microinjected into the VL-PAG resulted in greater open-arm time and more open-arm entries compared to the vehicle-microinjected group. The results on the DBB test confirmed an anxiolytic-like response of CCK-8 into the VL-PAG. In conclusion, CCK-8 microinjected into DL-PAG produced anxiety-like behavior on EPM, and for first time reported on DBB. Contrarily, CCK-8 microinjected into the VL-PAG reduced anxiety-like behavior also for first time reported using both behavioral models EPM and DBB.
Collapse
Affiliation(s)
- Priscila Vázquez-León
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico
| | - Carolina Campos-Rodríguez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico
| | - Carlos Gonzalez-Pliego
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico
| | - Abraham Miranda-Páez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico.
| |
Collapse
|
17
|
Carron SF, Yan EB, Allitt BJ, Rajan R. Immediate and Medium-term Changes in Cortical and Hippocampal Inhibitory Neuronal Populations after Diffuse TBI. Neuroscience 2018; 388:152-170. [PMID: 30036662 DOI: 10.1016/j.neuroscience.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 01/09/2023]
Abstract
Changes in inhibition following traumatic brain injury (TBI) appear to be one of the major factors that contribute to excitation:inhibition imbalance. Neuron pathology, interneurons in particular evolves from minutes to weeks post injury and follows a complex time course. Previously, we showed that in the long-term in diffuse TBI (dTBI), there was select reduction of specific dendrite-targeting neurons in sensory cortex and hippocampus while in motor cortex there was up-regulation of specific dendrite-targeting neurons. We now investigated the time course of dTBI effects on interneurons in neocortex and hippocampus. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) neuropeptide Y (NPY), and somatostatin (SOM) at 24 h and 2 weeks post dTBI. We found time-dependent, brain area-specific changes in inhibition at 24 h and 2 weeks. At 24 h post-injury, reduction of dendrite-targeting inhibitory neurons occurred in sensory cortex and hippocampus. At 2 weeks, we found compensatory changes in the somatosensory cortex and CA2/3 of hippocampus affected at 24 h, with affected interneuronal populations returning to sham levels. However, DG of hippocampus now showed reduction of dendrite-targeting inhibitory neurons. Finally, with respect to motor cortex, there was an upregulation of dendrite-targeting interneurons in the supragranular layers at 24 h returning to normal levels by 2 weeks. Overall, our findings reconfirm that dendritic inhibition is particularly susceptible to brain trauma, but also show that there are complex brain-area-specific changes in inhibitory neuronal numbers and in compensatory changes, rather than a simple monotonic progression of changes post-dTBI.
Collapse
Affiliation(s)
- Simone F Carron
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Edwin B Yan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Benjamin J Allitt
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Ramesh Rajan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
18
|
Neuropeptide Y (NPY) as a therapeutic target for neurodegenerative diseases. Neurobiol Dis 2016; 95:210-24. [PMID: 27461050 DOI: 10.1016/j.nbd.2016.07.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/29/2016] [Accepted: 07/20/2016] [Indexed: 12/16/2022] Open
Abstract
Neuropeptide Y (NPY) and NPY receptors are widely expressed in the mammalian central nervous system. Studies in both humans and rodent models revealed that brain NPY levels are altered in some neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Machado-Joseph disease. In this review, we will focus on the roles of NPY in the pathological mechanisms of these disorders, highlighting NPY as a neuroprotective agent, as a neural stem cell proliferative agent, as an agent that increases trophic support, as a stimulator of autophagy and as an inhibitor of excitotoxicity and neuroinflammation. Moreover, the effect of NPY in some clinical manifestations commonly observed in Alzheimer's disease, Parkinson's disease, Huntington's disease and Machado-Joseph disease, such as depressive symptoms and body weight loss, are also discussed. In conclusion, this review highlights NPY system as a potential therapeutic target in neurodegenerative diseases.
Collapse
|
19
|
Carron SF, Yan EB, Alwis DS, Rajan R. Differential susceptibility of cortical and subcortical inhibitory neurons and astrocytes in the long term following diffuse traumatic brain injury. J Comp Neurol 2016; 524:3530-3560. [PMID: 27072754 DOI: 10.1002/cne.24014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 02/02/2023]
Abstract
Long-term diffuse traumatic brain injury (dTBI) causes neuronal hyperexcitation in supragranular layers in sensory cortex, likely through reduced inhibition. Other forms of TBI affect inhibitory interneurons in subcortical areas but it is unknown if this occurs in cortex, or in any brain area in dTBI. We investigated dTBI effects on inhibitory neurons and astrocytes in somatosensory and motor cortex, and hippocampus, 8 weeks post-TBI. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) and neuropeptide Y (NPY), and somatostatin (SOM) and glial fibrillary acidic protein (GFAP), a marker for astrogliosis during neurodegeneration. Despite persistent behavioral deficits in rotarod performance up to the time of brain extraction (TBI = 73.13 ± 5.23% mean ± SEM, Sham = 92.29 ± 5.56%, P < 0.01), motor cortex showed only a significant increase, in NPY neurons in supragranular layers (mean cells/mm2 ± SEM, Sham = 16 ± 0.971, TBI = 25 ± 1.51, P = 0.001). In somatosensory cortex, only CR+ neurons showed changes, being decreased in supragranular (TBI = 19 ± 1.18, Sham = 25 ± 1.10, P < 0.01) and increased in infragranular (TBI = 28 ± 1.35, Sham = 24 ± 1.07, P < 0.05) layers. Heterogeneous changes were seen in hippocampal staining: CB+ decreased in dentate gyrus (TBI = 2 ± 0.382, Sham = 4 ± 0.383, P < 0.01), PV+ increased in CA1 (TBI = 39 ± 1.26, Sham = 33 ± 1.69, P < 0.05) and CA2/3 (TBI = 26 ± 2.10, Sham = 20 ± 1.49, P < 0.05), and CR+ decreased in CA1 (TBI = 10 ± 1.02, Sham = 14 ± 1.14, P < 0.05). Astrogliosis significantly increased in corpus callosum (TBI = 6.7 ± 0.69, Sham = 2.5 ± 0.38; P = 0.007). While dTBI effects on inhibitory neurons appear region- and type-specific, a common feature in all cases of decrease was that changes occurred in dendrite targeting interneurons involved in neuronal integration. J. Comp. Neurol. 524:3530-3560, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simone F Carron
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Edwin B Yan
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Dasuni S Alwis
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Ramesh Rajan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Cheetham CEJ, Grier BD, Belluscio L. Bulk regional viral injection in neonatal mice enables structural and functional interrogation of defined neuronal populations throughout targeted brain areas. Front Neural Circuits 2015; 9:72. [PMID: 26594154 PMCID: PMC4633521 DOI: 10.3389/fncir.2015.00072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/23/2015] [Indexed: 11/15/2022] Open
Abstract
The ability to label and manipulate specific cell types is central to understanding the structure and function of neuronal circuits. Here, we have developed a simple, affordable strategy for labeling of genetically defined populations of neurons throughout a targeted brain region: Bulk Regional Viral Injection (BReVI). Our strategy involves a large volume adeno-associated virus (AAV) injection in the targeted brain region of neonatal Cre driver mice. Using the mouse olfactory bulb (OB) as a model system, we tested the ability of BReVI to broadly and selectively label tufted cells, one of the two principal neuron populations of the OB, in CCK-IRES-Cre mice. BReVI resulted in labeling of neurons throughout the injected OB, with no spatial bias toward the injection site and no evidence of damage. The specificity of BReVI labeling was strikingly similar to that seen previously using immunohistochemical staining for cholecystokinin (CCK), an established tufted cell marker. Hence, the CCK-IRES-Cre line in combination with BReVI can provide an important tool for targeting and manipulation of OB tufted cells. We also found robust Cre-dependent reporter expression within three days of BReVI, which enabled us to assess developmental changes in the number and laminar distribution of OB tufted cells during the first three postnatal weeks. Furthermore, we demonstrate that BReVI permits structural and functional imaging in vivo, and can be combined with transgenic strategies to facilitate multi-color labeling of neuronal circuit components. BReVI is broadly applicable to different Cre driver lines and can be used to regionally manipulate genetically defined populations of neurons in any accessible brain region.
Collapse
Affiliation(s)
- Claire E. J. Cheetham
- National Institute of Neurological Disorders and StrokeBethesda, MD, USA
- Department of Biological Sciences, Carnegie Mellon UniversityPittsburgh, PA, USA
| | - Bryce D. Grier
- National Institute of Neurological Disorders and StrokeBethesda, MD, USA
| | - Leonardo Belluscio
- National Institute of Neurological Disorders and StrokeBethesda, MD, USA
| |
Collapse
|
21
|
Whissell PD, Cajanding JD, Fogel N, Kim JC. Comparative density of CCK- and PV-GABA cells within the cortex and hippocampus. Front Neuroanat 2015; 9:124. [PMID: 26441554 PMCID: PMC4585045 DOI: 10.3389/fnana.2015.00124] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/31/2015] [Indexed: 11/14/2022] Open
Abstract
Cholecystokinin (CCK)- and parvalbumin (PV)-expressing neurons constitute the two major populations of perisomatic GABAergic neurons in the cortex and the hippocampus. As CCK- and PV-GABA neurons differ in an array of morphological, biochemical and electrophysiological features, it has been proposed that they form distinct inhibitory ensembles which differentially contribute to network oscillations and behavior. However, the relationship and balance between CCK- and PV-GABA neurons in the inhibitory networks of the brain is currently unclear as the distribution of these cells has never been compared on a large scale. Here, we systemically investigated the distribution of CCK- and PV-GABA cells across a wide number of discrete forebrain regions using an intersectional genetic approach. Our analysis revealed several novel trends in the distribution of these cells. While PV-GABA cells were more abundant overall, CCK-GABA cells outnumbered PV-GABA cells in several subregions of the hippocampus, medial prefrontal cortex and ventrolateral temporal cortex. Interestingly, CCK-GABA cells were relatively more abundant in secondary/association areas of the cortex (V2, S2, M2, and AudD/AudV) than they were in corresponding primary areas (V1, S1, M1, and Aud1). The reverse trend was observed for PV-GABA cells. Our findings suggest that the balance between CCK- and PV-GABA cells in a given cortical region is related to the type of processing that area performs; inhibitory networks in the secondary cortex tend to favor the inclusion of CCK-GABA cells more than networks in the primary cortex. The intersectional genetic labeling approach employed in the current study expands upon the ability to study molecularly defined subsets of GABAergic neurons. This technique can be applied to the investigation of neuropathologies which involve disruptions to the GABAergic system, including schizophrenia, stress, maternal immune activation and autism.
Collapse
Affiliation(s)
- Paul D Whissell
- Department of Psychology, University of Toronto, Toronto ON, Canada
| | | | - Nicole Fogel
- Cell and Systems Biology, University of Toronto, Toronto ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto ON, Canada ; Cell and Systems Biology, University of Toronto, Toronto ON, Canada
| |
Collapse
|
22
|
Bari A, Dec A, Lee AW, Lee J, Song D, Dale E, Peterson J, Zorn S, Huang X, Campbell B, Robbins TW, West AR. Enhanced inhibitory control by neuropeptide Y Y5 receptor blockade in rats. Psychopharmacology (Berl) 2015; 232:959-73. [PMID: 25194952 DOI: 10.1007/s00213-014-3730-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/24/2014] [Indexed: 12/25/2022]
Abstract
RATIONALE The neuropeptide Y (NPY) system acts in synergy with the classic neurotransmitters to regulate a large variety of functions including autonomic, affective, and cognitive processes. Research on the effects of NPY in the central nervous system has focused on food intake control and affective processes, but growing evidence of NPY involvement in attention-deficit/hyperactivity disorder (ADHD) and other psychiatric conditions motivated the present study. OBJECTIVES We tested the effects of the novel and highly selective NPY Y5 receptor antagonist Lu AE00654 on impulsivity and the underlying cortico-striatal circuitry in rats to further explore the possible involvement of the NPY system in pathologies characterized by inattention and impulsive behavior. RESULTS A low dose of Lu AE00654 (0.03 mg/kg) selectively facilitated response inhibition as measured by the stop-signal task, whereas no effects were found at higher doses (0.3 and 3 mg/kg). Systemic administration of Lu AE00654 also enhanced the inhibitory influence of the dorsal frontal cortex on neurons in the caudate-putamen, this fronto-striatal circuitry being implicated in the executive control of behavior. Finally, by locally injecting a Y5 agonist, we observed reciprocal activation between dorsal frontal cortex and caudate-putamen neurons. Importantly, the effects of the Y5 agonist were attenuated by pretreatment with Lu AE00654, confirming the presence of Y5 binding sites modulating functional interactions within frontal-subcortical circuits. CONCLUSIONS These results suggest that the NPY system modulates inhibitory neurotransmission in brain areas important for impulse control, and may be relevant for the treatment of pathologies such as ADHD and drug abuse.
Collapse
Affiliation(s)
- A Bari
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Plescia F, Brancato A, Marino RAM, Vita C, Navarra M, Cannizzaro C. Effect of Acetaldehyde Intoxication and Withdrawal on NPY Expression: Focus on Endocannabinoidergic System Involvement. Front Psychiatry 2014; 5:138. [PMID: 25324788 PMCID: PMC4181239 DOI: 10.3389/fpsyt.2014.00138] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/18/2014] [Indexed: 01/17/2023] Open
Abstract
Acetaldehyde (ACD), the first alcohol metabolite, plays a pivotal role in the rewarding, motivational, and addictive properties of the parental compound. Many studies have investigated the role of ACD in mediating neurochemical and behavioral effects induced by alcohol administration, but very little is known about the modulation of neuropeptide systems following ACD intoxication and withdrawal. Indeed, the neuropeptide Y (NPY) system is altered during alcohol withdrawal in key regions for cerebrocortical excitability and neuroplasticity. The primary goal of this research was to investigate the effects of ACD intoxication and withdrawal by recording rat behavior and by measuring NPY immunoreactivity in hippocampus and NAcc, two brain regions mainly involved in processes which encompass neuroplasticity in alcohol dependence. Furthermore, on the basis of the involvement of endocannabinoidergic system in alcohol and ACD reinforcing effects, the role of the selective CB1 receptor antagonist AM281 in modulating NPY expression during withdrawal was assessed. Our results indicate that (i) ACD intoxication induced a reduction in NPY expression in hippocampus and NAcc; (ii) symptoms of physical dependence, similar to alcohol's, were scored at 12 h from the last administration of ACD; and (iii) NPY levels increased in early and prolonged acute withdrawal in both brain regions examined. The administration of AM281 was able to blunt signs of ACD-induced physical dependence, to modulate NPY levels, and to further increase NPY expression during ACD withdrawal both in hippocampus and NAcc. In conclusion, the present study shows that complex plastic changes take place in NPY system during ACD intoxication and subsequent withdrawal in rat hippocampal formation and NAcc. The pharmacological inhibition of CB1 signaling could counteract the neurochemical imbalance associated with ACD, and alcohol withdrawal, likely boosting the setting up of homeostatic functional recovery.
Collapse
Affiliation(s)
- Fulvio Plescia
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo , Palermo , Italy
| | - Anna Brancato
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo , Palermo , Italy
| | - Rosa Anna Maria Marino
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo , Palermo , Italy
| | - Carlotta Vita
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo , Palermo , Italy
| | - Michele Navarra
- Department of Drug Sciences and Products for Health, University of Messina , Messina , Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo , Palermo , Italy
| |
Collapse
|
24
|
Li X, Yu K, Zhang Z, Sun W, Yang Z, Feng J, Chen X, Liu CH, Wang H, Guo YP, He J. Cholecystokinin from the entorhinal cortex enables neural plasticity in the auditory cortex. Cell Res 2013; 24:307-30. [PMID: 24343575 PMCID: PMC3945883 DOI: 10.1038/cr.2013.164] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 10/03/2013] [Accepted: 11/12/2013] [Indexed: 01/12/2023] Open
Abstract
Patients with damage to the medial temporal lobe show deficits in forming new declarative memories but can still recall older memories, suggesting that the medial temporal lobe is necessary for encoding memories in the neocortex. Here, we found that cortical projection neurons in the perirhinal and entorhinal cortices were mostly immunopositive for cholecystokinin (CCK). Local infusion of CCK in the auditory cortex of anesthetized rats induced plastic changes that enabled cortical neurons to potentiate their responses or to start responding to an auditory stimulus that was paired with a tone that robustly triggered action potentials. CCK infusion also enabled auditory neurons to start responding to a light stimulus that was paired with a noise burst. In vivo intracellular recordings in the auditory cortex showed that synaptic strength was potentiated after two pairings of presynaptic and postsynaptic activity in the presence of CCK. Infusion of a CCKB antagonist in the auditory cortex prevented the formation of a visuo-auditory association in awake rats. Finally, activation of the entorhinal cortex potentiated neuronal responses in the auditory cortex, which was suppressed by infusion of a CCKB antagonist. Together, these findings suggest that the medial temporal lobe influences neocortical plasticity via CCK-positive cortical projection neurons in the entorhinal cortex.
Collapse
Affiliation(s)
- Xiao Li
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] University of Chinese Academy of Sciences (CAS) and CAS-Hong Kong Joint Laboratory, Institute of Biophysics, Beijing 100101, China [3] University of Chinese Academy of Sciences (CAS) and CAS-Hong Kong Joint Laboratory, Institute of Biophysics, Beijing 100101, China
| | - Kai Yu
- 1] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong [2] Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Zicong Zhang
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Wenjian Sun
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Zhou Yang
- 1] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong [2] Division of Life Science, Hong Kong University of Science and Technology, Clearwater Bay, N.T., Hong Kong SAR, China
| | - Jingyu Feng
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Xi Chen
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chun-Hua Liu
- 1] Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China [2] Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Guangdong 510530, China
| | - Haitao Wang
- 1] Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China [2] Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Guangdong 510530, China
| | - Yi Ping Guo
- 1] Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China [2] Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Guangdong 510530, China
| | - Jufang He
- 1] Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China [2] University of Chinese Academy of Sciences (CAS) and CAS-Hong Kong Joint Laboratory, Institute of Biophysics, Beijing 100101, China [3] University of Chinese Academy of Sciences (CAS) and CAS-Hong Kong Joint Laboratory, Institute of Biophysics, Beijing 100101, China [4] Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China [5] Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Guangdong 510530, China
| |
Collapse
|
25
|
Upregulation of gene expression in reward-modulatory striatal opioid systems by sleep loss. Neuropsychopharmacology 2013; 38:2578-87. [PMID: 23864029 PMCID: PMC3828536 DOI: 10.1038/npp.2013.174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/01/2013] [Accepted: 07/05/2013] [Indexed: 01/01/2023]
Abstract
Epidemiological studies have shown a link between sleep loss and the obesity 'epidemic,' and several observations indicate that sleep curtailment engenders positive energy balance via increased palatable-food 'snacking.' These effects suggest alterations in reward-modulatory brain systems. We explored the effects of 10 days of sleep deprivation in rats on the expression of striatal opioid peptide (OP) genes that subserve food motivation and hedonic reward, and compared effects with those seen in hypothalamic energy balance-regulatory systems. Sleep-deprived (Sleep-Dep) rats were compared with yoked forced-locomotion apparatus controls (App-Controls), food-restricted rats (Food-Restrict), and unmanipulated controls (Home-Cage). Detection of mRNA levels with in situ hybridization revealed a subregion-specific upregulation of striatal preproenkephalin and prodynorhin gene expression in the Sleep-Dep group relative to all other groups. Neuropeptide Y (NPY) gene expression in the hippocampal dentate gyrus and throughout neocortex was also robustly upregulated selectively in the Sleep-Dep group. In contrast, parallel gene expression changes were observed in the Sleep-Dep and Food-Restrict groups in hypothalamic energy-sensing systems (arcuate nucleus NPY was upregulated, and cocaine- and amphetamine-regulated transcript was downregulated), in alignment with leptin suppression in both groups. Together, these results reveal a novel set of sleep deprivation-induced transcriptional changes in reward-modulatory peptide systems, which are dissociable from the energy-balance perturbations of sleep loss or the potentially stressful effects of the forced-locomotion procedure. The recruitment of telencephalic food-reward systems may provide a feeding drive highly resistant to feedback control, which could engender obesity through the enhancement of palatable feeding.
Collapse
|
26
|
Santos-Carvalho A, Álvaro AR, Martins J, Ambrósio AF, Cavadas C. Emerging novel roles of neuropeptide Y in the retina: from neuromodulation to neuroprotection. Prog Neurobiol 2013; 112:70-9. [PMID: 24184719 DOI: 10.1016/j.pneurobio.2013.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 12/11/2022]
Abstract
Neuropeptide Y (NPY) and NPY receptors are widely expressed in the central nervous system, including the retina. Retinal cells, in particular neurons, astrocytes, and Müller, microglial and endothelial cells express this peptide and its receptors (Y1, Y2, Y4 and/or Y5). Several studies have shown that NPY is expressed in the retina of various mammalian and non-mammalian species. However, studies analyzing the distribution of NPY receptors in the retina are still scarce. Although the physiological roles of NPY in the retina have not been completely elucidated, its early expression strongly suggests that NPY may be involved in the development of retinal circuitry. NPY inhibits the increase in [Ca(2+)]i triggered by elevated KCl in retinal neurons, protects retinal neural cells against toxic insults and induces the proliferation of retinal progenitor cells. In this review, we will focus on the roles of NPY in the retina, specifically proliferation, neuromodulation and neuroprotection. Alterations in the NPY system in the retina might contribute to the pathogenesis of retinal degenerative diseases, such as diabetic retinopathy and glaucoma, and NPY and its receptors might be viewed as potentially novel therapeutic targets.
Collapse
Affiliation(s)
- Ana Santos-Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ana Rita Álvaro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Department of Biology and Environment, University of Trás-os-Montes and Alto Douro, Apartado 1013, 5001-801 Vila Real, Portugal
| | - João Martins
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - António Francisco Ambrósio
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal; AIBILI-Association for Innovation and Biomedical Research on Light and Image, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Cláudia Cavadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
27
|
Watakabe A, Hirokawa J, Ichinohe N, Ohsawa S, Kaneko T, Rockland KS, Yamamori T. Area-specific substratification of deep layer neurons in the rat cortex. J Comp Neurol 2013; 520:3553-73. [PMID: 22678985 DOI: 10.1002/cne.23160] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Gene markers are useful tools to identify cell types for fine mapping of neuronal circuits. Here we report area-specific sublamina structure of the rat cerebral cortex using cholecystokinin (cck) and purkinje cell protein4 (pcp4) mRNAs as the markers for excitatory neuron subtypes in layers 5 and 6. We found a segregated expression, especially pronounced in layer 6, where corticothalamic and corticocortical projecting neurons reside. To examine the relationship between gene expression and projection target, we injected retrograde tracers into several thalamic subnuclei, ventral posterior (VP), posterior (PO), mediodorsal (MD), medial and lateral geniculate nuclei (MGN and LGN); as well as into two cortical areas (M1 and V1). This combination of tracer-in situ hybridization (ISH) experiments revealed that corticocortical neurons predominantly express cck and corticothalamic neurons predominantly express pcp4 mRNAs in all areas tested. In general, cck(+) and pcp4(+) cells occupied the upper and lower compartment of layer 6a, respectively. However, the sublaminar distribution and the relative abundance of cck(+) and pcp4(+) cells were quite distinctive across areas. For example, layer 6 of the prelimbic cortex was almost devoid of cck(+) neurons, and was occupied instead by corticothalamic pcp4(+) neurons. In the lateral areas, such as S2, there was an additional layer of cck(+) cells positioned below the pcp4(+) compartment. The claustrum, which has a tight relationship with the cortex, mostly consisted of cck(+)/pcp4(-) cells. In summary, the combination of gene markers and retrograde tracers revealed a distinct sublaminar organization, with conspicuous cross-area variation in the arrangement and relative density of corticothalamic connections.
Collapse
Affiliation(s)
- Akiya Watakabe
- Division of Brain Biology, National Institute for Basic Biology, Graduate University for Advanced Studies, Okazaki, Japan.
| | | | | | | | | | | | | |
Collapse
|
28
|
Raghanti MA, Conley T, Sudduth J, Erwin JM, Stimpson CD, Hof PR, Sherwood CC. Neuropeptide Y-immunoreactive neurons in the cerebral cortex of humans and other haplorrhine primates. Am J Primatol 2012; 75:415-24. [PMID: 23042407 DOI: 10.1002/ajp.22082] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/13/2012] [Accepted: 08/14/2012] [Indexed: 11/09/2022]
Abstract
We examined the distribution of neurons immunoreactive for neuropeptide Y (NPY) in the posterior part of the superior temporal cortex (Brodmann's area 22 or area Tpt) of humans and nonhuman haplorrhine primates. NPY has been implicated in learning and memory and the density of NPY-expressing cortical neurons and axons is reduced in depression, bipolar disorder, schizophrenia, and Alzheimer's disease. Due to the role that NPY plays in both cognition and neurodegenerative diseases, we tested the hypothesis that the density of cortical and interstitial neurons expressing NPY was increased in humans relative to other primate species. The study sample included great apes (chimpanzee and gorilla), Old World monkeys (pigtailed macaque, moor macaque, and baboon) and New World monkeys (squirrel monkey and capuchin). Stereologic methods were used to estimate the density of NPY-immunoreactive (-ir) neurons in layers I-VI of area Tpt and the subjacent white matter. Adjacent Nissl-stained sections were used to calculate local densities of all neurons. The ratio of NPY-ir neurons to total neurons within area Tpt and the total density of NPY-ir neurons within the white matter were compared among species. Overall, NPY-ir neurons represented only an average of 0.006% of the total neuron population. While there were significant differences among species, phylogenetic trends in NPY-ir neuron distributions were not observed and humans did not differ from other primates. However, variation among species warrants further investigation into the distribution of this neuromodulator system.
Collapse
Affiliation(s)
- Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Fishell G, Rudy B. Mechanisms of inhibition within the telencephalon: "where the wild things are". Annu Rev Neurosci 2011; 34:535-67. [PMID: 21469958 DOI: 10.1146/annurev-neuro-061010-113717] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this review, we first provide a historical perspective of inhibitory signaling from the discovery of inhibition through to our present understanding of the diversity and mechanisms by which GABAergic interneuron populations function in different parts of the telencephalon. This is followed by a summary of the mechanisms of inhibition in the CNS. With this as a starting point, we provide an overview describing the variations in the subtypes and origins of inhibitory interneurons within the pallial and subpallial divisions of the telencephalon, with a focus on the hippocampus, somatosensory, paleo/piriform cortex, striatum, and various amygdala nuclei. Strikingly, we observe that marked variations exist in the origin and numerical balance between GABAergic interneurons and the principal cell populations in distinct regions of the telencephalon. Finally we speculate regarding the attractiveness and challenges of establishing a unifying nomenclature to describe inhibitory neuron diversity throughout the telencephalon.
Collapse
Affiliation(s)
- Gord Fishell
- Smilow Neuroscience Program, Smilow Research Center, New York University School of Medicine, New York, New York 10016, USA.
| | | |
Collapse
|
30
|
Blazquez-Llorca L, García-Marín V, DeFelipe J. GABAergic complex basket formations in the human neocortex. J Comp Neurol 2011; 518:4917-37. [PMID: 21031559 DOI: 10.1002/cne.22496] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Certain GABAergic interneurons in the cerebral cortex, basket cells, establish multiple connections with cell bodies that typically outline the somata and proximal dendrites of pyramidal cells. During studies into the distribution of the vesicular GABA transporter (VGAT) in the human cerebral cortex, we were struck by the presence of a very dense, pericellular arrangement of multiple VGAT-immunoreactive (-ir) terminals in certain cortical areas. We called these terminals "Complex basket formations" (Cbk-formations) to distinguish them from the simpler and more typical pericellular GABAergic innervations of most cortical neurons. Here we examined the distribution of these VGAT-ir Cbk-formations in various cortical areas, including the somatosensory (area 3b), visual (areas 17 and 18), motor (area 4), associative frontal (dorsolateral areas 9, 10, 45, 46, and orbital areas 11, 12, 13, 14, 47), associative temporal (areas 20, 21, 22, and 38), and limbic cingulate areas (areas 24, 32). Furthermore, we used dual or triple staining techniques to study the chemical nature of the innervated cells. We found that VGAT-ir Cbk-formations were most frequently found in area 4 followed by areas 3b, 13, and 18. In addition, they were mostly observed in layer III, except in area 17, where they were most dense in layer IV. We also found that 70% of the innervated neurons were pyramidal cells, while the remaining 30% were multipolar cells. Most of these multipolar cells expressed the calcium-binding protein parvalbumin and the lectin Vicia villosa agglutinin.
Collapse
|
31
|
Trent NL, Menard JL. Infusions of neuropeptide Y into the lateral septum reduce anxiety-related behaviors in the rat. Pharmacol Biochem Behav 2011; 99:580-90. [PMID: 21693128 DOI: 10.1016/j.pbb.2011.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/23/2011] [Accepted: 06/06/2011] [Indexed: 11/24/2022]
Abstract
Neuropeptide Y (NPY) is one of the most abundant peptides in mammalian brain and NPY-like-immunoreactivity is highly expressed in the lateral septum, an area extensively involved in anxiety regulation. NPY counteracts the neurochemical and behavioral responses to acute threat in animal models, and intracerebroventricular (i.c.v.) administration of NPY at low doses is anxiolytic. Less is known about the specific contributions of the lateral septum to NPY-mediated anxiety regulation. In Experiment 1, the effects of infusions of NPY (1.5 μg) into the lateral septum were investigated in three animal models of anxiety: the elevated plus-maze, novelty-induced suppression of feeding, and shock-probe burying tests. Experiment 2 examined the role of the NPY Y1 receptor in these models by co-infusing the Y1 antagonist BIBO 3304 (0.15 μg, 0.30 μg) with NPY into the lateral septum. In the elevated plus-maze, there were no changes in rats' open arm exploration, the index of anxiety reduction in this test. In the novelty-induced suppression of feeding test, rats infused with NPY showed decreases in the latency to consume a palatable snack in a novel (but not familiar) environment, suggesting a reduction in anxiety independent of increases in appetite. This anxiolysis was attenuated by co-infusion with BIBO 3304 (0.30 μg) in Experiment 2. Lastly, rats infused with NPY showed decreases in the duration of burying behavior in the shock-probe burying test, also indicative of anxiety reduction. However, unlike in the feeding test, BIBO 3304 did not attenuate the NPY-induced anxiolysis in the shock-probe test. It is concluded that NPY produces anxiolytic-like actions in the lateral septum in two animal models of anxiety: the novelty-induced suppression of feeding, and shock-probe burying tests, and that this anxiolysis is dependent on Y1 receptor activation in the feeding test.
Collapse
Affiliation(s)
- Natalie L Trent
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
32
|
Khaira SK, Nefzger CM, Beh SJ, Pouton CW, Haynes JM. Midbrain and forebrain patterning delivers immunocytochemically and functionally similar populations of neuropeptide Y containing GABAergic neurons. Neurochem Int 2011; 59:413-20. [PMID: 21349310 DOI: 10.1016/j.neuint.2011.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/21/2011] [Accepted: 02/11/2011] [Indexed: 10/18/2022]
Abstract
Neurons differentiated in vitro from embryonic stem cells (ESCs) have the potential to serve both as models of disease states and in drug discovery programs. In this study, we use sonic hedgehog (SHH) and fibroblast growth factor 8 (FGF-8) to enrich for forebrain and midbrain phenotypes from mouse ESCs. We then investigate, using Ca(2+) imaging and [(3)H]-GABA release studies, whether the GABAergic neurons produced exhibit distinct functional phenotypes. At day 24 of differentiation, reverse transcriptase-PCR showed the presence of both forebrain (Bf-1, Hesx1, Pgc-1α, Six3) and midbrain (GATA2, GATA3) selective mRNA markers in developing forebrain-enriched cultures. All markers were present in midbrain cultures except for Bf-1 and Pgc-1α. Irrespective of culture conditions all GABA immunoreactive neurons were also immunoreactive to neuropeptide Y (NPY) antibodies. Forebrain and midbrain GABAergic neurons responded to ATP (1 mM), L-glutamate (30 μM), noradrenaline (30 μM), acetylcholine (30 μM) and dopamine (30 μM), with similar elevations of intracellular Ca(2+)([Ca(2+)](i)). The presence of GABA(A) and GABA(B) antagonists, bicuculline (30 μM) and CGP55845 (1 μM), increased the elevation of [Ca(2+)](i) in response to dopamine (30 μM) in midbrain, but not forebrain GABAergic neurons. All agonists, except dopamine, elicited similar [(3)H]-GABA release from forebrain and midbrain cultures. Dopamine (30 μM) did not stimulate significant [(3)H]-GABA release in midbrain cultures, although it was effective in forebrain cultures. This study shows that differentiating neurons toward a midbrain fate restricts the expression of forebrain markers. Forebrain differentiation results in the expression of forebrain and midbrain markers. All GABA(+) neurons contain NPY, and show similar agonist-induced elevations of [Ca(2+)](i) and [(3)H]-GABA release. This study indicates that the pharmacological phenotype of these particular neurons may be independent of the addition of the patterning factors that direct neurons toward forebrain and midbrain fates.
Collapse
Affiliation(s)
- S K Khaira
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
33
|
Kubota Y, Shigematsu N, Karube F, Sekigawa A, Kato S, Yamaguchi N, Hirai Y, Morishima M, Kawaguchi Y. Selective coexpression of multiple chemical markers defines discrete populations of neocortical GABAergic neurons. ACTA ACUST UNITED AC 2011; 21:1803-17. [PMID: 21220766 DOI: 10.1093/cercor/bhq252] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Whether neocortical γ-aminobutyric acid (GABA) cells are composed of a limited number of distinct classes of neuron, or whether they are continuously differentiated with much higher diversity, remains a contentious issue for the field. Most GABA cells of rat frontal cortex have at least 1 of 6 chemical markers (parvalbumin, calretinin, alpha-actinin-2, somatostatin, vasoactive intestinal polypeptide, and cholecystokinin), with each chemical class comprising several distinct neuronal subtypes having specific physiological and morphological characteristics. To better clarify GABAergic neuron diversity, we assessed the colocalization of these 6 chemical markers with corticotropin-releasing factor (CRF), neuropeptide Y (NPY), the substance P receptor (SPR), and nitric oxide synthase (NOS); these 4 additional chemical markers suggested to be expressed diversely or specifically among cortical GABA cells. We further correlated morphological and physiological characteristics of identified some chemical subclasses of inhibitory neurons. Our results reveal expression specificity of CRF, NPY, SPR, and NOS in morphologically and physiologically distinct interneuron classes. These observations support the existence of a limited number of functionally distinct subtypes of GABA cells in the neocortex.
Collapse
Affiliation(s)
- Yoshiyuki Kubota
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Miyazaki T, Yamasaki M, Uchigashima M, Matsushima A, Watanabe M. Cellular expression and subcellular localization of secretogranin II in the mouse hippocampus and cerebellum. Eur J Neurosci 2010; 33:82-94. [PMID: 21044184 DOI: 10.1111/j.1460-9568.2010.07472.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Secretogranin II (SgII), or chromogranin C, is thought to participate in the sorting and packaging of peptide hormones and neuropeptides into secretory granules and large dense-core vesicle (LDCVs), and also functions as a precursor of neuropeptide secretoneurin. Although SgII is widely distributed in the brain and is predominantly localized at terminals of mossy fibers in the hippocampus and cerebellum and climbing fibers in the cerebellum, its cellular expression and ultrastructural localization remain largely unknown. In the present study, we addressed this issue in the adult mouse brain by multiple-labeling fluorescence in situ hybridization and immunofluorescence and by preembedding and postembedding immunoelectron microscopies. SgII was expressed in various neurons, distributed as either tiny puncta or coarse aggregates in the neuropil, and intensely accumulated in perikarya of particular neurons, such as parvalbumin-positive interneurons and mossy cells in the hippocampus and Purkinje cells in the cerebellum. Coarse aggregates were typical of terminals of mossy fibers and climbing fibers. In these terminals, numerous immunogold particles were clustered on individual LDCVs, and one or two particles also fell within small synaptic vesicle-accumulating portions. SgII was further detected as tiny puncta in neural elements lacking LDCVs, such as parallel fibers of cerebellar granule cells, somatodendritic elements of various neurons and Bergmann glia. Thus, SgII is present in LDCV and non-LDCV compartments of various neural cells. The wide subcellular localization of SgII may reflect diverse release sites of neuropeptides and secretorneurin, or suggests its role in the sorting and packaging of molecules other than neuropeptides in non-LDCV compartments.
Collapse
Affiliation(s)
- Taisuke Miyazaki
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | | | | | | | | |
Collapse
|
35
|
Novel animal models for studying complex brain disorders: BAC-driven miRNA-mediated in vivo silencing of gene expression. Mol Psychiatry 2010; 15:987-95. [PMID: 20125089 PMCID: PMC3011211 DOI: 10.1038/mp.2010.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In schizophrenia, glutamic acid decarboxylase 1 (GAD1) disturbances are robust, consistently observed, cell-type specific and represent a core feature of the disease. In addition, neuropeptide Y (NPY), which is a phenotypic marker of a sub-population of GAD1-containing interneurons, has shown reduced expression in the prefrontal cortex in subjects with schizophrenia, suggesting that dysfunction of the NPY+ cortical interneuronal sub-population might be a core feature of this devastating disorder. However, modeling gene expression disturbances in schizophrenia in a cell type-specific manner has been extremely challenging. To more closely mimic these molecular and cellular human post-mortem findings, we generated a transgenic mouse in which we downregulated GAD1 mRNA expression specifically in NPY+ neurons. This novel, cell type-specific in vivo system for reducing gene expression uses a bacterial artificial chromosome (BAC) containing the NPY promoter-enhancer elements, the reporter molecule (eGFP) and a modified intron containing a synthetic microRNA (miRNA) targeted to GAD1. The animals of isogenic strains are generated rapidly, providing a new tool for better understanding the molecular disturbances in the GABAergic system observed in complex neuropsychiatric disorders such as schizophrenia. In the future, because of the small size of the silencing miRNAs combined with our BAC strategy, this method may be modified to allow generation of mice with simultaneous silencing of multiple genes in the same cells with a single construct, and production of splice-variant-specific knockdown animals.
Collapse
|
36
|
Janzsó G, Valcz G, Thuma A, Szoke B, Lendvai Z, Abrahám H, Kozicz T, Halasy K. Cocaine- and amphetamine-regulated transcript (CART) peptide-immunopositive neuronal elements in the lateral septum: rostrocaudal distribution in the male rat. Brain Res 2010; 1362:40-7. [PMID: 20883668 DOI: 10.1016/j.brainres.2010.09.079] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 09/07/2010] [Accepted: 09/22/2010] [Indexed: 11/26/2022]
Abstract
The morphological features and distribution of cocaine- and amphetamine-regulated transcript peptide immunoreactivity (CART-IR) were studied in the lateral septum (LS) of male rats using light and electron microscopic immunocytochemistry and computer-aided densitometry. CART-IR was detected along the rostrocaudal axis of the LS in varicose axonal fibers only, although immunoreactive cell bodies and dendrites were not detected. Pericellular basket-like arrangements around immunonegative cell bodies were present. From among the targets of such pericellular baskets, glutamic acid decarboxylase (GAD)-immunopositive and NPY-immunoreactive somata were identified. Thin varicose axons were present in each section, whereas thick varicose axons were restricted to the sections of rostral position only. CART-IR was observed in varicose fibers forming a dense subependymal plexus, from which solitary varicose fibers entered the ependymal layer. The fine structure of varicosities was similar to that of other neuropeptide-containing fibers. Small varicosities established asymmetrical synaptic contacts mainly with dendrites and dendritic spines, and larger varicosities established symmetrical synapses with somata and dendritic shafts. CART-to-CART connections were not revealed. The density curve of the CART-IR along the rostrocaudal axis of LS was found to be paraboloid. CART is known as one of the most anorexigenic peptides. These results serve as basis for further physiological studies concerning the biological significance of lateral septal CART peptide in the regulation of food intake.
Collapse
Affiliation(s)
- Gergely Janzsó
- Department of Anatomy and Histology, Faculty of Veterinary Sciences, Szent István University, Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen X, Dzakpasu R. Observed network dynamics from altering the balance between excitatory and inhibitory neurons in cultured networks. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2010; 82:031907. [PMID: 21230108 DOI: 10.1103/physreve.82.031907] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 08/05/2010] [Indexed: 05/30/2023]
Abstract
Complexity in the temporal organization of neural systems may be a reflection of the diversity of their neural constituents. These constituents, excitatory and inhibitory neurons, comprise a well-defined ratio in vivo and form the substrate for rhythmic oscillatory activity. To begin to elucidate the dynamical implications that underlie this balance, we construct neural circuits not ordinarily found in nature and study the resulting temporal patterns. We culture several networks of neurons composed of varying fractions of excitatory and inhibitory cells and use a multielectrode array to study their temporal dynamics as this balance is modulated. We use the electrode burst as the temporal imprimatur to signify the presence of network activity. Burst durations, interburst intervals, and the number of spikes participating within a burst are used to illustrate the vivid differences in the temporal organization between the various cultured networks. When the network consists largely of excitatory neurons, no network temporal structure is apparent. However, the addition of inhibitory neurons evokes a temporal order. Calculation of the temporal autocorrelation shows that when the number of inhibitory neurons is a major fraction of the network, a striking network pattern materializes when none was previously present.
Collapse
Affiliation(s)
- Xin Chen
- Department of Physics, Georgetown University, Washington, DC 20057, USA
| | | |
Collapse
|
38
|
Muzzi P, Camera P, Di Cunto F, Vercelli A. Deletion of the citron kinase gene selectively affects the number and distribution of interneurons in barrelfield cortex. J Comp Neurol 2009; 513:249-64. [PMID: 19148892 DOI: 10.1002/cne.21927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Citron kinase (CIT-K), a ser/thr kinase, is required during neurogenesis for cytokinesis of neuronal precursors. Deletion of the cit-k gene in mice (cit-k(-/-) mice) leads to a severe malformative central nervous system syndrome characterized by microencephaly, ataxia, and epileptic seizures; affected mice die by the third week of postnatal life. We have used NADPH-diaphorase histochemistry, immunostaining for calbindin, calretinin, parvalbumin, and glutamic acid decarboxylase 67 (GAD67), and histological staining to undertake qualitative and quantitative analyses of the morphology and distribution of interneurons in the barrelfield cortex of cit-k(-/-) mice. By postnatal day 13, lack of CIT-K results in profoundly altered cortical cell morphology: the infragranular layers are populated by large, binucleate interneurons bearing many more dendrites than in control mice, an anatomical profile that has also been reported for the cortex of humans with cortical dysplasias and epilepsy. Tessellation analyses reveal that a clustered distribution of interneurons is maintained in cit-k(-/-) mice, but that their nearest neighbor distance is significantly increased, and thus their density is reduced; the overall number of interneurons is more dramatically decreased in the absence of CIT-K than would be predicted on the basis of the reduced brain size of affected mice. This reduction of inhibitory gamma-aminobutyric acid (GABA)ergic neurons likely underlies the occurrence of epileptic seizures in the cit-k(-/-) mice. Furthermore, the altered distribution of NADPH-diaphorase-positive interneurons could be responsible for an impaired coupling of cortical activity to blood flow, also affecting cortical growth and functioning.
Collapse
Affiliation(s)
- Patrizia Muzzi
- Department of Anatomy, Pharmacology and Forensic Medicine, University of Torino, 10126 Torino, Italy
| | | | | | | |
Collapse
|
39
|
Hilke S, Holm L, Man K, Hökfelt T, Theodorsson E. Rapid change of neuropeptide Y levels and gene-expression in the brain of ovariectomized mice after administration of 17beta-estradiol. Neuropeptides 2009; 43:327-32. [PMID: 19481799 DOI: 10.1016/j.npep.2009.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 03/23/2009] [Accepted: 04/23/2009] [Indexed: 10/20/2022]
Abstract
Estrogen alters excitability and changes synaptic morphology in the rat hippocampal formation. We have compared, by means of radioimmunoassay and in situ hybridization, the effects of short-term treatment with 17beta-estradiol on neuropeptide Y (NPY) in the brain of ovariectomized mice. A highly significant reduction in concentrations of NPY-like immunoreactivity (LI) was observed in the hippocampal formation, some cortical areas and the caudate nucleus 1h after administration of 17beta-estradiol as compared to the control group. In contrast, NPY transcript levels increased in the hippocampal formation (dentate gyrus) and the caudate nucleus, possibly representing a compensatory increase of NPY synthesis following increased estradiol-induced NPY release. These data suggest that 17beta-estradiol, via membrane-related mechanisms, increases NPY release and synthesis in forebrain areas involved in cognition, mood and motor functions.
Collapse
Affiliation(s)
- Susanne Hilke
- Department of Clinical and Experimental Medicine, Division of Clinical Chemistry, Faculty of Health and Sciences, University Hospital, SE-581 85 Linköping, Sweden.
| | | | | | | | | |
Collapse
|
40
|
Widerlöv E, Heilig M, Ekman R, Wahlestedt C. Possible relationship between neuropeptide Y (NPY) and major depression – evidence from human and animal studies. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/08039488809103218] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
41
|
Abstract
Neuropeptide Y (NPY) is an abundant neuropeptide of the neocortex involved in numerous physiological and pathological processes. Because of the large electrophysiological, molecular, and morphological diversity of NPY-expressing neurons their precise identity remains unclear. To define distinct populations of NPY neurons we characterized, in acute slices of rat barrel cortex, 200 cortical neurons of layers I-IV by means of whole-cell patch-clamp recordings, biocytin labeling, and single-cell reverse transcriptase-PCR designed to probe for the expression of well established molecular markers for cortical neurons. To classify reliably cortical NPY neurons, we used and compared different unsupervised clustering algorithms based on laminar location and electrophysiological and molecular properties. These classification schemes confirmed that NPY neurons are nearly exclusively GABAergic and consistently disclosed three main types of NPY-expressing interneurons. (1) Neurogliaform-like neurons exhibiting a dense axonal arbor, were the most frequent and superficial, and substantially expressed the neuronal isoform of nitric oxide synthase. (2) Martinotti-like cells characterized by an ascending axon ramifying in layer I coexpressed somatostatin and were the most excitable type. (3) Among fast-spiking and parvalbumin-positive basket cells, NPY expression was correlated with pronounced spike latency. By clarifying the diversity of cortical NPY neurons, this study establishes a basis for future investigations aiming at elucidating their physiological roles.
Collapse
|
42
|
The neuropeptides CCK and NPY and the changing view of cell-to-cell communication in the taste bud. Physiol Behav 2009; 97:581-91. [PMID: 19332083 DOI: 10.1016/j.physbeh.2009.02.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 02/16/2009] [Indexed: 11/24/2022]
Abstract
The evolving view of the taste bud increasingly suggests that it operates as a complex signal processing unit. A number of neurotransmitters and neuropeptides and their corresponding receptors are now known to be expressed in subsets of taste receptor cells in the mammalian bud. These expression patterns set up hard-wired cell-to-cell communication pathways whose exact physiological roles still remain obscure. As occurs in other cellular systems, it is likely that neuropeptides are co-expressed with neurotransmitters and function as neuromodulators. Several neuropeptides have been identified in taste receptor cells including cholecystokinin (CCK), neuropeptide Y (NPY), vasoactive intestinal peptide (VIP), and glucagon-like peptide 1 (GLP-1). Of these, CCK and NPY are the best studied. These two peptides are co-expressed in the same presynaptic cells; however, their postsynaptic actions are both divergent and antagonistic. CCK and its receptor, the CCK-1 subtype, are expressed in the same subset of taste receptor cells and the autocrine activation of these cells produces a number of excitatory physiological actions. Further, most of these cells are responsive to bitter stimuli. On the other hand, NPY and its receptor, the NPY-1 subtype, are expressed in different cells. NPY, acting in a paracrine fashion on NPY-1 receptors, results in inhibitory actions on the cell. Preliminary evidence suggests the NPY-1 receptor expressing cell co-expresses T1R3, a member of the T1R family of G-protein coupled receptors thought to be important in detection of sweet and umami stimuli. Thus the neuropeptide expressing cells co-express CCK, NPY, and CCK-1 receptor. Neuropeptides released from these cells during bitter stimulation may work in concert to both modulate the excitation of bitter-sensitive taste receptor cells while concurrently inhibiting sweet-sensitive cells. This modulatory process is similar to the phenomenon of lateral inhibition that occurs in other sensory systems.
Collapse
|
43
|
Sherrin T, Todorovic C, Zeyda T, Tan CH, Wong PTH, Zhu YZ, Spiess J, Spiess J. Chronic stimulation of corticotropin-releasing factor receptor 1 enhances the anxiogenic response of the cholecystokinin system. Mol Psychiatry 2009; 14:291-307. [PMID: 18195718 DOI: 10.1038/sj.mp.4002121] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Corticotropin-releasing factor (CRF) and cholecystokinin (CCK), two highly colocalized neuropeptides, have been linked to the etiology of stress-related anxiety disorders. Recent evidence points to the possibility that some of the anxiogenic effects of the central CCK system take place through interplay with the CRF system. The aim of the present study was to examine the effects of chronic, mild activation of CRF receptor 1 (CRF(1)) on the central CCK system of the C57BL/6J mouse. As shown by in situ hybridization, real-Time PCR and immunohistochemistry, 5 days of intracerebroventricular (i.c.v.) injections of a subeffective dose (2.3 pmol) of cortagine, a CRF(1)-selective agonist, resulted in an increase in CCK mRNA levels and CCK(2) receptor immunoreactivity in several brain regions, such as amygdala and hippocampus, known to be involved in the regulation of anxiety. Mice with elevated endogenous central CCK tone exhibited significantly higher anxiety-like behaviors in the open-field task and elevated plus maze, and enhanced conditioned fear. These behavioral changes were reversed by i.c.v. administration of the CCK(2)-selective antagonist LY225910, after 5 days of priming with cortagine. Under the same conditions, the intraperitoneal administration of the CRF(1) antagonist antalarmin was ineffective. This result indicated that once the CCK system was sensitized by prior CRF(1) activation, it exhibited its anxiogenic effects, without influence by CRF(1), possibly because of its observed downregulation. In sum, our results provide a novel model for the interaction of the CRF and CCK systems contributing to the development of hypersensitive emotional circuitry.
Collapse
Affiliation(s)
- T Sherrin
- Specialized Neuroscience Research Program, University of Hawaii, Honolulu, HI 96813, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hillman KL, Lei S, Doze VA, Porter JE. Alpha-1A adrenergic receptor activation increases inhibitory tone in CA1 hippocampus. Epilepsy Res 2009; 84:97-109. [PMID: 19201164 DOI: 10.1016/j.eplepsyres.2008.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 12/19/2008] [Accepted: 12/27/2008] [Indexed: 11/19/2022]
Abstract
The endogenous catecholamine norepinephrine (NE) exhibits anti-epileptic properties, however it is not well understood which adrenergic receptor (AR) mediates this effect. The aim of this study was to investigate alpha(1)-adrenergic receptor activation in region CA1 of the hippocampus, a subcortical structure often implicated in temporal lobe epilepsies. Using cell-attached and whole-cell recordings in rat hippocampal slices, we confirmed that selective alpha(1)-AR activation increases action potential firing in a subpopulation of CA1 interneurons. We found that this response is mediated via the alpha(1A)-AR subtype, initiated by sodium influx, and appears independent of second messenger signaling. In CA1 pyramidal cells, alpha(1A)-AR activation decreases activity due to increased pre-synaptic GABA and somatostatin release. Examination of post-synaptic receptor involvement revealed that while GABA(A) receptors mediate the majority of alpha(1A)-adrenergic effects on CA1 pyramidal cells, significant contributions are also made by GABA(B) and somatostatin receptors. Finally, to test whether alpha(1A)-AR activation could have potential therapeutic implications, we performed AR agonist challenges using two in vitro epileptiform models. When GABA(A) receptors were available, alpha(1A)-AR activation significantly decreased epileptiform bursting in CA1. Together, our findings directly link stimulation of the alpha(1A)-AR subtype to release of GABA and somatostatin at the single cell level and suggest that alpha(1A)-AR activation may represent one mechanism by which NE exerts anti-epileptic effects within the hippocampus.
Collapse
Affiliation(s)
- Kristin L Hillman
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, ND 58203, United States
| | | | | | | |
Collapse
|
45
|
Abstract
The distribution and morphology of neurons containing somatostatin (SOM) was investigated in the amygdala (CA) of the pig. The SOM-immunoreactive (SOM-IR) cell bodies and fibres were present in all subdivisions of the porcine CA, however, their number and density varied depending on the nucleus studied. The highest density of SOM-positive somata was observed in the layer III of the cortical nuclei, in the anterior (magnocellular) part of the basomedial nucleus and in the caudal (large-celled) part of the lateral nucleus. Moderate to high numbers of SOM-IR cells were also observed in the medial and basolateral nuclei. Many labeled neurons were also consistently observed in the lateral part of the central nucleus. In the remaining CA regions, the density of SOM-positive cell bodies varied from moderate to low. In any CA region studied SOM-IR neurons formed heterogeneous population consisting of small, rounded or slightly elongated cell bodies, with a few poorly branched smooth dendrites. In general, morphological features of these cells clearly resembled the non-pyramidal Golgi type II interneurons. The routine double-labeling studies with antisera directed against SOM and neuropeptide Y (NPY) demonstrated that a large number of SOM-IR cell bodies and fibers in all studied CA areas contained simultaneously NPY. In contrast, co-localization of SOM and cholecystokinin (CCK) or SOM and vasoactive intestinal polypeptide (VIP) was never seen in cell bodies and fibres in any of nuclei studied. In conclusion, SOM-IR neurons of the porcine amygdala form large and heterogeneous subpopulation of, most probably, interneurons that often contain additionally NPY. On the other hand, CCK- and/or VIP-IR neurons belonged to another, discrete subpopulations of porcine CA neurons.
Collapse
|
46
|
Disney AA, Aoki C. Muscarinic acetylcholine receptors in macaque V1 are most frequently expressed by parvalbumin-immunoreactive neurons. J Comp Neurol 2008; 507:1748-62. [PMID: 18265004 DOI: 10.1002/cne.21616] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Acetylcholine (ACh) is believed to underlie mechanisms of arousal and attention in mammals. ACh also has a demonstrated functional effect in visual cortex that is both diverse and profound. We have reported previously that cholinergic modulation in V1 of the macaque monkey is strongly targeted toward GABAergic interneurons. Here we examine the localization of m1 and m2 muscarinic receptor subtypes across subpopulations of GABAergic interneurons--identified by their expression of the calcium-binding proteins parvalbumin, calbindin, and calretinin--using dual-immunofluorescence confocal microscopy in V1 of the macaque monkey. In doing so, we find that the vast majority (87%) of parvalbumin-immunoreactive neurons express m1-type muscarinic ACh receptors. m1 receptors are also expressed by 60% of calbindin-immunoreactive neurons and 40% of calretinin-immunoreactive neurons. m2 AChRs, on the other hand, are expressed by only 31% of parvalbumin neurons, 23% of calbindin neurons, and 25% of calretinin neurons. Parvalbumin-immunoreactive cells comprise approximately 75% of the inhibitory neuronal population in V1 and included in this large subpopulation are neurons known to veto and regulate the synchrony of principal cell spiking. Through the expression of m1 ACh receptors on nearly all of these PV cells, the cholinergic system avails itself of powerful control of information flow through and processing within the network of principal cells in the cortical circuit.
Collapse
Affiliation(s)
- Anita A Disney
- Center for Neural Science, New York University, New York, New York 10003, USA.
| | | |
Collapse
|
47
|
Gilpin NW, Misra K, Koob GF. Neuropeptide Y in the central nucleus of the amygdala suppresses dependence-induced increases in alcohol drinking. Pharmacol Biochem Behav 2008; 90:475-80. [PMID: 18501411 DOI: 10.1016/j.pbb.2008.04.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 03/19/2008] [Accepted: 04/09/2008] [Indexed: 11/25/2022]
Abstract
The anxiolytic effects of neuropeptide Y (NPY) are mediated in part by the central nucleus of the amygdala (CeA), a brain region involved in the regulation of alcohol-drinking behaviors. Centrally administered NPY suppresses alcohol drinking in subpopulations of rats vulnerable to the development of high alcohol-drinking behavior. The purpose of the current study was to determine the role of NPY in the CeA on elevated alcohol drinking produced by alcohol dependence. Adult male Wistar rats were trained to respond for 10% w/v alcohol in an operant situation with the use of a supersaccharin fading procedure. Following stabilization of responding, rats were divided into two groups matched for intake and given daily access to either alcohol-containing (9.2% v/v) liquid diet or an isocaloric control diet. Following extended access to the diet and reliable separation of operant responding between dependent and non-dependent rats during 6-h withdrawal tests, all rats were implanted bilaterally with cannulae aimed at the CeA. Rats were then infused with 4 NPY doses (0.0, 0.25, 0.5, 1.0 microg/0.5 microl aCSF) in a within-subjects Latin-square design during acute withdrawal and tested for operant alcohol responding 30 min later. Alcohol-dependent rats exhibited higher operant alcohol responding than non-dependent rats when infused with vehicle, but responding was similar in the two groups following infusion of all doses of NPY. These results indicate that NPY abolishes dependence-induced elevations in alcohol drinking and implicate the recruitment of limbic NPY systems in the motivational drive to consume alcohol following the transition to dependence.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, United States.
| | | | | |
Collapse
|
48
|
Rasmussen K. Section Review—Central & Peripheral Nervous Systems: Therapeutic Potential of Cholecystokinin-B Antagonists. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.4.313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Abstract
Glutamate and GABA, the two most abundant neurotransmitters in the mammalian central nervous system, can act on metabotropic receptors that are structurally quite dissimilar from those targeted by most other neurotransmitters/modulators. Accordingly, metabotropic glutamate receptors (mGluRs) and GABA(B) receptors (GABA(B)Rs) are classified as members of family 3 (or family C) of G protein-coupled receptors. On the other hand, mGluRs and GABA(B)Rs exhibit pronounced and partly unresolved differences between each other. The most intriguing difference is that mGluRs exist as multiple pharmacologically as well as structurally distinct subtypes, whereas, in the case of GABA(B)Rs, molecular biologists have so far identified only one structurally distinct heterodimeric complex whose few variants seem unable to explain the pharmacological heterogeneity of GABA(B)Rs observed in many functional studies. Both mGluRs and GABA(B)Rs can be localized on axon terminals of different neuronal systems as presynaptic autoreceptors and heteroreceptors modulating the exocytosis of various transmitters.
Collapse
Affiliation(s)
- M Raiteri
- Department of Experimental Medicine, Pharmacology and Toxicology Section, University of Genoa, 16148 Genoa, Italy.
| |
Collapse
|
50
|
Bayatti N, Moss JA, Sun L, Ambrose P, Ward JFH, Lindsay S, Clowry GJ. A molecular neuroanatomical study of the developing human neocortex from 8 to 17 postconceptional weeks revealing the early differentiation of the subplate and subventricular zone. Cereb Cortex 2007; 18:1536-48. [PMID: 17965125 PMCID: PMC2430151 DOI: 10.1093/cercor/bhm184] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have employed immunohistochemistry for multiple markers to investigate the structure and possible function of the different compartments of human cerebral wall from the formation of cortical plate at 8 postconceptional weeks (PCW) to the arrival of thalamocortical afferents at 17 PCW. New observations include the subplate emerging as a discrete differentiated layer by 10 PCW, characterized by synaptophysin and vesicular gamma-aminobutyric acid transporter expression also seen in the marginal zone, suggesting that these compartments may maintain a spontaneously active synaptic network even before the arrival of thalamocortical afferents. The subplate expanded from 13 to 17 PCW, becoming the largest compartment and differentiated further, with NPY neurons located in the outer subplate and KCC2 neurons in the inner subplate. Glutamate decarboxylase and calretinin-positive inhibitory neurons migrated tangentially and radially from 11.5 PCW, appearing in larger numbers toward the rostral pole. The proliferative zones, marked by Ki67 expression, developed a complicated structure by 12.5 PCW reflected in transcription factor expression patterns, including TBR2 confined to the inner subventricular and outer ventricular zones and TBR1 weakly expressed in the subventricular zone (SVZ). PAX6 was extensively expressed in the proliferative zones such that the human outer SVZ contained a large reservoir of PAX6-positive potential progenitor cells.
Collapse
Affiliation(s)
- Nadhim Bayatti
- School of Clinical Medical Sciences, Department of Child Health, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP, UK
| | | | | | | | | | | | | |
Collapse
|