1
|
Borland JM. The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents. Neurosci Biobehav Rev 2024; 164:105809. [PMID: 39004323 DOI: 10.1016/j.neubiorev.2024.105809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BORLAND, J.M., The effects of different types of social interactions on the electrophysiology of neurons in the nucleus accumbens in rodents, NEUROSCI BIOBEH REV 21(1) XXX-XXX, 2024.-Sociality shapes an organisms' life. The nucleus accumbens is a critical brain region for mental health. In the following review, the effects of different types of social interactions on the physiology of neurons in the nucleus accumbens is synthesized. More specifically, the effects of sex behavior, aggression, social defeat, pair-bonding, play behavior, affiliative interactions, parental behaviors, the isolation from social interactions and maternal separation on measures of excitatory synaptic transmission, intracellular signaling and factors of transcription and translation in neurons in the nucleus accumbens in rodent models are reviewed. Similarities and differences in effects depending on the type of social interaction is then discussed. This review improves the understanding of the molecular and synaptic mechanisms of sociality.
Collapse
|
2
|
Benoit E, Lyons DG, Rihel J. Noradrenergic tone is not required for neuronal activity-induced rebound sleep in zebrafish. J Comp Physiol B 2024; 194:279-298. [PMID: 37480493 PMCID: PMC11233345 DOI: 10.1007/s00360-023-01504-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/03/2023] [Indexed: 07/24/2023]
Abstract
Sleep pressure builds during wakefulness, but the mechanisms underlying this homeostatic process are poorly understood. One zebrafish model suggests that sleep pressure increases as a function of global neuronal activity, such as during sleep deprivation or acute exposure to drugs that induce widespread brain activation. Given that the arousal-promoting noradrenergic system is important for maintaining heightened neuronal activity during wakefulness, we hypothesised that genetic and pharmacological reduction of noradrenergic tone during drug-induced neuronal activation would dampen subsequent rebound sleep in zebrafish larvae. During stimulant drug treatment, dampening noradrenergic tone with the α2-adrenoceptor agonist clonidine unexpectedly enhanced subsequent rebound sleep, whereas enhancing noradrenergic signalling with a cocktail of α1- and β-adrenoceptor agonists did not enhance rebound sleep. Similarly, CRISPR/Cas9-mediated elimination of the dopamine β-hydroxylase (dbh) gene, which encodes an enzyme required for noradrenalin synthesis, enhanced baseline sleep in larvae but did not prevent additional rebound sleep following acute induction of neuronal activity. Across all drug conditions, c-fos expression immediately after drug exposure correlated strongly with the amount of induced rebound sleep, but was inversely related to the strength of noradrenergic modulatory tone. These results are consistent with a model in which increases in neuronal activity, as reflected by brain-wide levels of c-fos induction, drive a sleep pressure signal that promotes rebound sleep independently of noradrenergic tone.
Collapse
Affiliation(s)
- Eleanor Benoit
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Declan G Lyons
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
3
|
Tan T, Jiang L, He Z, Ding X, Xiong X, Tang M, Chen Y, Tang Y. NR1 Splicing Variant NR1a in Cerebellar Granule Neurons Constitutes a Better Motor Learning in the Mouse. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1112-1120. [PMID: 37880519 PMCID: PMC11102416 DOI: 10.1007/s12311-023-01614-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 10/27/2023]
Abstract
As an excitatory neuron in the cerebellum, the granule cells play a crucial role in motor learning. The assembly of NMDAR in these neurons varies in developmental stages, while the significance of this variety is still not clear. In this study, we found that motor training could specially upregulate the expression level of NR1a, a splicing form of NR1 subunit. Interestingly, overexpression of this splicing variant in a cerebellar granule cell-specific manner dramatically elevated the NMDAR binding activity. Furthermore, the NR1a transgenic mice did not only show an enhanced motor learning, but also exhibit a higher efficacy for motor training in motor learning. Our results suggested that as a "junior" receptor, NR1a facilitates NMDAR activity as well as motor skill learning.
Collapse
Affiliation(s)
- Ting Tan
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Linyan Jiang
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Zhengxiao He
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xuejiao Ding
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoli Xiong
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Mingxi Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yuan Chen
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Yaping Tang
- Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
4
|
Eom K, Jung J, Kim B, Hyun JH. Molecular tools for recording and intervention of neuronal activity. Mol Cells 2024; 47:100048. [PMID: 38521352 PMCID: PMC11021360 DOI: 10.1016/j.mocell.2024.100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024] Open
Abstract
Observing the activity of neural networks is critical for the identification of learning and memory processes, as well as abnormal activities of neural circuits in disease, particularly for the purpose of tracking disease progression. Methodologies for describing the activity history of neural networks using molecular biology techniques first utilized genes expressed by active neurons, followed by the application of recently developed techniques including optogenetics and incorporation of insights garnered from other disciplines, including chemistry and physics. In this review, we will discuss ways in which molecular biological techniques used to describe the activity of neural networks have evolved along with the potential for future development.
Collapse
Affiliation(s)
- Kisang Eom
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jinhwan Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Byungsoo Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
5
|
Hosseindoost S, Inanloo SH, Pestehei SK, Rahimi M, Yekta RA, Khajehnasiri A, Rad MA, Majedi H, Dehpour AR. Cellular and molecular mechanisms involved in the analgesic effects of botulinum neurotoxin: A literature review. Drug Dev Res 2024; 85:e22177. [PMID: 38528637 DOI: 10.1002/ddr.22177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/02/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024]
Abstract
Botulinum neurotoxins (BoNTs), derived from Clostridium botulinum, have been employed to treat a range of central and peripheral neurological disease. Some studies indicate that BoNT may be beneficial for pain conditions as well. It has been hypothesized that BoNTs may exert their analgesic effects by preventing the release of pain-related neurotransmitters and neuroinflammatory agents from sensory nerve endings, suppressing glial activation, and inhibiting the transmission of pain-related receptors to the neuronal cell membrane. In addition, there is evidence to suggest that the central analgesic effects of BoNTs are mediated through their retrograde axonal transport. The purpose of this review is to summarize the experimental evidence of the analgesic functions of BoNTs and discuss the cellular and molecular mechanisms by which they can act on pain conditions. Most of the studies reviewed in this article were conducted using BoNT/A. The PubMed database was searched from 1995 to December 2022 to identify relevant literature.
Collapse
Affiliation(s)
- Saereh Hosseindoost
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Inanloo
- Department of Urology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Khalil Pestehei
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Rahimi
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Atef Yekta
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anesthesiology, Critical Care, and Pain, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khajehnasiri
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anesthesiology, Critical Care, and Pain, Dr. Ali Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Majedi
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Deska-Gauthier D, Borowska-Fielding J, Jones C, Zhang H, MacKay CS, Michail R, Bennett LA, Bikoff JB, Zhang Y. Embryonic temporal-spatial delineation of excitatory spinal V3 interneuron diversity. Cell Rep 2024; 43:113635. [PMID: 38160393 PMCID: PMC10877927 DOI: 10.1016/j.celrep.2023.113635] [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: 05/09/2023] [Revised: 10/24/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
Spinal neural circuits that execute movement are composed of cardinal classes of neurons that emerged from distinct progenitor lineages. Each cardinal class contains multiple neuronal subtypes characterized by distinct molecular, anatomical, and physiological characteristics. Through a focus on the excitatory V3 interneuron class, here we demonstrate that interneuron subtype diversity is delineated through a combination of neurogenesis timing and final laminar settling position. We have revealed that early-born and late-born embryonic V3 temporal classes further diversify into subclasses with spatially and molecularly discrete identities. While neurogenesis timing accounts for V3 morphological diversification, laminar settling position accounts for electrophysiological profiles distinguishing V3 subtypes within the same temporal classes. Furthermore, V3 interneuron subtypes display independent behavioral recruitment patterns demonstrating a functional modularity underlying V3 interneuron diversity. These studies provide a framework for how early embryonic temporal and spatial mechanisms combine to delineate spinal interneuron classes into molecularly, anatomically, and functionally relevant subtypes in adults.
Collapse
Affiliation(s)
- Dylan Deska-Gauthier
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Joanna Borowska-Fielding
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Chris Jones
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Han Zhang
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Colin S MacKay
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Ramez Michail
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Laura A Bennett
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ying Zhang
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
7
|
Lai TT, Tsai YH, Liou CW, Fan CH, Hou YT, Yao TH, Chuang HL, Wu WL. The gut microbiota modulate locomotion via vagus-dependent glucagon-like peptide-1 signaling. NPJ Biofilms Microbiomes 2024; 10:2. [PMID: 38228675 DOI: 10.1038/s41522-024-00477-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
Locomotor activity is an innate behavior that can be triggered by gut-motivated conditions, such as appetite and metabolic condition. Various nutrient-sensing receptors distributed in the vagal terminal in the gut are crucial for signal transduction from the gut to the brain. The levels of gut hormones are closely associated with the colonization status of the gut microbiota, suggesting a complicated interaction among gut bacteria, gut hormones, and the brain. However, the detailed mechanism underlying gut microbiota-mediated endocrine signaling in the modulation of locomotion is still unclear. Herein, we show that broad-spectrum antibiotic cocktail (ABX)-treated mice displayed hypolocomotion and elevated levels of the gut hormone glucagon-like peptide-1 (GLP-1). Blockade of the GLP-1 receptor and subdiaphragmatic vagal transmission rescued the deficient locomotor phenotype in ABX-treated mice. Activation of the GLP-1 receptor and vagal projecting brain regions led to hypolocomotion. Finally, selective antibiotic treatment dramatically increased serum GLP-1 levels and decreased locomotion. Colonizing Lactobacillus reuteri and Bacteroides thetaiotaomicron in microbiota-deficient mice suppressed GLP-1 levels and restored the hypolocomotor phenotype. Our findings identify a mechanism by which specific gut microbes mediate host motor behavior via the enteroendocrine and vagal-dependent neural pathways.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Tian Hou
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Wei-Li Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
8
|
Fergany A, Zong C, Ekuban FA, Wu B, Ueha S, Shichino S, Matsushima K, Iwakura Y, Ichihara S, Ichihara G. Transcriptome analysis of the cerebral cortex of acrylamide-exposed wild-type and IL-1β-knockout mice. Arch Toxicol 2024; 98:181-205. [PMID: 37971544 PMCID: PMC10761544 DOI: 10.1007/s00204-023-03627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Acrylamide is an environmental electrophile that has been produced in large amounts for many years. There is concern about the adverse health effects of acrylamide exposure due to its widespread industrial use and also presence in commonly consumed foods and others. IL-1β is a key cytokine that protects the brain from inflammatory insults, but its role in acrylamide-induced neurotoxicity remains unknown. We reported recently that deletion of IL-1β gene exacerbates ACR-induced neurotoxicity in mice. The aim of this study was to identify genes or signaling pathway(s) involved in enhancement of ACR-induced neurotoxicity by IL-1β gene deletion or ACR-induced neurotoxicity to generate a hypothesis mechanism explaining ACR-induced neurotoxicity. C57BL/6 J wild-type and IL-1β KO mice were exposed to ACR at 0, 12.5, 25 mg/kg by oral gavage for 7 days/week for 4 weeks, followed by extraction of mRNA from mice cerebral cortex for RNA sequence analysis. IL-1β deletion altered the expression of genes involved in extracellular region, including upregulation of PFN1 gene related to amyotrophic lateral sclerosis and increased the expression of the opposite strand of IL-1β. Acrylamide exposure enhanced mitochondria oxidative phosphorylation, synapse and ribosome pathways, and activated various pathways of different neurodegenerative diseases, such as Alzheimer disease, Parkinson disease, Huntington disease, and prion disease. Protein network analysis suggested the involvement of different proteins in related to learning and cognitive function, such as Egr1, Egr2, Fos, Nr4a1, and Btg2. Our results identified possible pathways involved in IL-1β deletion-potentiated and ACR-induced neurotoxicity in mice.
Collapse
Affiliation(s)
- Alzahraa Fergany
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
- Laboratory of Genetics and Genetic Engineering in Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Bin Wu
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
9
|
Zhou J, Khateeb K, Yazdan-Shahmorad A. Early Intervention with Electrical Stimulation Reduces Neural Damage After Stroke in Non-human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572235. [PMID: 38187669 PMCID: PMC10769281 DOI: 10.1101/2023.12.18.572235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Ischemic stroke is a neurological condition that results in significant mortality and long-term disability for adults, creating huge health burdens worldwide. For stroke patients, acute intervention offers the most critical therapeutic opportunity as it can reduce irreversible tissue injury and improve functional outcomes. However, currently available treatments within the acute window are highly limited. Although emerging neuromodulation therapies have been tested for chronic stroke patients, acute stimulation is rarely studied due to the risk of causing adverse effects related to ischemia-induced electrical instability. To address this gap, we combined electrophysiology and histology tools to investigate the effects of acute electrical stimulation on ischemic neural damage in non-human primates. Specifically, we induced photothrombotic lesions in the monkey sensorimotor cortex while collecting electrocorticography (ECoG) signals through a customized neural interface. Gamma activity in ECoG was used as an electrophysiological marker to track the effects of stimulation on neural activation. Meanwhile, histological analysis including Nissl, cFos, and microglial staining was performed to evaluate the tissue response to ischemic injury. Comparing stimulated monkeys to controls, we found that theta-burst stimulation administered directly adjacent to the ischemic infarct at 1 hour post-stroke briefly inhibits peri-infarct neuronal activation as reflected by decreased ECoG gamma power and cFos expression. Meanwhile, lower microglial activation and smaller lesion volumes were observed in animals receiving post-stroke stimulation. Together, these results suggest that acute electrical stimulation can be used safely and effectively as an early stroke intervention to reduce excitotoxicity and inflammation, thus mitigating neural damage and enhancing stroke outcomes.
Collapse
Affiliation(s)
- Jasmine Zhou
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
| | - Karam Khateeb
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
| | - Azadeh Yazdan-Shahmorad
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, 98195
| |
Collapse
|
10
|
Chitolina R, Gallas-Lopes M, Reis CG, Benvenutti R, Stahlhofer-Buss T, Calcagnotto ME, Herrmann AP, Piato A. Chemically-induced epileptic seizures in zebrafish: A systematic review. Epilepsy Res 2023; 197:107236. [PMID: 37801749 DOI: 10.1016/j.eplepsyres.2023.107236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
The use of zebrafish as a model organism is gaining evidence in the field of epilepsy as it may help to understand the mechanisms underlying epileptic seizures. As zebrafish assays became popular, the heterogeneity between protocols increased, making it hard to choose a standard protocol to conduct research while also impairing the comparison of results between studies. We conducted a systematic review to comprehensively profile the chemically-induced seizure models in zebrafish. Literature searches were performed in PubMed, Scopus, and Web of Science, followed by a two-step screening process based on inclusion/exclusion criteria. Qualitative data were extracted, and a sample of 100 studies was randomly selected for risk of bias assessment. Out of the 1058 studies identified after removing duplicates, 201 met the inclusion criteria. We found that the most common chemoconvulsants used in the reviewed studies were pentylenetetrazole (n = 180), kainic acid (n = 11), and pilocarpine (n = 10), which increase seizure severity in a dose-dependent manner. The main outcomes assessed were seizure scores and locomotion. Significant variability between the protocols was observed for administration route, duration of exposure, and dose/concentration. Of the studies subjected to risk of bias assessment, most were rated as low risk of bias for selective reporting (94%), baseline characteristics of the animals (67%), and blinded outcome assessment (54%). Randomization procedures and incomplete data were rated unclear in 81% and 68% of the studies, respectively. None of the studies reported the sample size calculation. Overall, these findings underscore the need for improved methodological and reporting practices to enhance the reproducibility and reliability of zebrafish models for studying epilepsy. Our study offers a comprehensive overview of the current state of chemically-induced seizure models in zebrafish, highlighting the common chemoconvulsants used and the variability in protocol parameters. This may be particularly valuable to researchers interested in understanding the underlying mechanisms of epileptic seizures and screening potential drug candidates in zebrafish models.
Collapse
Affiliation(s)
- Rafael Chitolina
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Matheus Gallas-Lopes
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and meta-Analysis (BRISA) Collaboration, Brazil; Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos G Reis
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Thailana Stahlhofer-Buss
- Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Neurobiologia e Neuroquímica da Excitabilidade Neuronal e Plasticidade Sináptica (NNNESP Lab), Departamento de bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Ana P Herrmann
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and meta-Analysis (BRISA) Collaboration, Brazil; Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angelo Piato
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
11
|
Manning A, Han V, Stephens A, Wang R, Bush N, Bard M, Ramirez JM, Kalume F. Elevated susceptibility to exogenous seizure triggers and impaired interneuron excitability in a mouse model of Leigh syndrome epilepsy. Neurobiol Dis 2023; 187:106288. [PMID: 37704057 PMCID: PMC10621616 DOI: 10.1016/j.nbd.2023.106288] [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: 05/28/2023] [Revised: 08/12/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023] Open
Abstract
Mutations in the NADH dehydrogenase (ubiquinone reductase) iron‑sulfur protein 4 (NDUFS4) gene, which encodes for a key structural subunit of the OXFOS complex I (CI), lead to the most common form of mitochondrial disease in children known as Leigh syndrome (LS). As in other mitochondrial diseases, epileptic seizures constitute one of the most significant clinical features of LS. These seizures are often very difficult to treat and are a sign of poor disease prognosis. Mice with whole-body Ndufs4 KO are a well-validated model of LS; they exhibit epilepsy and several other clinical features of LS. We have previously shown that mice with Ndufs4 KO in only GABAergic interneurons (Gad2-Ndufs4-KO) reproduce the severe epilepsy phenotype observed in the global KO mice. This observation indicated that these mice represent an excellent model of LS epilepsy isolated from other clinical manifestations of the disease. To further characterize this epilepsy phenotype, we investigated seizure susceptibility to selected exogenous seizure triggers in Gad2-Ndufs4-KO mice. Then, using electrophysiology, imaging, and immunohistochemistry, we studied the cellular, physiological, and neuroanatomical consequences of Ndufs4 KO in GABAergic interneurons. Homozygous KO of Ndufs4 in GABAergic interneurons leads to a prominent susceptibility to exogenous seizure triggers, impaired interneuron excitability and interneuron loss. Finally, we found that the hippocampus and cortex participate in the generation of seizure activity in Gad2-Ndufs4-KO mice. These findings further define the LS epilepsy phenotype and provide important insights into the cellular mechanisms underlying epilepsy in LS and other mitochondrial diseases.
Collapse
Affiliation(s)
- Arena Manning
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Victor Han
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Alexa Stephens
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Rose Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Nicholas Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Michelle Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Jan M Ramirez
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America
| | - Franck Kalume
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
12
|
Mosini A, Mazzonetto P, Calió M, Pompeu C, Massinhani F, Nakamura T, Pires J, Silva C, Porcionatto M, Mello L. Temporal pattern of Fos and Jun families expression after mitogenic stimulation with FGF-2 in rat neural stem cells and fibroblasts. Braz J Med Biol Res 2023; 56:e12546. [PMID: 37703106 PMCID: PMC10496756 DOI: 10.1590/1414-431x2023e12546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
Intense stimulation of most living cells triggers the activation of immediate early genes, such as Fos and Jun families. These genes are important in cellular and biochemical processes, such as mitosis and cell death. The present study focused on determining the temporal expression pattern of Fos and Jun families in fibroblasts and neural stem cells of cerebellum, hippocampus, and subventricular zone (SVZ) of rats of different ages at 0, 0.5, 1, 3, and 6 h after stimulation with fibroblast growth factor (FGF)-2. In neonates, a similar expression pattern was observed in all cells analyzed, with lower expression in basal condition, peak expression at 0.5 h after stimulation, returning to baseline values between 1 and 3 h after stimulation. On the other hand, cells from adult animals only showed Fra1 and JunD expression after stimulation. In fibroblasts and hippocampus, Fra1 reached peak expression at 0.5 h after stimulation, while in the SVZ, peak level was observed at 6 h after stimulation. JunD in fibroblasts presented two peak expressions, at 0.5 and 6 h after stimulation. Between these periods, the expression observed was at a basal level. Nevertheless, JunD expression in SVZ and hippocampus was low and without significant changes after stimulation. Differences in mRNA expression in neonate and adult animals characterize the significant differences in neurogenesis and cell response to stimulation at different stages of development. Characterizing these differences might be important for the development of cell cultures, replacement therapy, and the understanding of the physiological response profile of different cell types.
Collapse
Affiliation(s)
- A.C. Mosini
- Laboratório de Neurobiologia, Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - P.C. Mazzonetto
- Laboratório de Neurobiologia, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M.L. Calió
- Laboratório de Neurobiologia, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - C. Pompeu
- Laboratório de Neurobiologia, Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - F.H. Massinhani
- Laboratório de Neurobiologia, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - T.K.E. Nakamura
- Laboratório de Neurobiologia, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - J.M. Pires
- Laboratório de Neurobiologia, Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - C.S. Silva
- Laboratório de Neurobiologia, Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M.A. Porcionatto
- Laboratório de Neurobiologia, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L.E. Mello
- Laboratório de Neurobiologia, Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
- Instituto D’Or de Pesquisa e Ensino, São Paulo, SP, Brasil
| |
Collapse
|
13
|
Frey T, Murakami T, Maki K, Kawaue T, Tani N, Sugai A, Nakazawa N, Ishiguro K, Adachi T, Kengaku M, Ohki K, Gotoh Y, Kishi Y. Age-associated reduction of nuclear shape dynamics in excitatory neurons of the visual cortex. Aging Cell 2023; 22:e13925. [PMID: 37476844 PMCID: PMC10497821 DOI: 10.1111/acel.13925] [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: 01/10/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Neurons decline in their functionality over time, and age-related neuronal alterations are associated with phenotypes of neurodegenerative diseases. In nonneural tissues, an infolded nuclear shape has been proposed as a hallmark of aged cells and neurons with infolded nuclei have also been reported to be associated with neuronal activity. Here, we performed time-lapse imaging in the visual cortex of Nex-Cre;SUN1-GFP mice. Nuclear infolding was observed within 10 min of stimulation in young nuclei, while the aged nuclei were already infolded pre-stimulation and showed reduced dynamics of the morphology. In young nuclei, the depletion of the stimuli restored the nucleus to a spherical shape and reduced the dynamic behavior, suggesting that nuclear infolding is a reversible process. We also found the aged nucleus to be stiffer than the young one, further relating to the age-associated loss of nuclear shape dynamics. We reveal temporal changes in the nuclear shape upon external stimulation and observe that these morphological dynamics decrease with age.
Collapse
Affiliation(s)
- Tanita Frey
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Tomonari Murakami
- Graduate School of MedicineThe University of TokyoTokyoJapan
- Institute for AI and Beyond, The University of TokyoTokyoJapan
| | - Koichiro Maki
- Institute for Life and Medical Sciences, Kyoto UniversityKyotoJapan
| | - Takumi Kawaue
- Institute for Integrated Cell‐Material Sciences, Institute for Advanced Study, Kyoto UniversityKyotoJapan
| | - Naoki Tani
- Liaison Laboratory Research Promotion CenterIMEG, Kumamoto UniversityKumamotoJapan
| | - Ayaka Sugai
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- Institute for Quantitative Biosciences, The University of TokyoTokyoJapan
| | - Naotaka Nakazawa
- Institute for Integrated Cell‐Material Sciences, Institute for Advanced Study, Kyoto UniversityKyotoJapan
- Department of Energy and Materials, Faculty of Science and EngineeringKindai UniversityOsakaJapan
| | - Kei‐ichiro Ishiguro
- Department of Chromosome BiologyInstitute of Molecular Embryology and Genetics (IMEG), Kumamoto UniversityKumamotoJapan
| | - Taiji Adachi
- Institute for Life and Medical Sciences, Kyoto UniversityKyotoJapan
| | - Mineko Kengaku
- Institute for Integrated Cell‐Material Sciences, Institute for Advanced Study, Kyoto UniversityKyotoJapan
| | - Kenichi Ohki
- Graduate School of MedicineThe University of TokyoTokyoJapan
- Institute for AI and Beyond, The University of TokyoTokyoJapan
- International Research Center for Neurointelligence (WPI‐IRCN), The University of TokyoTokyoJapan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- International Research Center for Neurointelligence (WPI‐IRCN), The University of TokyoTokyoJapan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- Institute for Quantitative Biosciences, The University of TokyoTokyoJapan
| |
Collapse
|
14
|
Garcia-Keller C, Hohmeister M, Seidling K, Beloate L, Chioma V, Spencer S, Kalivas P, Neuhofer D. Δ 9 -Tetrahydrocannabinol self-administration induces cell type-specific adaptations in the nucleus accumbens core. Addict Biol 2023; 28:e13286. [PMID: 37500492 PMCID: PMC10924663 DOI: 10.1111/adb.13286] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 07/29/2023]
Abstract
Drugs of abuse induce cell type-specific adaptations in D1- and D2-medium spiny neurons (MSNs) in the nucleus accumbens core (NAcore) that can bias signalling towards D1-MSNs and enhance relapse vulnerability. Whether Δ9 -tetrahydrocannabinol (THC) use initiates similar neuroadaptations is unknown. D1- and D2-Cre transgenic rats were transfected with Cre-dependent reporters and trained to self-administer THC + cannabidiol (THC + CBD). After extinction training spine morphology, glutamate transmission, CB1R function and cFOS expression were quantified. We found that extinction from THC + CBD induced a loss of large spine heads in D1- but not D2-MSNs and commensurate reductions in glutamate synaptic transmission. Also, presynaptic CB1R function was impaired selectively at glutamatergic synapses on D1-MSNs, which augmented the capacity to potentiate glutamate transmission. Using cFOS expression as an activity marker, we found no change after extinction but increased cFOS expression in D1-MSNs after cue-induced drug seeking. Contrasting D1-MSNs, CB1R function and glutamate synaptic transmission on D2-MSN synapses were unaffected by THC + CBD use. However, cFOS expression was decreased in D2-MSNs of THC + CBD-extinguished rats and was restored after drug seeking. Thus, CB1R adaptations in D1-MSNs partially predicted neuronal activity changes, posing pathway specific modulation of eCB signalling in D1-MSNs as a potential treatment avenue for cannabis use disorder (CUD).
Collapse
Affiliation(s)
- Constanza Garcia-Keller
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226
| | - Madeline Hohmeister
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Kailyn Seidling
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Lauren Beloate
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Vivian Chioma
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Sade Spencer
- Department of Pharmacology, Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, MN 55455
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| | - Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC 29425
| |
Collapse
|
15
|
Veremeyko T, Jiang R, He M, Ponomarev ED. Complement C4-deficient mice have a high mortality rate during PTZ-induced epileptic seizures, which correlates with cognitive problems and the deficiency in the expression of Egr1 and other immediate early genes. Front Cell Neurosci 2023; 17:1170031. [PMID: 37234916 PMCID: PMC10206007 DOI: 10.3389/fncel.2023.1170031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Complement system plays an important role in the immune defense against pathogens; however, recent studies demonstrated an important role of complement subunits C1q, C4, and C3 in normal functions of the central nervous system (CNS) such as non-functional synapse elimination (synapse pruning), and during various neurologic pathologies. Humans have two forms of C4 protein encoded by C4A and C4B genes that share 99.5% homology, while mice have only one C4B gene that is functionally active in the complement cascade. Overexpression of the human C4A gene was shown to contribute to the development of schizophrenia by mediating extensive synapse pruning through the activation C1q-C4-C3 pathway, while C4B deficiency or low levels of C4B expression were shown to relate to the development of schizophrenia and autism spectrum disorders possibly via other mechanisms not related to synapse elimination. To investigate the potential role of C4B in neuronal functions not related to synapse pruning, we compared wildtype (WT) mice with C3- and C4B- deficient animals for their susceptibility to pentylenetetrazole (PTZ)- induced epileptic seizures. We found that C4B (but not C3)-deficient mice were highly susceptible to convulsant and subconvulsant doses of PTZ when compared to WT controls. Further gene expression analysis revealed that in contrast to WT or C3-deficient animals, C4B-deficient mice failed to upregulate expressions of multiple immediate early genes (IEGs) Egrs1-4, c-Fos, c-Jus, FosB, Npas4, and Nur77 during epileptic seizures. Moreover, C4B-deficient mice had low levels of baseline expression of Egr1 on mRNA and protein levels, which was correlated with the cognitive problems of these animals. C4-deficient animals also failed to upregulate several genes downstream of IEGs such as BDNF and pro-inflammatory cytokines IL-1β, IL-6, and TNF. Taken together, our study demonstrates a new role of C4B in the regulation of expression of IEGs and their downstream targets during CNS insults such as epileptic seizures.
Collapse
Affiliation(s)
- Tatyana Veremeyko
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
- Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Mingliang He
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Eugene D. Ponomarev
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
- Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
16
|
Oyaga MR, Serra I, Kurup D, Koekkoek SKE, Badura A. Delay eyeblink conditioning performance and brain-wide c-Fos expression in male and female mice. Open Biol 2023; 13:220121. [PMID: 37161289 PMCID: PMC10170203 DOI: 10.1098/rsob.220121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Delay eyeblink conditioning has been extensively used to study associative learning and the cerebellar circuits underlying this task have been largely identified. However, there is a little knowledge on how factors such as strain, sex and innate behaviour influence performance during this type of learning. In this study, we used male and female mice of C57BL/6J (B6) and B6CBAF1 strains to investigate the effect of sex, strain and locomotion in delay eyeblink conditioning. We performed a short and a long delay eyeblink conditioning paradigm and used a c-Fos immunostaining approach to explore the involvement of different brain areas in this task. We found that both B6 and B6CBAF1 females reach higher learning scores compared to males in the initial stages of learning. This sex-dependent difference was no longer present as the learning progressed. Moreover, we found a strong positive correlation between learning scores and voluntary locomotion irrespective of the training duration. c-Fos immunostainings after the short paradigm showed positive correlations between c-Fos expression and learning scores in the cerebellar cortex and brainstem, as well as previously unreported areas. By contrast, after the long paradigm, c-Fos expression was only significantly elevated in the brainstem. Taken together, we show that differences in voluntary locomotion and activity across brain areas correlate with performance in delay eyeblink conditioning across strains and sexes.
Collapse
Affiliation(s)
- Maria Roa Oyaga
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | - Ines Serra
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | - Devika Kurup
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
| | | | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC, 3000 Rotterdam, the Netherlands
- Netherlands Institute of Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam 1105 BA, the Netherlands
| |
Collapse
|
17
|
Gudenschwager-Basso EK, Shandra O, Volanth T, Patel DC, Kelly C, Browning JL, Wei X, Harris EA, Mahmutovic D, Kaloss AM, Correa FG, Decker J, Maharathi B, Robel S, Sontheimer H, VandeVord PJ, Olsen ML, Theus MH. Atypical Neurogenesis, Astrogliosis, and Excessive Hilar Interneuron Loss Are Associated with the Development of Post-Traumatic Epilepsy. Cells 2023; 12:1248. [PMID: 37174647 PMCID: PMC10177146 DOI: 10.3390/cells12091248] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/02/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) remains a significant risk factor for post-traumatic epilepsy (PTE). The pathophysiological mechanisms underlying the injury-induced epileptogenesis are under investigation. The dentate gyrus-a structure that is highly susceptible to injury-has been implicated in the evolution of seizure development. METHODS Utilizing the murine unilateral focal control cortical impact (CCI) injury, we evaluated seizure onset using 24/7 EEG video analysis at 2-4 months post-injury. Cellular changes in the dentate gyrus and hilus of the hippocampus were quantified by unbiased stereology and Imaris image analysis to evaluate Prox1-positive cell migration, astrocyte branching, and morphology, as well as neuronal loss at four months post-injury. Isolation of region-specific astrocytes and RNA-Seq were performed to determine differential gene expression in animals that developed post-traumatic epilepsy (PTE+) vs. those animals that did not (PTE-), which may be associated with epileptogenesis. RESULTS CCI injury resulted in 37% PTE incidence, which increased with injury severity and hippocampal damage. Histological assessments uncovered a significant loss of hilar interneurons that coincided with aberrant migration of Prox1-positive granule cells and reduced astroglial branching in PTE+ compared to PTE- mice. We uniquely identified Cst3 as a PTE+-specific gene signature in astrocytes across all brain regions, which showed increased astroglial expression in the PTE+ hilus. CONCLUSIONS These findings suggest that epileptogenesis may emerge following TBI due to distinct aberrant cellular remodeling events and key molecular changes in the dentate gyrus of the hippocampus.
Collapse
Affiliation(s)
| | - Oleksii Shandra
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA
| | - Troy Volanth
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061, USA
| | - Dipan C. Patel
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061, USA
| | - Colin Kelly
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Jack L. Browning
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xiaoran Wei
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA (E.A.H.)
| | - Elizabeth A. Harris
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA (E.A.H.)
| | - Dzenis Mahmutovic
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Alexandra M. Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA (E.A.H.)
| | | | - Jeremy Decker
- Department of Biomedical Engineering and Mechanics, Blacksburg, VA 24061, USA
| | - Biswajit Maharathi
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefanie Robel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - Pamela J. VandeVord
- Department of Biomedical Engineering and Mechanics, Blacksburg, VA 24061, USA
| | | | - Michelle H. Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA (E.A.H.)
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Engineered Health, Viginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
18
|
From Determining Brain Insulin Resistance in a Sporadic Alzheimer's Disease Model to Exploring the Region-Dependent Effect of Intranasal Insulin. Mol Neurobiol 2023; 60:2005-2023. [PMID: 36596966 DOI: 10.1007/s12035-022-03188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 12/25/2022] [Indexed: 01/05/2023]
Abstract
Impaired response to insulin has been linked to many neurodegenerative disorders like Alzheimer's disease (AD). Animal model of sporadic AD has been developed by intracerebroventricular (icv) administration of streptozotocin (STZ), which given peripherally causes insulin resistance. Difficulty in demonstrating insulin resistance in this model led to our aim: to determine brain regional and peripheral response after intranasal (IN) administration of insulin in control and STZ-icv rats, by exploring peripheral and central metabolic parameters. One month after STZ-icv or vehicle-icv administration to 3-month-old male Wistar rats, cognitive status was determined after which rats received 2 IU of fast-acting insulin aspart intranasally (CTR + INS; STZ + INS) or saline only (CTR and STZ). Rats were sacrificed 2 h after administration and metabolic and glutamatergic parameters were measured in plasma, CSF, and the brain. Insulin and STZ increased amyloid-β concentration in plasma (CTR + INS and STZ vs CTR), while there was no effect on glucose and insulin plasma and CSF levels. INS normalized the levels of c-fos in temporal cortex of STZ + INS vs STZ (co-localized with neurons), while hypothalamic c-fos was found co-localized with the microglial marker. STZ and insulin brain region specifically altered the levels and activity of proteins involved in cell metabolism and glutamate signaling. Central changes found after INS in STZ-icv rats suggest hippocampal and cortical insulin sensitivity. Altered hypothalamic metabolic parameters of STZ-icv rats were not normalized by INS, indicating possible hypothalamic insulin insensitivity. Brain insulin sensitivity depends on the affected brain region and presence of metabolic dysfunction induced by STZ-icv administration.
Collapse
|
19
|
Calderazzo S, Covert M, Alba DD, Bowley BE, Pessina MA, Rosene DL, Buller B, Medalla M, Moore TL. Neural recovery after cortical injury: Effects of MSC derived extracellular vesicles on motor circuit remodeling in rhesus monkeys. IBRO Neurosci Rep 2022; 13:243-254. [PMID: 36590089 PMCID: PMC9795302 DOI: 10.1016/j.ibneur.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 01/04/2023] Open
Abstract
Reorganization of motor circuits in the cortex and corticospinal tract are thought to underlie functional recovery after cortical injury, but the mechanisms of neural plasticity that could be therapeutic targets remain unclear. Recent work from our group have shown that systemic treatment with mesenchymal stem cell derived (MSCd) extracellular vesicles (EVs) administered after cortical damage to the primary motor cortex (M1) of rhesus monkeys resulted in a robust recovery of fine motor function and reduced chronic inflammation. Here, we used immunohistochemistry for cfos, an activity-dependent intermediate early gene, to label task-related neurons in the surviving primary motor and premotor cortices, and markers of axonal and synaptic plasticity in the spinal cord. Compared to vehicle, EV treatment was associated with a greater density of cfos+ pyramidal neurons in the deep layers of M1, greater density of cfos+ inhibitory interneurons in premotor areas, and lower density of synapses on MAP2+ lower motor neurons in the cervical spinal cord. These data suggest that the anti-inflammatory effects of EVs may reduce injury-related upper motor neuron damage and hyperexcitability, as well as aberrant compensatory re-organization in the cervical spinal cord to improve motor function.
Collapse
Key Words
- CB, Calbindin
- CR, Calretinin
- CSC, Cervical Spinal Cord
- Circuit Remodeling
- Cortical Injury
- DH, Dorsal Horn
- EVs, Extracellular Vesicles
- Extracellular Vesicles
- Ischemia
- LCST, Lateral Corticospinal Tract
- M1, Primary Motor Cortex
- MAP2, Microtubule Associated Protein 2
- MSCd, Mesenchymal Stem Cell derived
- Motor Cortex
- NHP, Non-Human Primate
- PV, Parvalbumin
- Plasticity
- ROS, Reactive Oxygen Species
- SYN, Synaptophysin
- Stem Cell-Based Treatments
- VH, Ventral Horn
- dPMC, dorsal Premotor Cortex
- miRNA, Micro RNA
- periM1, Perilesional Primary Motor Cortex
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas L. Rosene
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | | | - Maria Medalla
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | - Tara L. Moore
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| |
Collapse
|
20
|
Deep brain stimulation in animal models of dystonia. Neurobiol Dis 2022; 175:105912. [DOI: 10.1016/j.nbd.2022.105912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
|
21
|
Seizurogenic effect of perfluorooctane sulfonate in zebrafish larvae. Neurotoxicology 2022; 93:257-264. [DOI: 10.1016/j.neuro.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
|
22
|
Lara Aparicio SY, Laureani Fierro ÁDJ, Aranda Abreu GE, Toledo Cárdenas R, García Hernández LI, Coria Ávila GA, Rojas Durán F, Aguilar MEH, Manzo Denes J, Chi-Castañeda LD, Pérez Estudillo CA. Current Opinion on the Use of c-Fos in Neuroscience. NEUROSCI 2022; 3:687-702. [PMID: 39483772 PMCID: PMC11523728 DOI: 10.3390/neurosci3040050] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 11/03/2024] Open
Abstract
For years, the biochemical processes that are triggered by harmful and non-harmful stimuli at the central nervous system level have been extensively studied by the scientific community through numerous techniques and animal models. For example, one of these techniques is the use of immediate expression genes, which is a useful, accessible, and reliable method for observing and quantifying cell activation. It has been shown that both the c-fos gene and its protein c-Fos have rapid activation after stimulus, with the length of time that they remain active depending on the type of stimulus and the activation time depending on the stimulus and the structure studied. Fos requires the participation of other genes (such as c-jun) for its expression (during hetero-dimer forming). c-Fos dimerizes with c-Jun protein to form factor AP-1, which promotes the transcription of various genes. The production and removal of c-Fos is part of cellular homeostasis, but its overexpression results in increased cell proliferation. Although Fos has been used as a marker of cellular activity since the 1990s, which molecular mechanism participates in the regulation of the expression of this protein is still unknown because the gene and the protein are not specific to neurons or glial cells. For these reasons, this work has the objective of gathering information about this protein and its use in neuroscience.
Collapse
Affiliation(s)
- Sandra Yasbeth Lara Aparicio
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
- Laboratorio de Neurofisiología, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Av. Luis Castelazo S/N, Col. Industrial Las Ánimas, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | | | | - Rebeca Toledo Cárdenas
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Luis Isauro García Hernández
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Genaro Alfonso Coria Ávila
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Fausto Rojas Durán
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | | - Jorge Manzo Denes
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Lizbeth Donají Chi-Castañeda
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | |
Collapse
|
23
|
Qiu Y, O’Neill N, Maffei B, Zourray C, Almacellas-Barbanoj A, Carpenter JC, Jones SP, Leite M, Turner TJ, Moreira FC, Snowball A, Shekh-Ahmad T, Magloire V, Barral S, Kurian MA, Walker MC, Schorge S, Kullmann DM, Lignani G. On-demand cell-autonomous gene therapy for brain circuit disorders. Science 2022; 378:523-532. [PMID: 36378958 PMCID: PMC7613996 DOI: 10.1126/science.abq6656] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several neurodevelopmental and neuropsychiatric disorders are characterized by intermittent episodes of pathological activity. Although genetic therapies offer the ability to modulate neuronal excitability, a limiting factor is that they do not discriminate between neurons involved in circuit pathologies and "healthy" surrounding or intermingled neurons. We describe a gene therapy strategy that down-regulates the excitability of overactive neurons in closed loop, which we tested in models of epilepsy. We used an immediate early gene promoter to drive the expression of Kv1.1 potassium channels specifically in hyperactive neurons, and only for as long as they exhibit abnormal activity. Neuronal excitability was reduced by seizure-related activity, leading to a persistent antiepileptic effect without interfering with normal behaviors. Activity-dependent gene therapy is a promising on-demand cell-autonomous treatment for brain circuit disorders.
Collapse
Affiliation(s)
- Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nathanael O’Neill
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Benito Maffei
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Clara Zourray
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
| | - Amanda Almacellas-Barbanoj
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jenna C. Carpenter
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Steffan P. Jones
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Marco Leite
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas J. Turner
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Francisco C. Moreira
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Tawfeeq Shekh-Ahmad
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Vincent Magloire
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Serena Barral
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
| | - Manju A. Kurian
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Matthew C. Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Stephanie Schorge
- Department of Neuroscience, Physiology and Pharmacology University College London, London, UK
| | - Dimitri M. Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
24
|
Hosseinzadeh Sahafi O, Rezayof A, Ghasemzadeh Z, Alijanpour S, Rahimian S. Ameliorating effect offluoxetine on tamoxifen-induced memory loss: The role of corticolimbic NMDA receptors and CREB/BDNF/cFos signaling pathways in rats. Brain Res 2022; 1794:148058. [PMID: 36007581 DOI: 10.1016/j.brainres.2022.148058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/02/2022]
Abstract
Tamoxifen-induced cognitive dysfunction may lead to fluoxetine consumption in patients with breast cancer. Since the brain mechanisms are unclear in tamoxifen/fluoxetine therapy, the blockade effect of hippocampal/amygdala/prefrontal cortical NMDA receptors was examined in fluoxetine/tamoxifen-induced memory retrieval. We also assessed the corticolimbic signaling pathways in memory retrieval under the drug treatment in adult male Wistar rats. Using the Western blot technique, the expression levels of the cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and cFos were evaluated in the corticolimbic regions. The results showed that pre-test administration of fluoxetine (3 and 5 mg/kg, i.p.) improved tamoxifen-induced memory impairment in the passive avoidance learning task. Pre-test bilateral microinjection of D-AP5, a selective NMDA receptor antagonist, into the dorsal hippocampal CA1 regions and the central amygdala (CeA), but not the medial prefrontal cortex (mPFC), inhibited the improving effect of fluoxetine on tamoxifen response. It is important to note that the microinjection of D-AP5 into the different sites by itself did not affect memory retrieval. Memory retrieval increased the signaling pathway of pCREB/CREB/BDNF/cFos in the corticolimbic regions. Tamoxifen-induced memory impairment decreased the hippocampal/PFC BDNF level and the amygdala level of pCREB/CREB/cFos. The improving effect of fluoxetine on tamoxifen significantly increased the hippocampal/PFC expression levels of BDNF, the PFC/amygdala expression levels of cFos, and the ratio of pCREB/CREB in all targeted areas. Thus, NMDA receptors' activity in the different corticolimbic regions mediates fluoxetine/tamoxifen memory retrieval. The corticolimbic synaptic plasticity changes likely accompany the improving effect of fluoxetine on tamoxifen response.
Collapse
Affiliation(s)
- Oveis Hosseinzadeh Sahafi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran
| | - Sepehrdad Rahimian
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
25
|
Hedley KE, Callister RJ, Callister R, Horvat JC, Tadros MA. Alterations in brainstem respiratory centers following peripheral inflammation: A systematic review. J Neuroimmunol 2022; 369:577903. [DOI: 10.1016/j.jneuroim.2022.577903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/02/2022] [Accepted: 05/29/2022] [Indexed: 11/29/2022]
|
26
|
Tsuchida R, Yamaguchi T, Funabashi D, Koumi Y, Kita I, Nishijima T. Exercise type influences the effect of an acute bout of exercise on hippocampal neuronal activation in mice. Neurosci Lett 2022; 783:136707. [PMID: 35660647 DOI: 10.1016/j.neulet.2022.136707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
The effects of exercise on the hippocampus depend on exercise conditions. Exercise intensity is thought to be a dominant factor that influences the effects of exercise on the hippocampus; however, it is uncertain whether the type of exercise influences its effectiveness. This study investigated whether the effect of an acute bout of exercise on hippocampal neuronal activation differs between two different types of exercise: treadmill and rotarod exercise. The intensities of both exercises were matched at just below the lactate threshold (LT), based on blood lactate concentration. Immunohistochemical examination of c-Fos, a marker of neuronal activation, revealed that treadmill exercise at 15 m/min (T15) significantly increased c-Fos expression in all subfields of the hippocampus (dentate gyrus DG, CA1, CA3), but rotarod exercise at 30 rpm (R30) did not, as compared with the respective control groups. These results demonstrate that moderate treadmill exercise more efficiently evokes hippocampal neuronal activation than does intensity-matched rotarod exercise. This suggests that exercise type is another important factor affecting the effects of exercise on the hippocampus.
Collapse
Affiliation(s)
- Ryuki Tsuchida
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Taisei Yamaguchi
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Daisuke Funabashi
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Yusuke Koumi
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Ichiro Kita
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Takeshi Nishijima
- Department of Human Health Sciences, Graduate School of Human Health Sciences Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan.
| |
Collapse
|
27
|
Whisker trimming induces anti-anxiety like status via activation of dorsomedial hypothalamus nucleus in mice. Brain Res 2022; 1789:147946. [PMID: 35597326 DOI: 10.1016/j.brainres.2022.147946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/27/2022] [Accepted: 05/15/2022] [Indexed: 11/20/2022]
Abstract
Whiskers are highly developed tactile organs in mice. Here, we showed that mice with whisker trimming had a decreased anxiety behavior and activation of dorsomedial hypothalamus compared to control mice. Inhibition or damage of dorsomedial hypothalamus reversed the decrease of anxiety level induced by whisker trimming. These results expand the role of whiskers in regulating mouse behaviors to anxiety and suggest a novel function of dorsomedial hypothalamus. These findings indicate importance of normal sensory functions in modulating animal behavior.
Collapse
|
28
|
Karantysh GV, Mendzheritsky AM, Prokofiev VN, Lyangasova OV, Fomenko MP. Expression of Genes Regulating Synaptic Plasticity in the Hippocampus and Spatial Learning in Rats of Different Age with Streptozotocin-Induced Diabetes. NEUROCHEM J+ 2022. [DOI: 10.1134/s181971242201007x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Ventral midline thalamus activation is correlated with memory performance in a delayed spatial matching-to-sample task: A c-Fos imaging approach in the rat. Behav Brain Res 2022; 418:113670. [PMID: 34798168 DOI: 10.1016/j.bbr.2021.113670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
The reuniens (Re) and rhomboid (Rh) nuclei of the ventral midline thalamus are bi-directionally connected with the hippocampus and the medial prefrontal cortex. They participate in a variety of cognitive functions, including information holding for seconds to minutes in working memory tasks. What about longer delays? To address this question, we used a spatial working memory task in which rats had to reach a platform submerged in water. The platform location was changed every 2-trial session and rats had to use allothetic cues to find it. Control rats received training in a typical response-memory task. We interposed a 6 h interval between instruction (locate platform) and evaluation (return to platform) trials in both tasks. After the last session, rats were killed for c-Fos imaging. A home-cage group was used as additional control of baseline levels of c-Fos expression. C-Fos expression was increased to comparable levels in the Re (not Rh) of both spatial memory and response-memory rats as compared to their home cage counterparts. However, in spatial memory rats, not in their response-memory controls, task performance was correlated with c-Fos expression in the Re: the higher this expression, the better the performance. Furthermore, we noticed an activation of hippocampal region CA1 and of the anteroventral nucleus of the rostral thalamus. This activation was specific to spatial memory. The data point to a possible performance-determinant participation of the Re nucleus in the delayed engagement of spatial information encoded in a temporary memory.
Collapse
|
30
|
Grossman YS, Fillinger C, Manganaro A, Voren G, Waldman R, Zou T, Janssen WG, Kenny PJ, Dumitriu D. Structure and function differences in the prelimbic cortex to basolateral amygdala circuit mediate trait vulnerability in a novel model of acute social defeat stress in male mice. Neuropsychopharmacology 2022; 47:788-799. [PMID: 34799681 PMCID: PMC8782864 DOI: 10.1038/s41386-021-01229-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/22/2021] [Accepted: 10/30/2021] [Indexed: 02/03/2023]
Abstract
Stressful life events are ubiquitous and well-known to negatively impact mental health. However, in both humans and animal models, there is large individual variability in how individuals respond to stress, with some but not all experiencing long-term adverse consequences. While there is growing understanding of the neurobiological underpinnings of the stress response, much less is known about how neurocircuits shaped by lifetime experiences are activated during an initial stressor and contribute to this selective vulnerability versus resilience. We developed a model of acute social defeat stress (ASDS) that allows classification of male mice into "susceptible" (socially avoidant) versus "resilient" (expressing control-level social approach) one hour after exposure to six minutes of social stress. Using circuit tracing and high-resolution confocal imaging, we explored differences in activation and dendritic spine density and morphology in the prelimbic cortex to basolateral amygdala (PL→BLA) circuit in resilient versus susceptible mice. Susceptible mice had greater PL→BLA recruitment during ASDS and activated PL→BLA neurons from susceptible mice had more and larger mushroom spines compared to resilient mice. We hypothesized identified structure/function differences indicate an overactive PL→BLA response in susceptible mice and used an intersectional chemogenetic approach to inhibit the PL→BLA circuit during or prior to ASDS. We found in both cases that this blocked ASDS-induced social avoidance. Overall, we show PL→BLA structure/function differences mediate divergent behavioral responses to ASDS in male mice. These results support PL→BLA circuit overactivity during stress as a biomarker of trait vulnerability and potential target for prevention of stress-induced psychopathology.
Collapse
Affiliation(s)
- Yael S Grossman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Duke University School of Medicine, Durham, NC, USA
| | - Clementine Fillinger
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessia Manganaro
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - George Voren
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Waldman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tiffany Zou
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul J Kenny
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dani Dumitriu
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, Columbia University, New York, NY, USA.
- Sackler Institute, Columbia University, New York, NY, USA.
- Columbia Population Research Center, Columbia University, New York, NY, USA.
- Zuckerman Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
31
|
Inflammatory Stress Induced by Intraperitoneal Injection of LPS Increases Phoenixin Expression and Activity in Distinct Rat Brain Nuclei. Brain Sci 2022; 12:brainsci12020135. [PMID: 35203899 PMCID: PMC8870310 DOI: 10.3390/brainsci12020135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Due to phoenixin’s role in restraint stress and glucocorticoid stress, as well as its recently shown effects on the inflammasome, we aimed to investigate the effects of lipopolysaccharide (LPS)-induced inflammatory stress on the activity of brain nuclei-expressing phoenixin. Male Sprague Dawley rats (n = 6/group) were intraperitoneally injected with either LPS or control (saline). Brains were processed for c-Fos and phoenixin immunohistochemistry and the resulting slides were evaluated using ImageJ software. c-Fos was counted and phoenixin was evaluated using densitometry. LPS stress significantly increased c-Fos expression in the central amygdaloid nucleus (CeM, 7.2-fold), supraoptic nucleus (SON, 34.8 ± 17.3 vs. 0.0 ± 0.0), arcuate nucleus (Arc, 4.9-fold), raphe pallidus (RPa, 5.1-fold), bed nucleus of the stria terminalis (BSt, 5.9-fold), dorsal motor nucleus of the vagus nerve (DMN, 89-fold), and medial part of the nucleus of the solitary tract (mNTS, 121-fold) compared to the control-injected group (p < 0.05). Phoenixin expression also significantly increased in the CeM (1.2-fold), SON (1.5-fold), RPa (1.3-fold), DMN (1.3-fold), and mNTS (1.9-fold, p < 0.05), leading to a positive correlation between c-Fos and phoenixin in the RPa, BSt, and mNTS (p < 0.05). In conclusion, LPS stress induces a significant increase in activity in phoenixin immunoreactive brain nuclei that is distinctively different from restraint stress.
Collapse
|
32
|
Agee LA, Nemchek V, Malone CA, Lee HJ, Monfils MH. Appetitive Behavior in the Social Transmission of Food Preference Paradigm Predicts Activation of Orexin-A producing Neurons in a Sex-Dependent Manner. Neuroscience 2022; 481:30-46. [PMID: 34843892 PMCID: PMC9246717 DOI: 10.1016/j.neuroscience.2021.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/22/2022]
Abstract
Orexin-producing cells in the lateral hypothalamic area have been shown to be involved in a wide variety of behavioral and cognitive functions, including the recall of appetitive associations and a variety of social behaviors. Here, we investigated the role of orexin in the acquisition and recall of socially transmitted food preferences in the rat. Rats were euthanized following either acquisition, short-term recall, or long-term recall of a socially transmitted food preference and their brains were processed for orexin-A and c-Fos expression. We found that while there were no significant differences in c-Fos expression between control and experimental subjects at any of the tested timepoints, females displayed significantly more activity in both orexinergic and non-orexinergic cells in the lateral hypothalamus. In the infralimbic cortex, we found that social behavior was significantly predictive of c-Fos expression, with social behaviors related to olfactory exploration appearing to be particularly influential. We additionally found that appetitive behavior was significantly predictive of orexin-A activity in a sex-dependent matter, with the total amount eaten correlating negatively with orexin-A/c-Fos colocalization in male rats but not female rats. These findings suggest a potential sex-specific role for the orexin system in balancing the stimulation of feeding behavior with the sleep/wake cycle.
Collapse
Affiliation(s)
- Laura A Agee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Victoria Nemchek
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Cassidy A Malone
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Marie-H Monfils
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
33
|
Nedelescu H, Wagner GE, De Ness GL, Carroll A, Kerr TM, Wang J, Zhang S, Chang S, Than AH, Emerson NE, Suto N, Weiss F. Cannabidiol Produces Distinct U-Shaped Dose-Response Effects on Cocaine-Induced Conditioned Place Preference and Associated Recruitment of Prelimbic Neurons in Male Rats. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:70-78. [PMID: 35252951 PMCID: PMC8896771 DOI: 10.1016/j.bpsgos.2021.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Cannabidiol (CBD) has received attention for the treatment of substance use disorders. In preclinical models of relapse, CBD attenuates drug seeking across several drugs of abuse, including cocaine. However, in these models CBD has not been consistently effective. This inconsistency in CBD effects may be related to presently insufficient information on the full spectrum of CBD dose effects on drug-related behaviors. Methods We address this issue by establishing a full dose-response profile of CBD’s actions using expression of cocaine-induced conditioned place preference as a model for drug-motivated behavior in male rats and by concurrently identifying dose-dependent effects of CBD on underlying neuronal activation and distinct neuronal phenotypes showing dose-dependent activation changes. Additionally, we established CBD levels in plasma and brain samples. Results CBD produced linear increases in CBD brain/plasma concentrations but suppressed conditioned place preference in a distinct U-shaped manner. In parallel with its behavioral effects, CBD produced U-shaped suppressant effects on neuronal activation in the prelimbic but not infralimbic cortex or nucleus accumbens core and shell. RNAscope in situ hybridization identified suppression of glutamatergic and GABAergic (gamma-aminobutyric acidergic) signaling in the prelimbic cortex as a possible cellular mechanism for the attenuation of cocaine-induced conditioned place preference by CBD. Conclusions The findings extend previous evidence on the potential of CBD in preventing drug-motivated behavior. However, CBD’s dose-response profile may have important dosing implications for future clinical applications and may contribute to the understanding of discrepant CBD effects on drug seeking reported in the literature.
Collapse
|
34
|
Peripheral Infection after Traumatic Brain Injury Augments Excitability in the Perilesional Cortex and Dentate Gyrus. Biomedicines 2021; 9:biomedicines9121946. [PMID: 34944762 PMCID: PMC8698476 DOI: 10.3390/biomedicines9121946] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022] Open
Abstract
Peripheral infections occur in up to 28% of patients with traumatic brain injury (TBI), which is a major etiology for structural epilepsies. We hypothesized that infection occurring after TBI acts as a “second hit” and facilitates post-traumatic epileptogenesis. Adult male Sprague–Dawley rats were subjected to lateral fluid-percussion injury or sham-operation. At 8 weeks post-injury, rats were treated with lipopolysaccharide (LPS, 5 mg/kg) to mimic Gram-negative peripheral infection. T2-weighted magnetic resonance imaging was used to detect the cortical lesion type (small focal inflammatory [TBIFI] vs. large cavity-forming [TBICF]). Spontaneous seizures were detected with video-electroencephalography, and seizure susceptibility was determined by the pentylenetetrazole (PTZ) test. Post-PTZ neuronal activation was assessed using c-Fos immunohistochemistry. LPS treatment increased the percentage of rats with PTZ-induced seizures among animals with TBIFI lesions (p < 0.05). It also increased the cumulative duration of PTZ-induced seizures (p < 0.01), particularly in the TBIFI group (p < 0.05). The number of c-Fos immunopositive cells was higher in the perilesional cortex of injured animals compared with sham-operated animals (p < 0.05), particularly in the TBI-LPS group (p < 0.05). LPS treatment increased the percentage of injured rats with bilateral c-Fos staining in the dentate gyrus (p < 0.05), particularly in the TBIFI group (p < 0.05). Our findings demonstrate that peripheral infection after TBI increases PTZ-induced seizure susceptibility and neuronal activation in the perilesional cortex and bilaterally in the dentate gyrus, particularly in animals with prolonged perilesional T2 enhancement. Our data suggest that treatment of infections and reduction of post-injury neuro-inflammation are important components of the treatment regimen aiming at preventing epileptogenesis after TBI.
Collapse
|
35
|
Patodia S, Somani A, Thom M. Review: Neuropathology findings in autonomic brain regions in SUDEP and future research directions. Auton Neurosci 2021; 235:102862. [PMID: 34411885 PMCID: PMC8455454 DOI: 10.1016/j.autneu.2021.102862] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022]
Abstract
Autonomic dysfunction is implicated from clinical, neuroimaging and experimental studies in sudden and unexpected death in epilepsy (SUDEP). Neuropathological analysis in SUDEP series enable exploration of acquired, seizure-related cellular adaptations in autonomic and brainstem autonomic centres of relevance to dysfunction in the peri-ictal period. Alterations in SUDEP compared to control groups have been identified in the ventrolateral medulla, amygdala, hippocampus and central autonomic regions. These involve neuropeptidergic, serotonergic and adenosine systems, as well as specific regional astroglial and microglial populations, as potential neuronal modulators, orchestrating autonomic dysfunction. Future research studies need to extend to clinically and genetically characterized epilepsies, to explore if common or distinct pathways of autonomic dysfunction mediate SUDEP. The ultimate objective of SUDEP research is the identification of disease biomarkers for at risk patients, to improve post-mortem recognition and disease categorisation, but ultimately, for exposing potential treatment targets of pharmacologically modifiable and reversible cellular alterations.
Collapse
Affiliation(s)
- Smriti Patodia
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alyma Somani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.
| |
Collapse
|
36
|
Petrisko TJ, Konat GW. Peripheral viral challenge increases c-fos level in cerebral neurons. Metab Brain Dis 2021; 36:1995-2002. [PMID: 34406561 DOI: 10.1007/s11011-021-00819-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/05/2021] [Indexed: 12/01/2022]
Abstract
Peripheral viral infection can substantially alter brain function. We have previously shown that intraperitoneal (i.p.) injection of a viral mimetic, polyinosinic-polycytidylic acid (PIC), engenders hyperexcitability of cerebral neurons. Because neuronal activity is invariably associated with their expression of the Cfos gene, the present study was undertaken to determine whether PIC challenge also increases neuronal c-fos protein level. Female C57BL/6 mice were i.p. injected with PIC, and neuronal c-fos was analyzed in the motor cortex by immunohistochemistry. PIC challenge instigated a robust increase in the number of c-fos-positive neurons. This increase reached approximately tenfold over control at 24 h. Also, the c-fos staining intensity of individual neurons increased. AMG-487, a specific inhibitor of the chemokine receptor CXCR3, profoundly attenuated the accumulation of neuronal c-fos, indicating the activation of CXCL10/CXCR3 axis as the trigger of the process. Together, these results show that the accumulation of c-fos is a viable readout to assess the response of cerebral neurons to peripheral PIC challenge, and to elucidate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Tiffany J Petrisko
- Department of Biochemistry, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Gregory W Konat
- Department of Biochemistry, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Biochemistry, West Virginia University School of Medicine, 4052 HSCN, P.O. Box 9128, Morgantown, WV, 26506-9128, USA.
| |
Collapse
|
37
|
Selected Molecular Targets for Antiepileptogenesis. Int J Mol Sci 2021; 22:ijms22189737. [PMID: 34575901 PMCID: PMC8466306 DOI: 10.3390/ijms22189737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023] Open
Abstract
The term epileptogenesis defines the usually durable process of converting normal brain into an epileptic one. The resistance of a significant proportion of patients with epilepsy to the available pharmacotherapy prompted the concept of a causative treatment option consisting in stopping or modifying the progress of epileptogenesis. Most antiepileptic drugs possess only a weak or no antiepileptogenic potential at all, but a few of them appear promising in this regard; these include, for example, eslicarbazepine (a sodium and T-type channel blocker), lamotrigine (a sodium channel blocker and glutamate antagonist) or levetiracetam (a ligand of synaptic vehicle protein SV2A). Among the approved non-antiepileptic drugs, antiepileptogenic potential seems to reside in losartan (a blocker of angiotensin II type 1 receptors), biperiden (an antiparkinsonian drug), nonsteroidal anti-inflammatory drugs, antioxidative drugs and minocycline (a second-generation tetracycline with anti-inflammatory and antioxidant properties). Among other possible antiepileptogenic compounds, antisense nucleotides have been considered, among these an antagomir targeting microRNA-134. The drugs and agents mentioned above have been evaluated in post-status epilepticus models of epileptogenesis, so their preventive efficacy must be verified. Limited clinical data indicate that biperiden in patients with brain injuries is well-tolerated and seems to reduce the incidence of post-traumatic epilepsy. Exceptionally, in this regard, our own original data presented here point to c-Fos as an early seizure duration, but not seizure intensity-related, marker of early epileptogenesis. Further research of reliable markers of early epileptogenesis is definitely needed to improve the process of designing adequate antiepileptogenic therapies.
Collapse
|
38
|
Carver CM, DeWitt HR, Stoja AP, Shapiro MS. Blockade of TRPC Channels Limits Cholinergic-Driven Hyperexcitability and Seizure Susceptibility After Traumatic Brain Injury. Front Neurosci 2021; 15:681144. [PMID: 34489621 PMCID: PMC8416999 DOI: 10.3389/fnins.2021.681144] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022] Open
Abstract
We investigated the contribution of excitatory transient receptor potential canonical (TRPC) cation channels to posttraumatic hyperexcitability in the brain 7 days following controlled cortical impact model of traumatic brain injury (TBI) to the parietal cortex in male adult mice. We investigated if TRPC1/TRPC4/TRPC5 channel expression is upregulated in excitatory neurons after TBI in contribution to epileptogenic hyperexcitability in key hippocampal and cortical circuits that have substantial cholinergic innervation. This was tested by measuring TRPC1/TRPC4/TRPC5 protein and messenger RNA (mRNA) expression, assays of cholinergic function, neuronal Ca2+ imaging in brain slices, and seizure susceptibility after TBI. We found region-specific increases in expression of TRPC1, TRPC4, and TRPC5 subunits in the hippocampus and cortex following TBI. The dentate gyrus, CA3 region, and cortex all exhibited robust upregulation of TRPC4 mRNA and protein. TBI increased cFos activity in dentate gyrus granule cells (DGGCs) and layer 5 pyramidal neurons both at the time of TBI and 7 days post-TBI. DGGCs displayed greater magnitude and duration of acetylcholine-induced rises in intracellular Ca2+ in brain slices from mice subjected to TBI. The TBI mice also exhibited greater seizure susceptibility in response to pentylenetetrazol-induced kindling. Blockade of TRPC4/TRPC5 channels with M084 reduced neuronal hyperexcitation and impeded epileptogenic progression of kindling. We observed that the time-dependent upregulation of TRPC4/TRPC5-containing channels alters cholinergic responses and activity of principal neurons acting to increase proexcitatory sensitivity. The underlying mechanism includes acutely decreased acetylcholinesterase function, resulting in greater Gq/11-coupled muscarinic receptor activation of TRPC channels. Overall, our evidence suggests that TBI-induced plasticity of TRPC channels strongly contributes to overt hyperexcitability and primes the hippocampus and cortex for seizures.
Collapse
Affiliation(s)
- Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Haley R DeWitt
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Aiola P Stoja
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Mark S Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
39
|
Gantz SC, Ortiz MM, Belilos AJ, Moussawi K. Excitation of medium spiny neurons by 'inhibitory' ultrapotent chemogenetics via shifts in chloride reversal potential. eLife 2021; 10:64241. [PMID: 33822716 PMCID: PMC8024007 DOI: 10.7554/elife.64241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/28/2021] [Indexed: 11/13/2022] Open
Abstract
Ultrapotent chemogenetics, including the chloride-permeable inhibitory PSAM4-GlyR receptor, were recently proposed as a powerful strategy to selectively control neuronal activity in awake, behaving animals. We aimed to validate the inhibitory function of PSAM4-GlyR in dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) in the ventral striatum. Activation of PSAM4-GlyR with the uPSEM792 ligand enhanced rather than suppressed the activity of D1-MSNs in vivo as indicated by increased c-fos expression in D1-MSNs and in vitro as indicated by cell-attached recordings from D1-MSNs in mouse brain slices. Whole-cell recordings showed that activation of PSAM4-GlyR depolarized D1-MSNs, attenuated GABAergic inhibition, and shifted the reversal potential of PSAM4-GlyR current to more depolarized potentials, perpetuating the depolarizing effect of receptor activation. These data show that 'inhibitory' PSAM4-GlyR chemogenetics may activate certain cell types and highlight the pitfalls of utilizing chloride conductances to inhibit neurons.
Collapse
Affiliation(s)
- Stephanie C Gantz
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Maria M Ortiz
- Biological and Biomedical Neuroscience Program, University of North Carolina, Chapel Hill, United States
| | | | - Khaled Moussawi
- National Institute on Drug Abuse, Baltimore, United States.,Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
40
|
Schlabitz S, Monni L, Ragot A, Dipper-Wawra M, Onken J, Holtkamp M, Fidzinski P. Spatiotemporal Correlation of Epileptiform Activity and Gene Expression in vitro. Front Mol Neurosci 2021; 14:643763. [PMID: 33859552 PMCID: PMC8042243 DOI: 10.3389/fnmol.2021.643763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/03/2021] [Indexed: 11/14/2022] Open
Abstract
Epileptiform activity alters gene expression in the central nervous system, a phenomenon that has been studied extensively in animal models. Here, we asked whether also in vitro models of seizures are in principle suitable to investigate changes in gene expression due to epileptiform activity and tested this hypothesis mainly in rodent and additionally in some human brain slices. We focused on three genes relevant for seizures and epilepsy: FOS proto-oncogene (c-Fos), inducible cAMP early repressor (Icer) and mammalian target of rapamycin (mTor). Seizure-like events (SLEs) were induced by 4-aminopyridine (4-AP) in rat entorhinal-hippocampal slices and by 4-AP/8 mM potassium in human temporal lobe slices obtained from surgical treatment of epilepsy. SLEs were monitored simultaneously by extracellular field potentials and intrinsic optical signals (IOS) for 1–4 h, mRNA expression was quantified by real time PCR. In rat slices, both duration of SLE exposure and SLE onset region were associated with increased expression of c-Fos and Icer while no such association was shown for mTor expression. Similar to rat slices, c-FOS induction in human tissue was increased in slices with epileptiform activity. Our results indicate that irrespective of limitations imposed by ex vivo conditions, in vitro models represent a suitable tool to investigate gene expression. Our finding is of relevance for the investigation of human tissue that can only be performed ex vivo. Specifically, it presents an important prerequisite for future studies on transcriptome-wide and cell-specific changes in human tissue with the goal to reveal novel candidates involved in the pathophysiology of epilepsy and possibly other CNS pathologies.
Collapse
Affiliation(s)
- Sophie Schlabitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Laura Monni
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Alienor Ragot
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Matthias Dipper-Wawra
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Julia Onken
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurosurgery, Berlin, Germany
| | - Martin Holtkamp
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany.,Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany
| | - Pawel Fidzinski
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany.,Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Neuroscience Research Center, Berlin, Germany
| |
Collapse
|
41
|
Magalhães SA, Foresti ML, Barros VN, Mello LE. Marmosets have a greater diversity of c-Fos response after hyperstimulation in distinct cortical regions as compared to rats. J Comp Neurol 2020; 529:1628-1641. [PMID: 32975324 DOI: 10.1002/cne.25044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/12/2020] [Accepted: 09/18/2020] [Indexed: 11/06/2022]
Abstract
Previous evidence indicated a potential mechanism that might support the fact that primates exhibit greater neural integration capacity as a result of the activation of different structures of the central nervous system, as compared to rodents. The current study aimed to provide further evidence to confirm previous findings by analyzing the patterns of c-Fos expression in more neocortical structures of rats and marmosets using a more robust quantitative technique and evaluating a larger number of brain areas. Nineteen Wistar rats and 21 marmosets (Callithrix jacchus) were distributed among control groups (animals without injections) and animals injected with pentylenetetrazol (PTZ) and euthanized at different time points after stimulus. Immunohistochemical detection of c-Fos was quantified using unbiased and efficient stereological cell counting in eight neocortical regions. Marmosets had a c-Fos expression that was notably more widely expressed (5× more cells) and longer lasting (up to 3 hr) than rats. c-Fos expression in rats presented similar patterns of expression according to the function of the brain cortical structures (associative, sensorial, and motor functions), which was not observed for marmosets (in which no clear pattern could be drawn, and a more diverse profile emerged). Our results provide evidence that the marmoset brain has a greater neuronal activation after intense stimulation by means of PTZ and a more complex pattern of brain activation. We speculate that these functional differences may contribute for the understanding of the different neuronal processing capacities of the neocortex in these mammals' orders.
Collapse
Affiliation(s)
| | - Maira Licia Foresti
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto D'Or de Pesquisa e Ensino, Botafogo, Brazil
| | | | - Luiz E Mello
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto D'Or de Pesquisa e Ensino, Botafogo, Brazil
| |
Collapse
|
42
|
Patodia S, Tan I, Ellis M, Somani A, Scheffer IE, Sisodiya SM, Thom M. Medullary tyrosine hydroxylase catecholaminergic neuronal populations in sudden unexpected death in epilepsy. Brain Pathol 2020; 31:133-143. [PMID: 32852867 PMCID: PMC8018054 DOI: 10.1111/bpa.12891] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/12/2020] [Accepted: 08/06/2020] [Indexed: 12/26/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is mechanistically complex and one probable cause is seizure‐related respiratory dysfunction. Medullary respiratory regulatory nuclei include the pre‐Bötzinger complex (pre‐BötC) in the ventrolateral medulla (VLM), the medullary raphé nuclei (MR) and nucleus of solitary tract in the dorsomedial medulla (DMM). The region of the VLM also contains intermingled tyrosine hydroxylase (TH) catecholaminergic neurones which directly project to the pre‐BötC and regulate breathing under hypoxic conditions and our aim was to evaluate these neurones in SUDEP cases. In post‐mortem cases from three groups [SUDEP (18), epilepsy controls (8) and non‐epilepsy controls (16)] serial sections of medulla (obex + 2 to + 13 mm) were immunolabeled for TH. Three regions of interest (ROI) were outlined (VLM, DMM and MR) and TH‐immunoreactive (TH‐IR) neurones were evaluated using automated detection for overall labeling index (neurones and processes) and neuronal densities and compared between groups and relative to obex level. C‐fos immunoreactivity was also semi‐quantitatively evaluated in these regions. We found no significant difference in the density of TH‐IR neurones or labeling index between the groups in all regions. Significantly more TH‐IR neurones were present in the DMM region than VLM in non‐epilepsy cases only (P < 0.01). Regional variations in TH‐IR neurones with obex level were seen in all groups except SUDEP. We also identified occasional TH neurones in the MR region in all groups. There was significantly less c‐fos labeling in the VLM and MR in SUDEP than non‐epilepsy controls but no difference with epilepsy controls. In conclusion, in this series we found no evidence for alteration of total medullary TH‐IR neuronal numbers in SUDEP but noted some differences in their relative distribution in the medulla and c‐fos neurones compared to control groups which may be relevant to the mechanism of death.
Collapse
Affiliation(s)
- Smriti Patodia
- Department of Clinical and Experimental epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 2BG, UK
| | - Ian Tan
- Department of Clinical and Experimental epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 2BG, UK
| | - Matthew Ellis
- School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Alyma Somani
- Department of Clinical and Experimental epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 2BG, UK
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine (Neurology), University of Melbourne, Victoria, 3052, Australia
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 2BG, UK.,Chalfont Centre for Epilepsy, Bucks, SL9 0RJ, UK
| | - Maria Thom
- Department of Clinical and Experimental epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 2BG, UK.,School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
43
|
Phillips ML, Pozzo-Miller L. Identification of Socially-activated Neurons. Bio Protoc 2020; 10:e3744. [PMID: 33659404 DOI: 10.21769/bioprotoc.3744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/21/2020] [Accepted: 07/15/2020] [Indexed: 11/02/2022] Open
Abstract
Determining the neuronal circuitry responsible for specific behaviors is a major focus in the field of neurobiology. Activity-dependent immediate early genes (IEGs), transcribed and translated shortly after neurons discharge action potentials, have been used extensively to either identify or gain genetic access to neurons and brain regions involved in such behaviors. By using immunohistochemistry for the protein product of the IEG c-Fos combined with retrograde labeling of specific neuronal populations, precise experimental timing, and identical data acquisition and processing, we present a method to quantitatively identify specific neuronal subpopulations that were active during social encounters. We have previously used this method to show a stronger recruitment of ventral hippocampal neurons that project to the medial prefrontal cortex, compared to those that project to the lateral hypothalamus, following social interactions. After optimization of surgeries for the injection of retrograde tracers, this method will be useful for the identification and mapping of neuronal populations engaged in many different behaviors.
Collapse
Affiliation(s)
- Mary L Phillips
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, 1825 University Blvd, Birmingham, AL, 35294, United States.,Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL, 33458, United States
| | - Lucas Pozzo-Miller
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, 1825 University Blvd, Birmingham, AL, 35294, United States
| |
Collapse
|
44
|
Forget B, Icick R, Robert J, Correia C, Prevost MS, Gielen M, Corringer PJ, Bellivier F, Vorspan F, Besson M, Maskos U. Alterations in nicotinic receptor alpha5 subunit gene differentially impact early and later stages of cocaine addiction: a translational study in transgenic rats and patients. Prog Neurobiol 2020; 197:101898. [PMID: 32841724 DOI: 10.1016/j.pneurobio.2020.101898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/06/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022]
Abstract
Cocaine addiction is a chronic and relapsing disorder with an important genetic component. Human candidate gene association studies showed that the single nucleotide polymorphism (SNP) rs16969968 in the α5 subunit (α5SNP) of nicotinic acetylcholine receptors (nAChRs), previously associated with increased tobacco dependence, was linked to a lower prevalence of cocaine use disorder (CUD). Three additional SNPs in the α5 subunit, previously shown to modify α5 mRNA levels, were also associated with CUD, suggesting an important role of the subunit in this pathology. To investigate the link between this subunit and CUD, we submitted rats knockout for the α5 subunit gene (α5KO), or carrying the α5SNP, to cocaine self-administration (SA) and showed that the acquisition of cocaine-SA was impaired in α5SNP rats while α5KO rats exhibited enhanced cocaine-induced relapse associated with altered neuronal activity in the nucleus accumbens. In addition, we observed in a human cohort of patients with CUD that the α5SNP was associated with a slower transition from first cocaine use to CUD. We also identified a novel SNP in the β4 nAChR subunit, part of the same gene cluster in the human genome and potentially altering CHRNA5 expression, associated with shorter time to relapse to cocaine use in patients. In conclusion, the α5SNP is protective against CUD by influencing early stages of cocaine exposure while CHRNA5 expression levels may represent a biomarker for the risk to relapse to cocaine use. Drugs modulating α5 containing nAChR activity may thus represent a novel therapeutic strategy against CUD.
Collapse
Affiliation(s)
- Benoît Forget
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France.
| | - Romain Icick
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France; Département de Psychiatrie et de Médecine Addictologique, Groupe Hospitalier Saint-Louis - Lariboisière - Fernand Widal, Assistance-Publique Hôpitaux de Paris, 75010, Paris, France; INSERM UMR_S1144, 4 avenue de l'Observatoire, 75006, Paris, France; Université Sorbonne - Paris - Cité, Paris, France
| | - Jonathan Robert
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Caroline Correia
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Marie S Prevost
- Unité Récepteurs-Canaux, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Marc Gielen
- Université Sorbonne - Paris - Cité, Paris, France; Unité Récepteurs-Canaux, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Pierre-Jean Corringer
- Unité Récepteurs-Canaux, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Frank Bellivier
- Département de Psychiatrie et de Médecine Addictologique, Groupe Hospitalier Saint-Louis - Lariboisière - Fernand Widal, Assistance-Publique Hôpitaux de Paris, 75010, Paris, France; Université Sorbonne - Paris - Cité, Paris, France; Unité Récepteurs-Canaux, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Florence Vorspan
- Département de Psychiatrie et de Médecine Addictologique, Groupe Hospitalier Saint-Louis - Lariboisière - Fernand Widal, Assistance-Publique Hôpitaux de Paris, 75010, Paris, France; INSERM UMR_S1144, 4 avenue de l'Observatoire, 75006, Paris, France; Université Sorbonne - Paris - Cité, Paris, France
| | - Morgane Besson
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France.
| | - Uwe Maskos
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France.
| |
Collapse
|
45
|
Evaluation of Anti-Convulsive Properties of Aqueous Kava Extract on Zebrafish Using the PTZ-Induced Seizure Model. Brain Sci 2020; 10:brainsci10080541. [PMID: 32796575 PMCID: PMC7463627 DOI: 10.3390/brainsci10080541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022] Open
Abstract
Kava roots have been extensively studied in clinical trials as potential candidate anti-anxiety drugs. However, anti-convulsive properties of various tissues of stems of Kava have not been reported to date. The objective of the study was to evaluate the anti-convulsive potential of aqueous extracts prepared from specific tissues of Kava (Piper methysticum) stems in zebrafish, using the PTZ-induced seizure model. The potency of each extract was compared in terms of the intensity of seizure scores and onset time after pre-treating the zebrafish before the PTZ challenge. The results indicate that aqueous extract of Kava stems without peel after 45 min of pre-treatment exhibited anti-convulsive potential at the dose of 50 mg/L. This study provides evidence to the anti-convulsive properties of peeled Kava stems and its potential for investigation and design of candidate anti-convulsive drugs.
Collapse
|
46
|
The retinoid X receptor: a nuclear receptor that modulates the sleep-wake cycle in rats. Psychopharmacology (Berl) 2020; 237:2055-2073. [PMID: 32472163 DOI: 10.1007/s00213-020-05518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/01/2020] [Indexed: 10/24/2022]
Abstract
RATIONALE The nuclear receptor retinoid X receptor (RXR) belongs to a nuclear receptor superfamily that modulates diverse functions via homodimerization with itself or several other nuclear receptors, including PPARα. While the activation of PPARα by natural or synthetic agonists regulates the sleep-wake cycle, the role of RXR in the sleep modulation is unknown. OBJECTIVES We investigated the effects of bexarotene (Bexa, a RXR agonist) or UVI 3003 (UVI, a RXR antagonist) on sleep, sleep homeostasis, levels of neurochemical related to sleep modulation, and c-Fos and NeuN expression. METHODS The sleep-wake cycle and sleep homeostasis were analyzed after application of Bexa or UVI. Moreover, we also evaluated whether Bexa or UVI could induce effects on dopamine, serotonin, norepinephrine epinephrine, adenosine, and acetylcholine contents, collected from either the nucleus accumbens or basal forebrain. In addition, c-Fos and NeuN expression in the hypothalamus was determined after Bexa or UVI treatments. RESULTS Systemic application of Bexa (1 mM, i.p.) attenuated slow-wave sleep and rapid eye movement sleep. In addition, Bexa increased the levels of dopamine, serotonin, norepinephrine epinephrine, adenosine, and acetylcholine sampled from either the nucleus accumbens or basal forebrain. Moreover, Bexa blocked the sleep rebound period after total sleep deprivation, increased in the hypothalamus the expression of c-Fos, and decreased NeuN activity. Remarkably, UVI 3003 (1 mM, i.p.) induced opposite effects in sleep, sleep homeostasis, neurochemicals levels, and c-Fos and NeuN activity. CONCLUSIONS The administration of RXR agonist or antagonist significantly impaired the sleep-wake cycle and exerted effects on the levels of neurochemicals related to sleep modulation. Moreover, Bexa or UVI administration significantly affected c-Fos and NeuN expression in the hypothalamus. Our findings highlight the neurobiological role of RXR on sleep modulation.
Collapse
|
47
|
Lewis V, Laberge F, Heyland A. Temporal Profile of Brain Gene Expression After Prey Catching Conditioning in an Anuran Amphibian. Front Neurosci 2020; 13:1407. [PMID: 31992968 PMCID: PMC6971186 DOI: 10.3389/fnins.2019.01407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
A key goal in modern neurobiology is to understand the mechanisms underlying learning and memory. To that end, it is essential to identify the patterns of gene expression and the temporal sequence of molecular events associated with learning and memory processes. It is also important to ascertain if and how these molecular events vary between organisms. In vertebrates, learning and memory processes are characterized by distinct phases of molecular activity involving gene transcription, structural change, and long-term maintenance of such structural change in the nervous system. Utilizing next generation sequencing techniques, we profiled the temporal expression patterns of genes in the brain of the fire-bellied toad Bombina orientalis after prey catching conditioning. The fire-bellied toad is a basal tetrapod whose neural architecture and molecular pathways may help us understand the ancestral state of learning and memory mechanisms in tetrapods. Differential gene expression following conditioning revealed activity in molecular pathways related to immediate early genes (IEG), cytoskeletal modification, axon guidance activity, and apoptotic processes. Conditioning induced early IEG activity coinciding with transcriptional activity and neuron structural modification, followed by axon guidance and cell adhesion activity, and late neuronal pruning. While some of these gene expression patterns are similar to those found in mammals submitted to conditioning, some interesting divergent expression profiles were seen, and differential expression of some well-known learning-related mammalian genes is missing altogether. These results highlight the importance of using a comparative approach in the study of the mechanisms of leaning and memory and provide molecular resources for a novel vertebrate model in the relatively poorly studied Amphibia.
Collapse
Affiliation(s)
- Vern Lewis
- Integrative Biology, University of Guelph, Guelph, ON, Canada
| | | | - Andreas Heyland
- Integrative Biology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
48
|
Yavuz M, Albayrak N, Özgür M, Gülçebi İdriz Oğlu M, Çavdar S, Onat F. The effect of prenatal and postnatal caffeine exposure on pentylentetrazole induced seizures in the non-epileptic and epileptic offsprings. Neurosci Lett 2019; 713:134504. [PMID: 31539618 DOI: 10.1016/j.neulet.2019.134504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 11/29/2022]
Abstract
Caffeine, a central nervous system stimulant, has been reported to modulate seizure activity in various studies. In this study the effects of caffeine exposure on the pentylenetetrazole (PTZ) induced seizure thresholds and seizure stages in the Wistar and genetic absence epilepsy model offsprings were examined. Adult female and male Wistar rats and genetic absence epilepsy rats from Strasbourg (GAERS) consumed caffeine dissolved in water (0.3 g/L) before conception, during the gestational periods and lactation period whereas control groups of each strain received tap water. All offsprings at postnatal day 30 (PN30) subjected to 70 mg/kg of PTZ were evaluated in terms of overall seizure stages, the latency to the first generalized seizure and the c-Fos protein activity in the brain regions of somatosensorial cortex (SSCx), reticular thalamic nucleus (Rt), ventrobasal thalamus (VB), centromedial nucleus (CM) and lateral geniculate nucleus (LGN). The Wistar caffeine group had significantly shorter latency to the first generalized seizure (1.53 ± 0.49 min) comparing to the Wistar control offsprings (3.40 ± 0.68 min). GAERS caffeine group (6.52 ± 2.48 min) showed significantly longer latency comparing to Wistar caffeine group (1.53 ± 0.49 min). Although statistically not significant, GAERS caffeine group showed a longer latency comparing to the GAERS control group (4.71 ± 1.82 min). In all regions of SSCx, Rt, VB, CM and LGN, GAERS caffeine group had lower c-Fos protein expression comparing to the GAERS control group (p < 0.05). Wistar caffeine rats had lower expression of c-Fos protein comparing to the Wistar control group only in SSCx. In CM, GAERS rats expressed lower c-Fos protein comparing to the Wistar control (p < 0.05). In conclusion differential effects of caffeine in the seizure modulation may involve c-Fos protein activity-dependent protection mechanisms.
Collapse
Affiliation(s)
- Melis Yavuz
- Department of Medical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Nazlı Albayrak
- School of Medicine, Acibadem M. A. Aydınlar University, Istanbul, Turkey
| | - Merve Özgür
- Department of Anatomy, School of Medicine, Koç University, Istanbul, Turkey
| | - Medine Gülçebi İdriz Oğlu
- Department of Medical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Epilepsy Research Centre (EPAM), Marmara University, Istanbul, Turkey
| | - Safiye Çavdar
- Department of Anatomy, School of Medicine, Koç University, Istanbul, Turkey
| | - Filiz Onat
- Department of Medical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Epilepsy Research Centre (EPAM), Marmara University, Istanbul, Turkey.
| |
Collapse
|
49
|
Koyuncuoğlu T, Arabacı Tamer S, Erzik C, Karagöz A, Akakın D, Yüksel M, Yeğen BÇ. Oestrogen receptor ERα and ERβ agonists ameliorate oxidative brain injury and improve memory dysfunction in rats with an epileptic seizure. Exp Physiol 2019; 104:1911-1928. [PMID: 31608530 DOI: 10.1113/ep087986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
NEW FINDINGS What is the central question of this study? Could different hormonally active substances, including oestrogen receptor (ER) agonists, protect against oxidative brain damage and memory impairment induced by a single epileptic seizure in rats? If so, which signalling mechanisms are involved in their anti-inflammatory effects? What is the main finding and its importance? Chronic administration of oestrogen, progesterone, ER modulators/agonists or blockade of testosterone exhibited anti-inflammatory and antioxidant actions on single seizure-induced neuronal injury, while ER agonists additionally improved memory function and up-regulated CREB signalling and hippocampal GABA(A)α1 receptor density, suggesting that ERα or ERβ receptor activation may be beneficial in protecting against seizure-related oxidative brain injury and cognitive dysfunction. ABSTRACT The susceptibility to epileptic seizures is dependent on sex as well as fluctuations in oestrogen levels, while exogenous oestrogen was shown to have no effect or to facilitate or to inhibit seizure activity. Oestrogen receptors (ERs) mediate antioxidant and anti-inflammatory actions in several inflammatory models, but the involvement of ERs in seizure-induced neuronal injury has not been evaluated previously. In order to assess the effects of resveratrol, progesterone, oestradiol (E2), an anti-testosterone (cyproterone acetate; CPA), a selective ER modulator (tamoxifen; TMX) and ERα/ERβ agonists (propyl pyrazole triol (PPT), diarylpropionitrile (DPN)) on oxidative brain damage and memory impairment due to epileptic seizure, male Wistar rats (n = 120) received one of the treatment choices either in drinking water or intraperitoneally for 31 days, and epileptic seizure was induced on the 28th day by injection of a single-dose of pentylenetetrazole (45 mg kg-1 ). The results demonstrate that chronic pretreatment with resveratrol, progesterone, E2, CPA or TMX suppressed most of the inflammatory parameters indicative of oxidative neuronal injury, while treatment with the ER agonists DPN or PPT were found to be even more effective in limiting the oxidative damage. Treatment with DPN resulted in the up-regulation of cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) expression, while PPT up-regulated expression of CREB without affecting BDNF levels. Moreover, both ER agonists provided protection against seizure-induced memory loss with a concomitant increase in hippocampal GABA(A)α1-positive cells. In conclusion, ER agonists, and more specifically ERβ agonist, appear to provide maximum protection against seizure-induced oxidative brain injury and associated memory dysfunction by up-regulating the expression of CREB, BDNF and GABA(A)α1 receptors.
Collapse
Affiliation(s)
- Türkan Koyuncuoğlu
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Sevil Arabacı Tamer
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Can Erzik
- Department of Medical Biology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ayça Karagöz
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Dilek Akakın
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Meral Yüksel
- Department of Medical Laboratory, Vocational School of Health-Related Professions, Marmara University, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
50
|
Çelik H, Kucukler S, Çomaklı S, Özdemir S, Caglayan C, Yardım A, Kandemir FM. Morin attenuates ifosfamide-induced neurotoxicity in rats via suppression of oxidative stress, neuroinflammation and neuronal apoptosis. Neurotoxicology 2019; 76:126-137. [PMID: 31722249 DOI: 10.1016/j.neuro.2019.11.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/06/2019] [Accepted: 11/09/2019] [Indexed: 01/10/2023]
Abstract
Ifosfamide (IFA), a commonly used chemotherapeutic drug, has been frequently associated with encephalopathy and central nervous system toxicity. The present study aims to investigate whether morin could protect against acute IFA-induced neurotoxicity. Morin was administered to male rats once daily for 2 consecutive days at doses of 100 and 200 mg/kg body weight (BW) orally. IFA (500 mg/kg BW; i.p.) was administered on second day. The results showed that morin markedly inhibited the production of acetylcholinesterase (AChE), butrylcholinesterase (BChE), carbonic anhydrase (CA), glial fibrillary acidic protein (GFAP), brain-derived neurotrophic factor (BDNF) and nuclear factor erythroid 2-related factor 2 (Nrf-2) induced by IFA. Morin ameliorated IFA-induced lipid peroxidation, glutathione (GSH) depletion, and decrease antioxidant enzyme activities, catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Histopathological changes and immunohistochemical expressions of c-Jun N-terminal kinase (JNK) and c-Fos in the IFA-induced brain tissues were decreased after administration of morin. Furthermore, morin was able to down regulate the levels of inflammatory and apoptotic markers such as nuclear factor kappa B (NF-κB), neuronal nitric oxide synthase (nNOS), tumor necrosis factor-α (TNF-α), p53, cysteine aspartate specific protease-3 (caspase-3) and B-cell lymphoma-2 (Bcl-2). Taken together, our results demonstrated that morin elicited a typical chemoprotective effect on IFA-induced acute neurotoxicity.
Collapse
Affiliation(s)
- Hamit Çelik
- Department of Neurology, Private Buhara Hospital, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Cuneyt Caglayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, Bingol,Turkey.
| | - Ahmet Yardım
- Department of Neurosurgery, Private Buhara Hospital, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| |
Collapse
|