1
|
Park MG, Lim J, Kim D, Lee WS, Yoon BE, Lee CJ. Suppressing astrocytic GABA transaminase enhances tonic inhibition and weakens hippocampal spatial memory. Exp Mol Med 2025; 57:379-389. [PMID: 39894826 PMCID: PMC11873293 DOI: 10.1038/s12276-025-01398-0] [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: 04/26/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
Pharmacological suppression of γ-aminobutyric acid (GABA) transaminase (GABA-T), the sole GABA-degrading enzyme and a potential therapeutic target for treating brain disorders such as epilepsy, increases not only phasic inhibition but also tonic inhibition. However, the specific cellular source, neuromodulatory effects and potential therapeutic benefits of this enhanced tonic inhibition remain unexplored due to the lack of cell-type-specific gene manipulation studies. Here we report that the increase in tonic GABA currents observed after GABA-T suppression is predominantly due to increased tonic GABA release from astrocytes rather than action-potential-dependent synaptic GABA spillover. General GABA-T knockdown (KD) by a short hairpin RNA considerably increased tonic GABA currents in dentate granule cells, thereby enhancing tonic inhibition. An astrocyte-specific rescue of GABA-T following general GABA-T KD normalized the elevated tonic GABA currents to near control levels. Tetrodotoxin-insensitive tonic GABA currents were significantly increased after general GABA-T KD, whereas tetrodotoxin-sensitive tonic GABA currents showed no significant increase, suggesting that this enhanced tonic inhibition is primarily action-potential independent. General GABA-T KD reduced the spike probability of granule cells and impaired dorsal hippocampus-dependent spatial memory, which were fully reversed by astrocyte-specific GABA-T rescue. These findings suggest that suppressing astrocytic GABA-T may be sufficient to influence the excitatory/inhibitory balance in the brain and associated behaviors. Our study implies that the therapeutic benefits of pharmacological GABA-T suppression may be largely attributed to the modulation of astrocytic GABA-T and its impact on tonic GABA release from astrocytes. Here, we report distinct effects of GABA-T suppression depending on cell type; suppressing GABA-T in astrocytes enhances tonic inhibition, while its suppression in GABAergic neurons augments phasic inhibition. Our findings demonstrate that targeted suppression of astrocytic GABA-T not only enhances tonic GABA release from astrocytes but also significantly influences the excitation/inhibition balance in the brain, with consequential effects on behavior. This suggests that astrocytic GABA-T modulation holds promising potential for developing novel therapeutic strategies aimed at treating cognitive and neurological disorders through the regulation of astrocytic GABA metabolism. GAD glutamate decarboxylase, MAO-B monoamine oxidase B, BEST1 bestrophin 1, GABA-T GABA transaminase, GAT GABA transporter, DG dentate gyrus, SSA succinic semialdehyde.
Collapse
Affiliation(s)
- Mingu Gordon Park
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
- IBS School, University of Science and Technology, Daejeon, South Korea
| | - Daeun Kim
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
| | - Won-Seok Lee
- Department of Molecular Biology, Dankook University, Cheonan, South Korea
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea.
- IBS School, University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
2
|
Dentel B, Angeles-Perez L, Flores AY, Lei K, Ren C, Sanchez AP, Tsai PT. Neuronal cell type specific roles for Nprl2 in neurodevelopmental disorder-relevant behaviors. Neurobiol Dis 2025; 205:106790. [PMID: 39765274 DOI: 10.1016/j.nbd.2025.106790] [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: 05/20/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Loss of function in the subunits of the GTPase-activating protein (GAP) activity toward Rags-1 (GATOR1) complex, an amino-acid sensitive negative regulator of the mechanistic target of rapamycin complex 1 (mTORC1), is implicated in both genetic familial epilepsies and Neurodevelopmental Disorders (NDDs) (Baldassari et al., 2018). Previous studies have found seizure phenotypes and increased activity resulting from conditional deletion of GATOR1 function from forebrain excitatory neurons (Yuskaitis et al., 2018; Dentel et al., 2022); however, studies focused on understanding mechanisms contributing to NDD-relevant behaviors are lacking, especially studies understanding the contributions of GATOR1's critical GAP catalytic subunit, nitrogen permease regulator like-2 (Nprl2). Given the clinical phenotypes observed in patients with Nprl2 mutations, in this study, we sought to investigate the neuronal cell type contributions of Nprl2 to NDD behaviors. We conditionally deleted Nprl2 broadly in most neurons (Synapsin1cre), in inhibitory neurons only (Vgatcre), and in Purkinje cells within the cerebellum (L7cre). Broad neuronal deletion of Nprl2 resulted in seizures, social and learning deficits, and hyperactivity. In contrast, deleting Nprl2 from inhibitory neurons led to increased motor learning, hyperactive behavior, in addition to social and learning deficits. Lastly, Purkinje cell (PC) loss of Nprl2 also led to learning and social deficits but did not affect locomotor activity. These phenotypes enhance understanding of the spectrum of disease found in human populations with GATOR1 loss of function and highlight the significance of distinct cellular populations to NDD-related behaviors. SIGNIFICANCE STATEMENT: We aim to elucidate the neuronal-specific contributions of nitrogen permease regulator like-2 (Nprl2) to its neurodevelopmental disorder (NDD)-relevant phenotypes. We conditionally deleted Nprl2 broadly in neurons (Syn1cre), in inhibitory neurons (Vgatcre), and in cerebellar Purkinje cells (L7cre). We identify seizures only in the Syn1cre conditional mutant (cKO); hyperactivity, learning difficulties, social deficits, and impulsivity in the Syn1cre and Vgatcre cKOs; and social deficits, and fear learning deficits in L7cre cKOs. To our knowledge, we are the first to describe the behavioral contributions of Nprl2's function across multiple cell types. Our findings highlight both critical roles for Nprl2 in learning and behavior and also distinct contributions of select neuronal populations to these NDD-relevant behaviors.
Collapse
Affiliation(s)
- Brianne Dentel
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Lidiette Angeles-Perez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Abigail Y Flores
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Katherine Lei
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Chongyu Ren
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Andrea Pineda Sanchez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Peter T Tsai
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Psychiatry, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Pediatrics, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Neuroscience; O'Donnell Brain Institute, Dallas, TX, United States of America.
| |
Collapse
|
3
|
Ding C, Wu Y, Zhan C, Naseem A, Chen L, Li H, Yang B, Liu Y. Research progress on the role and inhibitors of Keap1 signaling pathway in inflammation. Int Immunopharmacol 2024; 141:112853. [PMID: 39159555 DOI: 10.1016/j.intimp.2024.112853] [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: 06/18/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
Inflammation is a protective mechanism against endogenous and exogenous pathogens. It is a typical feature of numerous chronic diseases and their complications. Keap1 is an essential target in oxidative stress and inflammatory diseases. Among them, the Keap1-Nrf2-ARE pathway (including Keap1-Nrf2-HO-1) is the most significant pathway of Keap1 targets, which participates in the control of inflammation in multiple organs (including renal inflammation, lung inflammation, liver inflammation, neuroinflammation, etc.). Identifying new Keap1 inhibitors is crucial for new drug discovery. However, most drugs have specificity issues as they covalently bind to cysteine residues of Keap1, causing off-target effects. Therefore, direct inhibition of Keap1-Nrf2 PPIs is a new research idea. Through non-electrophilic and non-covalent binding, its inhibitors have better specificity and ability to activate Nrf2, and targeting therapy against Keap1-Nrf2 PPIs has become a new method for drug development in chronic diseases. This review summarizes the members and downstream genes of the Keap1-related pathway and their roles in inflammatory disease models. In addition, we summarize all the research progress of anti-inflammatory drugs targeting Keap1 from 2010 to 2024, mainly describing their biological functions, molecular mechanisms of action, and therapeutic roles in inflammatory diseases.
Collapse
Affiliation(s)
- Chao Ding
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Ying Wu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Chaochao Zhan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Anam Naseem
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Yan Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| |
Collapse
|
4
|
Parodi G, Brofiga M, Pastore VP, Chiappalone M, Martinoia S. Deepening the role of excitation/inhibition balance in human iPSCs-derived neuronal networks coupled to MEAs during long-term development. J Neural Eng 2023; 20:056011. [PMID: 37678214 DOI: 10.1088/1741-2552/acf78b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023]
Abstract
Objective.The purpose of this study is to investigate whether and how the balance between excitation and inhibition ('E/I balance') influences the spontaneous development of human-derived neuronal networksin vitro. To achieve that goal, we performed a long-term (98 d) characterization of both homogeneous (only excitatory or inhibitory neurons) and heterogeneous (mixed neuronal types) cultures with controlled E/I ratios (i.e. E:I 0:100, 25:75, 50:50, 75:25, 100:0) by recording their electrophysiological activity using micro-electrode arrays.Approach.Excitatory and inhibitory neurons were derived from human induced pluripotent stem cells (hiPSCs). We realized five different configurations by systematically varying the glutamatergic and GABAergic percentages.Main results.We successfully built both homogeneous and heterogeneous neuronal cultures from hiPSCs finely controlling the E/I ratios; we were able to maintain them for up to 3 months. Homogeneity differentially impacted purely inhibitory (no bursts) and purely excitatory (few bursts) networks, deviating from the typical traits of heterogeneous cultures (burst dominated). Increased inhibition in heterogeneous cultures strongly affected the duration and organization of bursting and network bursting activity. Spike-based functional connectivity and image-based deep learning analysis further confirmed all the above.Significance.Healthy neuronal activity is controlled by a well-defined E/I balance whose alteration could lead to the onset of neurodevelopmental disorders like schizophrenia or epilepsy. Most of the commonly usedin vitromodels are animal-derived or too simplified and thus far from thein vivohuman condition. In this work, by performing a long-term study of hiPSCs-derived neuronal networks obtained from healthy human subjects, we demonstrated the feasibility of a robustin vitromodel which can be further exploited for investigating pathological conditions where the E/I balance is impaired.
Collapse
Affiliation(s)
- Giulia Parodi
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | - Martina Brofiga
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- ScreenNeuroPharm s.r.l, Sanremo, Italy
- Neurofacility, Istituto Italiano di Tecnologia, Genova, Italy
| | - Vito Paolo Pastore
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- Machine Learning Genoa Center (MaLGa), Department of Informatics, Bioengineering, Robotics, and Systems Engineering, University of Genova, Genova, Italy
| | - Michela Chiappalone
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| |
Collapse
|
5
|
Kim HJ, Kim SY, Kim GE, Jin HJ. Association between genetic polymorphisms of synaptophysin (SYP) gene and attention deficit hyperactivity disorder in Korean subjects. Genes Genomics 2023; 45:1097-1105. [PMID: 37133725 DOI: 10.1007/s13258-023-01393-7] [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: 12/10/2021] [Accepted: 04/19/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) is a common childhood neurodevelopmental disorder, and the prevalence of ADHD among Korean children has attained about 8.5%. Various genetic factors can contribute to the etiology of the disease. Synaptophysin (SYP) regulates neurotransmitter release and synaptic plasticity. According to previous studies, several genetic polymorphisms on SYP were risk factors for ADHD. OBJECTIVE We investigated the effect of the SYP gene polymorphisms (rs2293945 and rs3817678) on ADHD in Korean children. METHODS In this study, we examined the case-control study in 150 ADHD cases and 322 controls. The genotyping of SYP gene polymorphisms was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). RESULTS Significant associations in the genotype and genetic models of SYP rs2293945 polymorphism between girls with ADHD and control girls were found. The girls with ADHD having the C/T genotype were significantly associated with ADHD. In the dominant model of rs3817678, C/T + T/T genotypes were significantly associated with ADHD. The haplotype analyses showed significant associations from haplotypes of rs2293945 T-rs3817678 G and rs2293945 C-rs3817678 A. CONCLUSION Our results imply that the SYP rs2293945 C/T polymorphism in female participants may provide a possible effect on the genetic etiology of ADHD.
Collapse
Affiliation(s)
- Hyung Jun Kim
- Department of Biological Science, College of Science & Technology, Dankook University, 31116, Cheonan, South Korea
| | - Seong Yong Kim
- Department of Biological Science, College of Science & Technology, Dankook University, 31116, Cheonan, South Korea
| | - Ga Eun Kim
- Department of Biological Science, College of Science & Technology, Dankook University, 31116, Cheonan, South Korea
| | - Han Jun Jin
- Department of Biological Science, College of Science & Technology, Dankook University, 31116, Cheonan, South Korea.
| |
Collapse
|
6
|
Koh W, Kwak H, Cheong E, Lee CJ. GABA tone regulation and its cognitive functions in the brain. Nat Rev Neurosci 2023; 24:523-539. [PMID: 37495761 DOI: 10.1038/s41583-023-00724-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/28/2023]
Abstract
γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter released at GABAergic synapses, mediating fast-acting phasic inhibition. Emerging lines of evidence unequivocally indicate that a small amount of extracellular GABA - GABA tone - exists in the brain and induces a tonic GABA current that controls neuronal activity on a slow timescale relative to that of phasic inhibition. Surprisingly, studies indicate that glial cells that synthesize GABA, such as astrocytes, release GABA through non-vesicular mechanisms, such as channel-mediated release, and thereby act as the source of GABA tone in the brain. In this Review, we first provide an overview of major advances in our understanding of the cell-specific molecular and cellular mechanisms of GABA synthesis, release and clearance that regulate GABA tone in various brain regions. We next examine the diverse ways in which the tonic GABA current regulates synaptic transmission and synaptic plasticity through extrasynaptic GABAA-receptor-mediated mechanisms. Last, we discuss the physiological mechanisms through which tonic inhibition modulates cognitive function on a slow timescale. In this Review, we emphasize that the cognitive functions of tonic GABA current extend beyond mere inhibition, laying a foundation for future research on the physiological and pathophysiological roles of GABA tone regulation in normal and abnormal psychiatric conditions.
Collapse
Affiliation(s)
- Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - Hankyul Kwak
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea.
| |
Collapse
|
7
|
Genetic association study between Astrotactin-2 (ASTN2) rs10817999 gene polymorphism and attention deficit hyperactivity disorder in Korean children. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
8
|
Sex and Age-Dependent Olfactory Memory Dysfunction in ADHD Model Mice. Life (Basel) 2023; 13:life13030686. [PMID: 36983841 PMCID: PMC10056048 DOI: 10.3390/life13030686] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
ADHD is a typical neurodevelopmental disorder with a high prevalence rate. NSCs in the subventricular zone (SVZ) are closely related to neurodevelopmental disorder and can affect olfactory function by neurogenesis and migratory route. Although olfactory dysfunction is one of the symptoms of ADHD, the relevance of cells in the olfactory bulb derived from NSCs has not been studied. Therefore, we investigated olfactory memory and NSCs in Git1-deficient mice, under the ADHD model. Interestingly, only adult male G protein-coupled receptor kinase-interacting protein-1 (GIT1)-deficient (+/−, HE) mice showed impaired olfactory memory, suggesting sex and age dependence. We performed adult NSCs culture from the SVZ and observed distinct cell population in both sex and genotype. Taken together, our study suggests that the altered differentiation of NSCs in GIT1+/− mice can contribute to olfactory dysfunction in ADHD.
Collapse
|
9
|
Ottappilakkil H, Babu S, Balasubramanian S, Manoharan S, Perumal E. Fluoride Induced Neurobehavioral Impairments in Experimental Animals: a Brief Review. Biol Trace Elem Res 2023; 201:1214-1236. [PMID: 35488996 DOI: 10.1007/s12011-022-03242-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/09/2022] [Indexed: 02/07/2023]
Abstract
Fluoride is one of the major toxicants in the environment and is often found in drinking water at higher concentrations. Living organisms including humans exposed to high fluoride levels are found to develop mild-to-severe detrimental pathological conditions called fluorosis. Fluoride can cross the hematoencephalic barrier and settle in various brain regions. This accumulation affects the structure and function of both the central and peripheral nervous systems. The neural ultrastructure damages are reflected in metabolic and cognitive activities. Hindrances in synaptic plasticity and signal transmission, early neuronal apoptosis, functional alterations of the intercellular signaling pathway components, improper protein synthesis, dyshomeostasis of the transcriptional and neurotrophic factors, oxidative stress, and inflammatory responses are accounted for the fluoride neurotoxicity. Fluoride causes a decline in brain functions that directly influence the overall quality of life in both humans and animals. Animal studies are widely used to explore the etiology of fluoride-induced neurotoxicity. A good number of these studies support a positive correlation between fluoride intake and toxicity phenotypes closely associated with neurotoxicity. However, the experimental dosages highly surpass the normal environmental concentrations and are difficult to compare with human exposures. The treatment procedures are highly dependent on the dosage, duration of exposure, sex, and age of specimens among other factors which make it difficult to arrive at general conclusions. Our review aims to explore fluoride-induced neuronal damage along with associated histopathological, behavioral, and cognitive effects in experimental models. Furthermore, the correlation of various molecular mechanisms upon fluoride intoxication and associated neurobehavioral deficits has been discussed. Since there is no well-established mechanism to prevent fluorosis, phytochemical-based alleviation of its characteristic indications has been proposed as a possible remedial measure.
Collapse
Affiliation(s)
| | - Srija Babu
- Bharathiar University, Coimbatore, Tamilnadu, India
| | | | | | | |
Collapse
|
10
|
Custodio RJP, Kim M, Chung YC, Kim BN, Kim HJ, Cheong JH. Thrsp Gene and the ADHD Predominantly Inattentive Presentation. ACS Chem Neurosci 2023; 14:573-589. [PMID: 36716294 DOI: 10.1021/acschemneuro.2c00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
There are three presentations of attention-deficit/hyperactivity disorder (ADHD): the predominantly inattention (ADHD-PI), predominantly hyperactive-impulsive (ADHD-HI), and combined (ADHD-C) presentations of ADHD. These may represent distinct childhood-onset neurobehavioral disorders with separate etiologies. ADHD diagnoses are behaviorally based, so investigations into potential etiologies should be founded on behavior. Animal models of ADHD demonstrate face, predictive, and construct validity when they accurately reproduce elements of the symptoms, etiology, biochemistry, and disorder treatment. Spontaneously hypertensive rats (SHR/NCrl) fulfill many validation criteria and compare well with clinical cases of ADHD-C. Compounding the difficulty of selecting an ideal model to study specific presentations of ADHD is a simple fact that our knowledge regarding ADHD neurobiology is insufficient. Accordingly, the current review has explored a potential animal model for a specific presentation, ADHD-PI, with acceptable face, predictive, and construct validity. The Thrsp gene could be a biomarker for ADHD-PI presentation, and THRSP OE mice could represent an animal model for studying this distinct ADHD presentation.
Collapse
Affiliation(s)
- Raly James Perez Custodio
- Department of Ergonomics, Leibniz Research Centre for Working Environment and Human Factors─IfADo, Ardeystraße 67, 44139 Dortmund, Germany
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, 815 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea.,Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University Medical School, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Bung-Nyun Kim
- Department of Psychiatry and Behavioral Science, College of Medicine, Seoul National University, 101 Daehakro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Jae Hoon Cheong
- Institute for New Drug Development, School of Pharmacy, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| |
Collapse
|
11
|
Inhibiting peripheral and central MAO-B ameliorates joint inflammation and cognitive impairment in rheumatoid arthritis. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1188-1200. [PMID: 35982301 PMCID: PMC9440195 DOI: 10.1038/s12276-022-00830-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/07/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by chronic inflammation and the destruction of joints and systemic organs. RA is commonly accompanied by neuropsychiatric complications, such as cognitive impairment and depression. However, the role of monoamine oxidase (MAO) and its inhibitors in controlling neurotransmitters associated with these complications in RA have not been clearly identified. Here, we report that peripheral and central MAO-B are highly associated with joint inflammation and cognitive impairment in RA, respectively. Ribonucleic acid (RNA) sequencing and protein expression quantification were used to show that MAO-B and related molecules, such as gamma aminobutyric acid (GABA), were elevated in the inflamed synovium of RA patients. In primary cultured fibroblast-like synoviocytes in the RA synovium, MAO-B expression was significantly increased by tumor necrosis factor (TNF)-α-induced autophagy, which produces putrescine, the polyamine substrate for GABA synthesis. We also observed that MAO-B-mediated aberrant astrocytic production of GABA was augmented by interleukin (IL)-1β and inhibited CA1-hippocampal pyramidal neurons, which are responsible for memory storage, in an animal model of RA. Moreover, a newly developed reversible inhibitor of MAO-B ameliorated joint inflammation by inhibiting cyclooxygenase (Cox)-2. Therefore, MAO-B can be an effective therapeutic target for joint inflammation and cognitive impairment in patients with RA. Inhibiting an enzyme that is upregulated during joint inflammation may prove a valuable therapy for rheumatoid arthritis (RA). As well as causing considerable pain and discomfort in the joints, RA can also trigger neuropsychiatric problems including depression and memory impairment. The monoamine oxidase (MAO) enzyme family is involved in the control of neurotransmitters, and there is evidence that links MAO-B levels with systemic inflammation. C. Justin Lee at Center for Cognition and Sociality, Institute for Basic Science,, Daejeon, South Korea, and co-workers examined the role of MAO-B in RA using patient tissue samples and mouse models. MAO-B and related molecules were upregulated in patients’ inflamed joint tissues. In mice, elevated MAO-B triggered the inhibition of nerve cell activity related to memory storage. A novel drug that inhibits MAO-B reduced RA-related inflammation and cognitive impairment in mice, suggesting a promising approach to treatment.
Collapse
|
12
|
Lee WS, Kang JH, Lee JH, Kim YS, Kim JJ, Kim HS, Kim HW, Shin US, Yoon BE. Improved gliotransmission by increasing intracellular Ca 2+ via TRPV1 on multi-walled carbon nanotube platforms. J Nanobiotechnology 2022; 20:367. [PMID: 35953847 PMCID: PMC9367080 DOI: 10.1186/s12951-022-01551-1] [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: 05/16/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Astrocyte is a key regulator of neuronal activity and excitatory/inhibitory balance via gliotransmission. Recently, gliotransmission has been identified as a novel target for neurological diseases. However, using the properties of nanomaterials to modulate gliotransmission has not been uncovered. Results We prepared non-invasive CNT platforms for cells with different nanotopography and properties such as hydrophilicity and conductivity. Using CNT platforms, we investigated the effect of CNT on astrocyte functions participating in synaptic transmission by releasing gliotransmitters. Astrocytes on CNT platforms showed improved cell adhesion and proliferation with upregulated integrin and GFAP expression. In addition, intracellular GABA and glutamate in astrocytes were augmented on CNT platforms. We also demonstrated that gliotransmitters in brain slices were increased by ex vivo incubation with CNT. Additionally, intracellular resting Ca2+ level, which is important for gliotransmission, was also increased via TRPV1 on CNT platforms. Conclusion CNT can improve astrocyte function including adhesion, proliferation and gliotransmission by increasing resting Ca2+ level. Therefore, our study suggests that CNT would be utilized as a new therapeutic platform for central nervous system diseases by modulating gliotransmission. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01551-1.
Collapse
Affiliation(s)
- Won-Seok Lee
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Ji-Hye Kang
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.,Dental Medicine Innovation Centre, UCL Eastman-Korea, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jongmin Joseph Kim
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Han-Sem Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.,Dental Medicine Innovation Centre, UCL Eastman-Korea, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Ueon Sang Shin
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea. .,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
13
|
S-ketamine administration in pregnant mice induces ADHD- and depression-like behaviors in offspring mice. Behav Brain Res 2022; 433:113996. [PMID: 35817136 DOI: 10.1016/j.bbr.2022.113996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Anesthesia and psychotropic drugs in pregnant women may cause long-term effects on the brain development of unborn babies. The authors set out to investigate the neurotoxicity of S-ketamine, which possesses anesthetic and antidepressant effects and may cause attention deficit hyperactivity disorder (ADHD)- and depression-like behaviors in offspring mice. METHODS Pregnant mice were administered with low-, medium-, and high-dose S-ketamine (15, 30, and 60 mg/kg) by intraperitoneal injection for 5 days from gestational day 14-18. At 21 days after birth, an elevated plus-maze test, fear conditioning, open field test, and forced swimming test were used to assess ADHD- and depression-like behaviors. Neuronal amount, glial activation, synaptic function indicated by ki67, and inhibitory presynaptic proteins revealed by GAD2 in the hippocampus, amygdala, habenula nucleus, and lateral hypothalamus (LHA) were determined by immunofluorescence assay. RESULTS All the pregnant mice exposed to high-dose S-ketamine administration had miscarriage after the first injection. Both low-dose and medium-dose S-ketamine administration significantly increased the open-arm time and attenuated frozen time in the fear conditioning, which indicates impulsivity and memory dysfunction-like behaviors. Medium-dose S-ketamine administration reduced locomotor activity in the open field and increased immobility time in the forced swimming test, indicating depression-like behaviors. Changes in astrocytic activation, synaptic dysfunction, and decreased inhibitory presynaptic proteins were found in the hippocampus, amygdala, and habenula nucleus. CONCLUSIONS These results demonstrate that S-ketamine may lead to detrimental effects, including ADHD-and depression-like behaviors in offspring mice. More studies should be promoted to determine the neurotoxicity of S-ketamine in the developing brain.
Collapse
|
14
|
Lee JM, Sa M, An H, Kim JMJ, Kwon J, Yoon BE, Lee CJ. Generation of Astrocyte-Specific MAOB Conditional Knockout Mouse with Minimal Tonic GABA Inhibition. Exp Neurobiol 2022; 31:158-172. [PMID: 35786639 PMCID: PMC9272118 DOI: 10.5607/en22016] [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: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
Monoamine oxidase B (MAOB) is a key enzyme for GABA production in astrocytes in several brain regions. To date, the role of astrocytic MAOB has been studied in MAOB null knockout (KO) mice, although MAOB is expressed throughout the body. Therefore, there has been a need for genetically engineered mice in which only astrocytic MAOB is targeted. Here, we generated an astrocyte-specific MAOB conditional KO (cKO) mouse line and characterized it in the cerebellar and striatal regions of the brain. Using the CRISPR-Cas9 gene-editing technique, we generated Maob floxed mice (B6-Maobem1Cjl/Ibs) which have floxed exons 2 and 3 of Maob with two loxP sites. By crossing these mice with hGFAP-CreERT2, we obtained Maob floxed::hGFAP-CreERT2 mice which have a property of tamoxifen-inducible ablation of Maob under the human GFAP (hGFAP) promoter. When we treated Maob floxed::hGFAP-CreERT2 mice with tamoxifen for 5 consecutive days, MAOB and GABA immunoreactivity were significantly reduced in striatal astrocytes as well as in Bergmann glia and lamellar astrocytes in the cerebellum, compared to sunflower oil-injected control mice. Moreover, astrocyte-specific MAOB cKO led to a 74.6% reduction in tonic GABA currents from granule cells and a 76.8% reduction from medium spiny neurons. Our results validate that astrocytic MAOB is a critical enzyme for the synthesis of GABA in astrocytes. We propose that this new mouse line could be widely used in studies of various brain diseases to elucidate the pathological role of astrocytic MAOB in the future.
Collapse
Affiliation(s)
- Jung Moo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Moonsun Sa
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | | | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Bo-Eun Yoon
- Department of Molecular biology, Dankook University, Cheonan 31116, Korea
| | - C. Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| |
Collapse
|
15
|
Labba NA, Wæhler HA, Houdaifi N, Zosen D, Haugen F, Paulsen RE, Hadera MG, Eskeland R. Paracetamol perturbs neuronal arborization and disrupts the cytoskeletal proteins SPTBN1 and TUBB3 in both human and chicken in vitro models. Toxicol Appl Pharmacol 2022; 449:116130. [PMID: 35714712 DOI: 10.1016/j.taap.2022.116130] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/28/2022] [Accepted: 06/10/2022] [Indexed: 11/26/2022]
Abstract
Epidemiological studies have linked long-term/high-dose usage of paracetamol (N-acetyl-para-aminophenol, APAP) during pregnancy to adverse neuropsychiatric outcomes, primarily attention-deficit hyperactive disorder (ADHD), in the offspring. Though variable, ADHD has been associated with phenotypic alterations characterized by reductions in grey matter densities and aberrations in structural connectivity, effects which are thought to originate in neurodevelopment. We used embryonic chicken cerebellar granule neurons (CGNs) and neuronally differentiating human NTERA2 cells (NT2Ns) to investigate the in vitro effects of APAP on cell viability, migration, neuritogenesis, and the intracellular levels of various proteins involved in neurodevelopment as well as in the maintenance of the structure and function of neurites. Exposure to APAP ranging from 100 to 1600 μM yielded concentration- and time-dependent reductions in cell viability and levels of neurite arborization, as well as reductions in the levels of the cytoskeletal protein β2-spectrin, with the highest APAP concentration resulting in between 50 and 75% reductions in the aforementioned metrics over the course of 72 h. Exposure to APAP also reduced migration in the NT2Ns but not CGNs. Moreover, we found concentration- and time-dependent increases in punctate aggregation of the cytoskeletal protein β3-tubulin following exposure to APAP in both cell model systems, with the highest APAP concentration approximately doubling the number of aggregates over 72-120 h. Our findings demonstrate that APAP negatively perturbs neurite arborization degree, with concurrent reductions in the protein levels of β2-spectrin and disruption of the integrity of β3-tubulin, both proteins of which play important roles in neuronal structure and function.
Collapse
Affiliation(s)
- Nils-Anders Labba
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Hallvard Austin Wæhler
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Nora Houdaifi
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Denis Zosen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Fred Haugen
- Department of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Mussie Ghezu Hadera
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway
| | - Ragnhild Eskeland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Norway.
| |
Collapse
|
16
|
Kovács Z, Skatchkov SN, Veh RW, Szabó Z, Németh K, Szabó PT, Kardos J, Héja L. Critical Role of Astrocytic Polyamine and GABA Metabolism in Epileptogenesis. Front Cell Neurosci 2022; 15:787319. [PMID: 35069115 PMCID: PMC8770812 DOI: 10.3389/fncel.2021.787319] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence indicate that astrocytes are essential players of the excitatory and inhibitory signaling during normal and epileptiform activity via uptake and release of gliotransmitters, ions, and other substances. Polyamines can be regarded as gliotransmitters since they are almost exclusively stored in astrocytes and can be released by various mechanisms. The polyamine putrescine (PUT) is utilized to synthesize GABA, which can also be released from astrocytes and provide tonic inhibition on neurons. The polyamine spermine (SPM), synthesized form PUT through spermidine (SPD), is known to unblock astrocytic Cx43 gap junction channels and therefore facilitate astrocytic synchronization. In addition, SPM released from astrocytes may also modulate neuronal NMDA, AMPA, and kainate receptors. As a consequence, astrocytic polyamines possess the capability to significantly modulate epileptiform activity. In this study, we investigated different steps in polyamine metabolism and coupled GABA release to assess their potential to control seizure generation and maintenance in two different epilepsy models: the low-[Mg2+] model of temporal lobe epilepsy in vitro and in the WAG/Rij rat model of absence epilepsy in vivo. We show that SPM is a gliotransmitter that is released from astrocytes and significantly contributes to network excitation. Importantly, we found that inhibition of SPD synthesis completely prevented seizure generation in WAG/Rij rats. We hypothesize that this antiepileptic effect is attributed to the subsequent enhancement of PUT to GABA conversion in astrocytes, leading to GABA release through GAT-2/3 transporters. This interpretation is supported by the observation that antiepileptic potential of the Food and Drug Administration (FDA)-approved drug levetiracetam can be diminished by specifically blocking astrocytic GAT-2/3 with SNAP-5114, suggesting that levetiracetam exerts its effect by increasing surface expression of GAT-2/3. Our findings conclusively suggest that the major pathway through which astrocytic polyamines contribute to epileptiform activity is the production of GABA. Modulation of astrocytic polyamine levels, therefore, may serve for a more effective antiepileptic drug development in the future.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Biology, ELTE Eötvös Loránd University, Savaria University Centre, Szombathely, Hungary
| | - Serguei N. Skatchkov
- Department of Physiology, Universidad Central Del Caribe, Bayamon, PR, United States
- Department of Biochemistry, Universidad Central Del Caribe, Bayamon, PR, United States
| | - Rüdiger W. Veh
- Institut für Zell- und Neurobiologie, Centrum 2, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Krisztina Németh
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Pál T. Szabó
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| |
Collapse
|
17
|
Sieghart W, Chiou LC, Ernst M, Fabjan J, M Savić M, Lee MT. α6-Containing GABA A Receptors: Functional Roles and Therapeutic Potentials. Pharmacol Rev 2022; 74:238-270. [PMID: 35017178 DOI: 10.1124/pharmrev.121.000293] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
GABAA receptors containing the α6 subunit are highly expressed in cerebellar granule cells and less abundantly in many other neuronal and peripheral tissues. Here, we for the first time summarize their importance for the functions of the cerebellum and the nervous system. The cerebellum is not only involved in motor control but also in cognitive, emotional, and social behaviors. α6βγ2 GABAA receptors located at cerebellar Golgi cell/granule cell synapses enhance the precision of inputs required for cerebellar timing of motor activity and are thus involved in cognitive processing and adequate responses to our environment. Extrasynaptic α6βδ GABAA receptors regulate the amount of information entering the cerebellum by their tonic inhibition of granule cells, and their optimal functioning enhances input filtering or contrast. The complex roles of the cerebellum in multiple brain functions can be compromised by genetic or neurodevelopmental causes that lead to a hypofunction of cerebellar α6-containing GABAA receptors. Animal models mimicking neuropsychiatric phenotypes suggest that compounds selectively activating or positively modulating cerebellar α6-containing GABAA receptors can alleviate essential tremor and motor disturbances in Angelman and Down syndrome as well as impaired prepulse inhibition in neuropsychiatric disorders and reduce migraine and trigeminal-related pain via α6-containing GABAA receptors in trigeminal ganglia. Genetic studies in humans suggest an association of the human GABAA receptor α6 subunit gene with stress-associated disorders. Animal studies support this conclusion. Neuroimaging and post-mortem studies in humans further support an involvement of α6-containing GABAA receptors in various neuropsychiatric disorders, pointing to a broad therapeutic potential of drugs modulating α6-containing GABAA receptors. SIGNIFICANCE STATEMENT: α6-Containing GABAA receptors are abundantly expressed in cerebellar granule cells, but their pathophysiological roles are widely unknown, and they are thus out of the mainstream of GABAA receptor research. Anatomical and electrophysiological evidence indicates that these receptors have a crucial function in neuronal circuits of the cerebellum and the nervous system, and experimental, genetic, post-mortem, and pharmacological studies indicate that selective modulation of these receptors offers therapeutic prospects for a variety of neuropsychiatric disorders and for stress and its consequences.
Collapse
Affiliation(s)
- Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Lih-Chu Chiou
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Margot Ernst
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Jure Fabjan
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Miroslav M Savić
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Ming Tatt Lee
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| |
Collapse
|
18
|
Twible C, Abdo R, Zhang Q. Astrocyte Role in Temporal Lobe Epilepsy and Development of Mossy Fiber Sprouting. Front Cell Neurosci 2021; 15:725693. [PMID: 34658792 PMCID: PMC8514632 DOI: 10.3389/fncel.2021.725693] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy affects approximately 50 million people worldwide, with 60% of adult epilepsies presenting an onset of focal origin. The most common focal epilepsy is temporal lobe epilepsy (TLE). The role of astrocytes in the presentation and development of TLE has been increasingly studied and discussed within the literature. The most common histopathological diagnosis of TLE is hippocampal sclerosis. Hippocampal sclerosis is characterized by neuronal cell loss within the Cornu ammonis and reactive astrogliosis. In some cases, mossy fiber sprouting may be observed. Mossy fiber sprouting has been controversial in its contribution to epileptogenesis in TLE patients, and the mechanisms surrounding the phenomenon have yet to be elucidated. Several studies have reported that mossy fiber sprouting has an almost certain co-existence with reactive astrogliosis within the hippocampus under epileptic conditions. Astrocytes are known to play an important role in the survival and axonal outgrowth of central and peripheral nervous system neurons, pointing to a potential role of astrocytes in TLE and associated cellular alterations. Herein, we review the recent developments surrounding the role of astrocytes in the pathogenic process of TLE and mossy fiber sprouting, with a focus on proposed signaling pathways and cellular mechanisms, histological observations, and clinical correlations in human patients.
Collapse
Affiliation(s)
- Carolyn Twible
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada
| | - Rober Abdo
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Qi Zhang
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Pathology and Lab Medicine, London Health Sciences Centre, University Hospital, London, ON, Canada
| |
Collapse
|
19
|
Rudolph S, Guo C, Pashkovski SL, Osorno T, Gillis WF, Krauss JM, Nyitrai H, Flaquer I, El-Rifai M, Datta SR, Regehr WG. Cerebellum-Specific Deletion of the GABA A Receptor δ Subunit Leads to Sex-Specific Disruption of Behavior. Cell Rep 2021; 33:108338. [PMID: 33147470 PMCID: PMC7700496 DOI: 10.1016/j.celrep.2020.108338] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 08/04/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022] Open
Abstract
Granule cells (GCs) of the cerebellar input layer express high-affinity δ GABAA subunit-containing GABAA receptors (δGABAARs) that respond to ambient GABA levels and context-dependent neuromodulators like steroids. We find that GC-specific deletion of δGABAA (cerebellar [cb] δ knockout [KO]) decreases tonic inhibition, makes GCs hyperexcitable, and in turn, leads to differential activation of cb output regions as well as many cortical and subcortical brain areas involved in cognition, anxiety-like behaviors, and the stress response. Cb δ KO mice display deficits in many behaviors, but motor function is normal. Strikingly, δGABAA deletion alters maternal behavior as well as spontaneous, stress-related, and social behaviors specifically in females. Our findings establish that δGABAARs enable the cerebellum to control diverse behaviors not previously associated with the cerebellum in a sex-dependent manner. These insights may contribute to a better understanding of the mechanisms that underlie behavioral abnormalities in psychiatric and neurodevelopmental disorders that display a gender bias. Rudolph et al. show that deletion of the neuromodulator and hormone-sensitive δGABAA receptor subunit from cerebellar granule cells results in anxiety-like behaviors and female-specific deficits in social behavior and maternal care. δGABAA deletion is associated with hyperexcitability of the cerebellar input layer and altered activation of many stress-related brain regions.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Stan L Pashkovski
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tomas Osorno
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Winthrop F Gillis
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy M Krauss
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isabella Flaquer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mahmoud El-Rifai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Lockhofen DEL, Mulert C. Neurochemistry of Visual Attention. Front Neurosci 2021; 15:643597. [PMID: 34025339 PMCID: PMC8133366 DOI: 10.3389/fnins.2021.643597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/12/2021] [Indexed: 11/25/2022] Open
Abstract
Visual attention is the cognitive process that mediates the selection of important information from the environment. This selection is usually controlled by bottom-up and top-down attentional biasing. Since for most humans vision is the dominant sense, visual attention is critically important for higher-order cognitive functions and related deficits are a core symptom of many neuropsychiatric and neurological disorders. Here, we summarize the importance and relative contributions of different neuromodulators and neurotransmitters to the neural mechanisms of top-down and bottom-up attentional control. We will not only review the roles of widely accepted neuromodulators, such as acetylcholine, dopamine and noradrenaline, but also the contributions of other modulatory substances. In doing so, we hope to shed some light on the current understanding of the role of neurochemistry in shaping neuron properties contributing to the allocation of attention in the visual field.
Collapse
Affiliation(s)
| | - Christoph Mulert
- Center for Psychiatry and Psychotherapy, Justus-Liebig University, Hessen, Germany
| |
Collapse
|
21
|
Excessive Astrocytic GABA Causes Cortical Hypometabolism and Impedes Functional Recovery after Subcortical Stroke. Cell Rep 2021; 32:107861. [PMID: 32640227 DOI: 10.1016/j.celrep.2020.107861] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 05/18/2020] [Accepted: 06/12/2020] [Indexed: 11/20/2022] Open
Abstract
Glucose hypometabolism in cortical structures after functional disconnection is frequently reported in patients with white matter diseases such as subcortical stroke. However, the molecular and cellular mechanisms have been poorly elucidated. Here we show, in an animal model of internal capsular infarct, that GABA-synthesizing reactive astrocytes in distant cortical areas cause glucose hypometabolism via tonic inhibition of neighboring neurons. We find that reversal of aberrant astrocytic GABA synthesis, by pharmacological inhibition and astrocyte-specific gene silencing of MAO-B, reverses the reduction in cortical glucose metabolism. Moreover, induction of aberrant astrocytic GABA synthesis by cortical injection of putrescine or adenovirus recapitulates cortical hypometabolism. Furthermore, MAO-B inhibition causes a remarkable recovery from post-stroke motor deficits when combined with a rehabilitation regimen. Collectively, our data indicate that cortical glucose hypometabolism in subcortical stroke is caused by aberrant astrocytic GABA and MAO-B inhibition and that attenuating cortical hypometabolism can be a therapeutic approach in subcortical stroke.
Collapse
|
22
|
McCaffrey TA, St Laurent G, Shtokalo D, Antonets D, Vyatkin Y, Jones D, Battison E, Nigg JT. Biomarker discovery in attention deficit hyperactivity disorder: RNA sequencing of whole blood in discordant twin and case-controlled cohorts. BMC Med Genomics 2020; 13:160. [PMID: 33115496 PMCID: PMC7594430 DOI: 10.1186/s12920-020-00808-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Background A variety of DNA-based methods have been applied to identify genetic markers of attention deficit hyperactivity disorder (ADHD), but the connection to RNA-based gene expression has not been fully exploited. Methods Using well defined cohorts of discordant, monozygotic twins from the Michigan State University Twin Registry, and case-controlled ADHD cases in adolescents, the present studies utilized advanced single molecule RNA sequencing to identify expressed changes in whole blood RNA in ADHD. Multiple analytical strategies were employed to narrow differentially expressed RNA targets to a small set of potential biomarkers of ADHD.
Results RNA markers common to both the discordant twin study and case-controlled subjects further narrowed the putative targets, some of which had been previously associated with ADHD at the DNA level. The potential role of several differentially expressed genes, including ABCB5, RGS2, GAK, GIT1 and 3 members of the galactose metabolism pathway (GALE, GALT, GALK1) are substantiated by prior associations to ADHD and by established mechanistic connections to molecular pathways relevant to ADHD and behavioral control. Conclusions The convergence of DNA, RNA, and metabolic data suggests these may be promising targets for diagnostics and therapeutics in ADHD.
Collapse
Affiliation(s)
- Timothy A McCaffrey
- Division of Genomic Medicine, Department of Medicine, The George Washington University, 2300 Eye St., Washington, DC, 20037, USA. .,The St. Laurent Institute, Vancouver, WA, USA.
| | | | - Dmitry Shtokalo
- The St. Laurent Institute, Vancouver, WA, USA.,A.P. Ershov Institute of Informatics Systems, Novosibirsk, Russia.,AcademGene, LLC, Novosibirsk, Russia
| | - Denis Antonets
- A.P. Ershov Institute of Informatics Systems, Novosibirsk, Russia.,AcademGene, LLC, Novosibirsk, Russia
| | | | | | | | - Joel T Nigg
- Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
23
|
Kim YS, Choi J, Yoon BE. Neuron-Glia Interactions in Neurodevelopmental Disorders. Cells 2020; 9:cells9102176. [PMID: 32992620 PMCID: PMC7601502 DOI: 10.3390/cells9102176] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have revealed synaptic dysfunction to be a hallmark of various psychiatric diseases, and that glial cells participate in synapse formation, development, and plasticity. Glial cells contribute to neuroinflammation and synaptic homeostasis, the latter being essential for maintaining the physiological function of the central nervous system (CNS). In particular, glial cells undergo gliotransmission and regulate neuronal activity in tripartite synapses via ion channels (gap junction hemichannel, volume regulated anion channel, and bestrophin-1), receptors (for neurotransmitters and cytokines), or transporters (GLT-1, GLAST, and GATs) that are expressed on glial cell membranes. In this review, we propose that dysfunction in neuron-glia interactions may contribute to the pathogenesis of neurodevelopmental disorders. Understanding the mechanisms of neuron-glia interaction for synapse formation and maturation will contribute to the development of novel therapeutic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Juwon Choi
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
- Department of Nanobiomedical science, Dankook University, Cheonan 31116, Korea
- Correspondence: ; Tel.: +82-41-529-6085
| |
Collapse
|
24
|
Ferreira S, Pitman KA, Wang S, Summers BS, Bye N, Young KM, Cullen CL. Amyloidosis is associated with thicker myelin and increased oligodendrogenesis in the adult mouse brain. J Neurosci Res 2020; 98:1905-1932. [PMID: 32557778 PMCID: PMC7540704 DOI: 10.1002/jnr.24672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022]
Abstract
In Alzheimer's disease, amyloid plaque formation is associated with the focal death of oligodendrocytes and soluble amyloid β impairs the survival of oligodendrocytes in vitro. However, the response of oligodendrocyte progenitor cells (OPCs) to early amyloid pathology remains unclear. To explore this, we performed a histological, electrophysiological, and behavioral characterization of transgenic mice expressing a pathological form of human amyloid precursor protein (APP), containing three single point mutations associated with the development of familial Alzheimer's disease (PDGFB‐APPSw.Ind, also known as J20 mice). PDGFB‐APPSw.Ind transgenic mice had impaired survival from weaning, were hyperactive by 2 months of age, and developed amyloid plaques by 6 months of age, however, their spatial memory remained intact over this time course. Hippocampal OPC density was normal in P60‐P180 PDGFB‐APPSw.Ind transgenic mice and, by performing whole‐cell patch‐clamp electrophysiology, we found that their membrane properties, including their response to kainate (100 µM), were largely normal. However, by P100, the response of hippocampal OPCs to GABA was elevated in PDGFB‐APPSw.Ind transgenic mice. We also found that the nodes of Ranvier were shorter, the paranodes longer, and the myelin thicker for hippocampal axons in young adult PDGFB‐APPSw.Ind transgenic mice compared with wildtype littermates. Additionally, oligodendrogenesis was normal in young adulthood, but increased in the hippocampus, entorhinal cortex, and fimbria of PDGFB‐APPSw.Ind transgenic mice as pathology developed. As the new oligodendrocytes were not associated with a change in total oligodendrocyte number, these cells are likely required for cell replacement.
Collapse
Affiliation(s)
- Solène Ferreira
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Shiwei Wang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin S Summers
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
25
|
Yang C, Shi Z, You L, Du Y, Ni J, Yan D. Neuroprotective Effect of Catalpol via Anti-Oxidative, Anti-Inflammatory, and Anti-Apoptotic Mechanisms. Front Pharmacol 2020; 11:690. [PMID: 32477145 PMCID: PMC7240050 DOI: 10.3389/fphar.2020.00690] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation and neuro-oxidative damage are now considered to be key factors in the neurological diseases. Therefore, it is important to study anti-inflammatory and neuroprotective agents. The present study investigated the anti-inflammatory and neuroprotective effects of catalpol (CAT), and the potential molecular mechanisms involved. The findings revealed that CAT markedly downregulated pro-inflammatory mediator nitric oxide (NO) and cytokines, including interleukin (IL)-6 and tumor necrosis factor (TNF)-a in lipopolysaccharide (LPS)-treated BV2 microglial cells. Moreover, CAT significantly decreased the levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA), increased superoxide dismutase (SOD) activity and glutathione (GSH) level, reversed apoptosis, and restored mitochondrial membrane potential (MMP) in primary cortical neurons stimulated with hydrogen peroxide (H2O2). Furthermore, mechanistic studies showed that CAT inhibited nuclear factor-κB (NF-κB) pathway and p53-mediated Bcl-2/Bax/casaspe-3 apoptotic pathway. Moreover, it targeted the Kelch-like ECH-associated protein 1(Keap1)/Nuclear factor E2-related factor 2 (Nrf2) pathway. In summary, CAT may exert neuroprotective potential by attenuating microglial-mediated neuroinflammatory response through inhibition of the NF-κB signaling pathway. It blocked cortical neuronal oxidative damage by inhibiting p53-mediated Bcl-2/Bax/casaspe-3 apoptosis pathway and regulating Keap1/Nrf2 pathway. These results collectively indicate the potential of CAT as a highly effective therapeutic agent for neuroinflammatory and neuro-oxidative disorders.
Collapse
Affiliation(s)
- Chunjing Yang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China.,International Cooperation & Joint Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Zhengyuan Shi
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China.,International Cooperation & Joint Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| | - Longtai You
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jian Ni
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Yan
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China.,International Cooperation & Joint Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, China
| |
Collapse
|
26
|
Abstract
Astrocytes, initially described as merely support cells, are now known as a heterogeneous population of cells actively involved in a variety of biological functions such as: neuronal migration and differentiation; regulation of cerebral blood flow; metabolic control of extracellular potassium concentration; and modulation of synapse formation and elimination; among others. Cerebellar glial cells have been shown to play a significant role in proliferation, differentiation, migration, and synaptogenesis. However, less evidence is available about the role of neuron-astrocyte interactions during cerebellar development and their impact on diseases of the cerebellum. In this review, we will focus on the mechanisms underlying cellular interactions, specifically neuron-astrocyte interactions, during cerebellar development, function, and disease. We will discuss how cerebellar glia, astrocytes, and Bergmann glia play a fundamental role in several steps of cerebellar development, such as granule cell migration, axonal growth, neuronal differentiation, and synapse formation, and in diseases associated with the cerebellum. We will focus on how astrocytes and thyroid hormones impact cerebellar development. Furthermore, we will provide evidence of how growth factors secreted by glial cells, such as epidermal growth factor and transforming growth factors, control cerebellar organogenesis. Finally, we will argue that glia are a key mediator of cerebellar development and that identification of molecules and pathways involved in neuron-glia interactions may contribute to a better understanding of cerebellar development and associated disorders.
Collapse
|
27
|
Neurofibromatosis Type 1 Implicates Ras Pathways in the Genetic Architecture of Neurodevelopmental Disorders. Behav Genet 2020; 50:191-202. [DOI: 10.1007/s10519-020-09991-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 01/04/2020] [Indexed: 01/12/2023]
|
28
|
Glial Factors Regulating White Matter Development and Pathologies of the Cerebellum. Neurochem Res 2020; 45:643-655. [PMID: 31974933 PMCID: PMC7058568 DOI: 10.1007/s11064-020-02961-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
The cerebellum is a brain region that undergoes extremely dynamic growth during perinatal and postnatal development which is regulated by the proper interaction between glial cells and neurons with a complex concert of growth factors, chemokines, cytokines, neurotransmitters and transcriptions factors. The relevance of cerebellar functions for not only motor performance but also for cognition, emotion, memory and attention is increasingly being recognized and acknowledged. Since perturbed circuitry of cerebro-cerebellar trajectories can play a role in many central nervous system pathologies and thereby contribute to neurological symptoms in distinct neurodevelopmental and neurodegenerative diseases, is it the aim with this mini-review to highlight the pathways of glia–glia interplay being involved. The designs of future treatment strategies may hence be targeted to molecular pathways also playing a role in development and disease of the cerebellum.
Collapse
|
29
|
Pandit S, Neupane C, Woo J, Sharma R, Nam MH, Lee GS, Yi MH, Shin N, Kim DW, Cho H, Jeon BH, Kim HW, Lee CJ, Park JB. Bestrophin1-mediated tonic GABA release from reactive astrocytes prevents the development of seizure-prone network in kainate-injected hippocampi. Glia 2019; 68:1065-1080. [PMID: 31833596 DOI: 10.1002/glia.23762] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022]
Abstract
Tonic extrasynaptic GABAA receptor (GABAA R) activation is under the tight control of tonic GABA release from astrocytes to maintain the brain's excitation/inhibition (E/I) balance; any slight E/I balance disturbance can cause serious pathological conditions including epileptic seizures. However, the pathophysiological role of tonic GABA release from astrocytes has not been tested in epileptic seizures. Here, we report that pharmacological or genetic intervention of the GABA-permeable Bestrophin-1 (Best1) channel prevented the generation of tonic GABA inhibition, disinhibiting CA1 pyramidal neuronal firing and augmenting seizure susceptibility in kainic acid (KA)-induced epileptic mice. Astrocyte-specific Best1 over-expression in KA-injected Best1 knockout mice fully restored the generation of tonic GABA inhibition and effectively suppressed seizure susceptibility. We demonstrate for the first time that tonic GABA from reactive astrocytes strongly contributes to the compensatory shift of E/I balance in epileptic hippocampi, serving as a good therapeutic target against altered E/I balance in epileptic seizures.
Collapse
Affiliation(s)
- Sudip Pandit
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Chiranjivi Neupane
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Junsung Woo
- Center for Glia-Neuron Interaction and Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Ramesh Sharma
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Ho Nam
- Center for Glia-Neuron Interaction and Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Gyu-Seung Lee
- Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Hee Yi
- Department of Anatomy, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Anatomy, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Anatomy, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Hyunsill Cho
- Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Woo Kim
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Glia-Neuron Interaction and Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jin Bong Park
- Department of Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
30
|
Xie J, Goodbourn PT, Bui BV, Sztal TE, Jusuf PR. Correspondence Between Behavioral, Physiological, and Anatomical Measurements of Visual Function in Inhibitory Neuron–Ablated Zebrafish. ACTA ACUST UNITED AC 2019; 60:4681-4690. [DOI: 10.1167/iovs.19-27544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Jiaheng Xie
- School of Biosciences, The University of Melbourne, Melbourne, Australia
| | - Patrick T. Goodbourn
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Bang V. Bui
- Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Australia
| | - Tamar E. Sztal
- School of Biological Sciences, Monash University, Melbourne, Australia
| | - Patricia R. Jusuf
- School of Biosciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
31
|
Diverse Actions of Astrocytes in GABAergic Signaling. Int J Mol Sci 2019; 20:ijms20122964. [PMID: 31216630 PMCID: PMC6628243 DOI: 10.3390/ijms20122964] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023] Open
Abstract
An imbalance of excitatory and inhibitory neurotransmission leading to over excitation plays a crucial role in generating seizures, while enhancing GABAergic mechanisms are critical in terminating seizures. In recent years, it has been reported in many studies that astrocytes are deeply involved in synaptic transmission. Astrocytes form a critical component of the “tripartite” synapses by wrapping around the pre- and post-synaptic elements. From this location, astrocytes are known to greatly influence the dynamics of ions and transmitters in the synaptic cleft. Despite recent extensive research on excitatory tripartite synapses, inhibitory tripartite synapses have received less attention, even though they influence inhibitory synaptic transmission by affecting chloride and GABA concentration dynamics. In this review, we will discuss the diverse actions of astrocytic chloride and GABA homeostasis at GABAergic tripartite synapses. We will then consider the pathophysiological impacts of disturbed GABA homeostasis at the tripartite synapse.
Collapse
|
32
|
Li X, Zhang J, Niu R, Manthari RK, Yang K, Wang J. Effect of fluoride exposure on anxiety- and depression-like behavior in mouse. CHEMOSPHERE 2019; 215:454-460. [PMID: 30336322 DOI: 10.1016/j.chemosphere.2018.10.070] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/29/2018] [Accepted: 10/11/2018] [Indexed: 05/27/2023]
Abstract
We established the mouse model of fluoride (68 mg F ion/L deionized water) exposure for 90 days, 120 days and 150 days, and applied diverse methods as behavioral models of anxiety and depression, and analyzed the levels of the anxiety- and depression-like related genes like BDNF1, BDNF4, 5-HT1A, VGLUT, GAD67, and VGAT in the mouse hippocampus. In the mice exposed to NaF for 120 days, compared to the control group, chalky opacity was observed on the enamel of teeth; the results of anxiety-like behavior, like elevated zero maze, light/dark exploration test, novel object recognition test and emergence test were significantly altered, however in the mice exposed for 150 days, only the elevated zero maze and emergence test were significantly altered. Also, the results of depression-like behavior were significantly altered in the 120 days treated mice. Exposure to NaF for 120 days significantly decreased the mRNA expression levels of the BDNF4 with a concomitant increase in the 5-HT1A compared to the control mice. Especially the mRNA expression levels of GAD67 and VGAT were significantly decreased in all the three NaF treated groups. However, no significant changes were observed in the mRNA expression levels of the VGLUT compared to the control mice. In summary, we speculated that fluoride exposure had adverse effects on nervous system, inducing an imbalance between excitation and inhibition, which resulted in abnormal behavior and depression.
Collapse
Affiliation(s)
- Xuehua Li
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Jianmeng Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Ruiyan Niu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Ram Kumar Manthari
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Kaidong Yang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China.
| |
Collapse
|
33
|
Kim YS, Jung HM, Yoon BE. Exploring glia to better understand Alzheimer's disease. Anim Cells Syst (Seoul) 2018; 22:213-218. [PMID: 30460100 PMCID: PMC6138241 DOI: 10.1080/19768354.2018.1508498] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
Abstract
The amyloid-β (Aβ) hypothesis has been the leading explanation for the pathogenesis of Alzheimer’s disease (AD). The most common traits of AD are cognitive impairments and memory loss, which are associated with the accumulation of Aβ. Aβ aggregates activate glial cells, which in turn remove Aβ. Because microglia act as immune cells in the brain, most glia-related studies of AD have focused primarily on this cell type. However, astrocytes, another type of glial cell, also participate in the brain immune system, synaptic formation, brain homeostasis, and various other brain functions. Accordingly, many studies on the underlying mechanisms of AD have investigated not only neurons but also glial cells. Although these studies suggest that microglia and astrocytes are effective targets for AD therapeutics, other recent studies have raised questions regarding whether microglial cells and/or astrocytes serve a neuroprotective or neurotoxic function in AD. To gain a better understanding of the mechanisms of AD and identify novel targets for AD treatment, in this review, we consider the role of both microglia and astrocytes in AD.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan, Korea
| | - Hae Myeong Jung
- Department of Molecular Biology, Dankook University, Cheonan, Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan, Korea
| |
Collapse
|
34
|
Kawaharada S, Nakanishi M, Nakanishi N, Hazama K, Higashino M, Yasuhiro T, Lewis A, Clark GS, Chambers MS, Maidment SA, Katsumata S, Kaneko S. ONO-8590580, a Novel GABA Aα5 Negative Allosteric Modulator Enhances Long-Term Potentiation and Improves Cognitive Deficits in Preclinical Models. J Pharmacol Exp Ther 2018; 366:58-65. [PMID: 29674331 DOI: 10.1124/jpet.117.247627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/17/2018] [Indexed: 03/08/2025] Open
Abstract
GABAA receptors containing α5 subunits (GABAAα5) are highly expressed in the hippocampus and negatively involved in memory processing, as shown by the fact that GABAAα5-deficient mice show higher hippocampus-dependent performance than wild-type mice. Accordingly, small-molecule GABAAα5 negative allosteric modulators (NAMs) are known to enhance spatial learning and memory in rodents. Here we introduce a new, orally available GABAAα5 NAM that improves hippocampal functions. ONO-8590580 [1-(cyclopropylmethyl)-5-fluoro-4-methyl-N-[5-(1-methyl-1H-imidazol-4-yl)-2-pyridinyl]-1H-benzimidazol-6-amine] binds to the benzodiazepine binding sites on recombinant human α5-containing GABAA receptors with a Ki of 7.9 nM, and showed functionally selective GABAAα5 NAM activity for GABA-induced Cl- channel activity with a maximum 44.4% inhibition and an EC50 of 1.1 nM. In rat hippocampal slices, tetanus-induced long-term potentiation of CA1 synapse response was significantly augmented in the presence of 300 nM ONO-8590580. Orally administered ONO-8590580 (1-20 mg/kg) dose-dependently occupied hippocampal GABAAα5 in a range of 40%-90% at 1 hour after intake. In the rat passive avoidance test, ONO-8590580 (3-20 mg/kg, by mouth) significantly prevented (+)-MK-801 hydrogen maleate (MK-801)-induced memory deficit. In addition, ONO-8590580 (20 mg/kg, p.o.) was also effective in improving the cognitive deficit induced by scopolamine and MK-801 in the rat eight-arm radial maze test with equal or greater activity than 0.5 mg/kg donepezil. No anxiogenic-like or proconvulsant effect was associated with ONO-8590580 at 20 mg/kg p.o. in the elevated plus maze test or pentylenetetrazole-induced seizure test, respectively. In sum, ONO-8590580 is a novel GABAAα5 NAM that enhances hippocampal memory function without an anxiogenic or proconvulsant risk.
Collapse
Affiliation(s)
- Soichi Kawaharada
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Miki Nakanishi
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Nobuto Nakanishi
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Keisuke Hazama
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Masato Higashino
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Tetsuya Yasuhiro
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Arwel Lewis
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Gary S Clark
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Mark S Chambers
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Scott A Maidment
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Seishi Katsumata
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Shuji Kaneko
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| |
Collapse
|