1
|
Xin Q, Wang J, Zheng J, Tan Y, Jia X, Ni Z, Xu Z, Feng J, Wu Z, Li Y, Li XM, Ma H, Hu H. Neuron-astrocyte coupling in lateral habenula mediates depressive-like behaviors. Cell 2025; 188:3291-3309.e24. [PMID: 40280131 DOI: 10.1016/j.cell.2025.04.010] [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: 09/08/2024] [Revised: 01/08/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
The lateral habenula (LHb) neurons and astrocytes have been strongly implicated in depression etiology, but it was not clear how the two dynamically interact during depression onset. Here, using multi-brain-region calcium photometry recording in freely moving mice, we discover that stress induces a most rapid astrocytic calcium rise and a bimodal neuronal response in the LHb. LHb astrocytic calcium requires the α1A-adrenergic receptor and depends on a recurrent neural network between the LHb and locus coeruleus (LC). Through the gliotransmitter glutamate and ATP/adenosine, LHb astrocytes mediate the second-wave LHb neuronal activation and norepinephrine (NE) release. Activation or inhibition of LHb astrocytic calcium signaling facilitates or prevents stress-induced depressive-like behaviors, respectively. These results identify a stress-induced positive feedback loop in the LHb-LC axis, with astrocytes being a critical signaling relay. The identification of this prominent neuron-glia interaction may shed light on stress management and depression prevention.
Collapse
Affiliation(s)
- Qianqian Xin
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Junying Wang
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Jinkun Zheng
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Yi Tan
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Psychiatry and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiaoning Jia
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Zheyi Ni
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Zijie Xu
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Xiao-Ming Li
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Huan Ma
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Hailan Hu
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Psychiatry and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China.
| |
Collapse
|
2
|
Kim KJ, Lee JH, Lim J, Lee T, Joo J, Bhalla M, Wang T, Feng R, Lee CJ. Astrocyte-Specific Phenotyping of FAD 4T as an Alzheimer's Disease Mouse Model. Glia 2025; 73:1258-1271. [PMID: 40009021 DOI: 10.1002/glia.70002] [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: 11/22/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, characterized by memory decline and behavioral changes. Its pathological features include senile plaques, neurofibrillary tangles, and reactive gliosis, comprising abnormal accumulations of β-amyloid peptide (Aβ) and hyperphosphorylated tau protein surrounded by reactive astrocytes and microglia. Recently, it has emerged that severe reactive astrocytes and MAOB-dependent production of GABA and H2O2 are the real causes of learning and memory impairment and neurodegeneration. Diverse mouse models for AD have been developed to clarify pathological mechanisms and discover therapeutic strategies and drugs. However, there are many shortfalls and discrepancies among them. A new AD mouse model named FAD4T has been developed to overcome various shortcomings. Here, we employed astrocyte-focused screening procedures to examine the pathological features of FAD4T as an AD model. Our results revealed that the FAD4T mice showed abnormal accumulation of Aβ plaques in overall brain regions at 6 and 12 months. We found astrocytic hypertrophy with a significant elevation of GFAP and LCN2. However, the expressions of MAOB and iNOS, a severe reactive astrocyte marker, were unchanged. Electrophysiological and behavioral analysis indicated aberrant tonic GABA release, reduced neuronal activity, and impaired CA1-specific memory. These findings demonstrate that FAD4T mice mimic pathological and functional features of AD, different from other AD mouse models. These findings demonstrate that FAD4T mimics some features of AD patients but lacks other important features, such as severe reactive astrocytes and neurodegeneration. This astrocyte-focused screening method offers valuable tools for advancing AD research and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Ki Jung Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Taehee Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jinhyeong Joo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Tao Wang
- GemPharmatech Co Ltd., Guangdong, China
| | - Rui Feng
- GemPharmatech Co. Ltd., Nanjing, China
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
3
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
4
|
Kim KW, Lee E, Ko A, Hwang J, Park K, Lee BC, Kim KW, Oh WJ, Kim K, Lim HH. Cryo-EM structures of mouse bestrophin 1 channel in closed and partially open conformations. Mol Cells 2025; 48:100208. [PMID: 40043778 PMCID: PMC11964753 DOI: 10.1016/j.mocell.2025.100208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Bestrophin 1 (BEST1) channels are calcium-activated Cl- channels involved in diverse physiological processes, including gliotransmitter release in astrocytes. Although human and chicken BEST1 orthologs have been extensively studied, the structural and functional properties of mouse BEST1 (mBEST1) remain poorly understood. In this study, we characterized the structure-function of mBEST1-BF, a C-terminally tagged variant, using whole-cell patch-clamp recordings, surface biotinylation assays, and single-particle cryo-electron microscopy. Cryo-electron microscopy structural analysis of mBEST1-BF revealed closed and partially open conformations. Comparative analysis with human and chicken BEST1 orthologs highlighted conserved calcium-binding and gating mechanisms, with distinct features in mBEST1, including a wider aperture sufficient to accommodate dehydrated Cl- ions and potential anion-binding sites near Val205 and Gln208 residues. The disordered C-terminal region of mBEST1 remains unresolved, suggesting it may require stabilizing factors for structural determination. Additionally, the autoinhibitory domain, which includes Ser354, likely plays a key role in regulating gating, with Ser354 potentially serving as a phosphorylation site that modulates channel activity. Our findings provide structural and functional insights into mBEST1 and suggest mechanisms underlying its unique gating and ion permeation properties.
Collapse
Affiliation(s)
- Kwon-Woo Kim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Euna Lee
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Ara Ko
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Junmo Hwang
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Kunwoong Park
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Byoung-Cheol Lee
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Ki Woo Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Kyuhyung Kim
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
5
|
Gonzalez L, Bezzi P. Astrocyte Dysfunctions in Obsessive Compulsive Disorder: Rethinking Neurobiology and Therapeutic Targets. J Neurochem 2025; 169:e70092. [PMID: 40400176 PMCID: PMC12095986 DOI: 10.1111/jnc.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Obsessive-compulsive disorder (OCD) has long been conceptualized as a neuron-centric disorder of cortico-striato-thalamo-cortical (CSTC) circuit dysregulation. However, a growing body of evidence is now reframing this narrative, placing astrocytes-once relegated to passive support roles-at the center of OCD pathophysiology. Astrocytes are critical regulators of glutamate and GABA homeostasis, calcium signaling, and synaptic plasticity, all of which are disrupted in OCD. Recent high-resolution molecular and proteomic studies reveal that specific astrocyte subpopulations, including Crym-positive astrocytes, directly shape excitatory/inhibitory balance and control perseverative behaviors by modulating presynaptic inputs from the orbitofrontal cortex. Disruptions in astrocytic neurotransmitter clearance and dopamine metabolism amplify CSTC circuit hyperactivity and reinforce compulsions. This review reframes OCD as a disorder of neuro-glial dysfunctions, proposing that targeting astrocytic signaling, metabolism, and structural plasticity may unlock transformative therapeutic strategies. By integrating human and animal data, we advocate for a glial-centric model of OCD that not only enhances mechanistic understanding but also opens new frontiers for precision treatment.
Collapse
Affiliation(s)
- Laurine Gonzalez
- Department of Fundamental Neurosciences (DNF)University of Lausanne (UNIL)LausanneSwitzerland
| | - Paola Bezzi
- Department of Fundamental Neurosciences (DNF)University of Lausanne (UNIL)LausanneSwitzerland
- Department of Physiology and PharmacologyUniversity of Rome SapienzaRomeItaly
| |
Collapse
|
6
|
Pant S, Tam SW, Long SB. The pentameric chloride channel BEST1 is activated by extracellular GABA. Proc Natl Acad Sci U S A 2025; 122:e2424474122. [PMID: 40249777 PMCID: PMC12037058 DOI: 10.1073/pnas.2424474122] [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: 11/22/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025] Open
Abstract
Bestrophin-1 (BEST1) is a chloride channel expressed in the eye and other tissues of the body. A link between BEST1 and the principal inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been proposed. The most appreciated receptors for extracellular GABA are the GABAB G-protein-coupled receptors and the pentameric GABAA chloride channels, both of which have fundamental roles in the central nervous system. Here, we demonstrate that BEST1 is directly activated by GABA. Through functional studies and atomic-resolution structures of human and chicken BEST1, we identify a GABA binding site on the channel's extracellular side and determine the mechanism by which GABA binding stabilizes opening of the channel's central gate. This same gate, "the neck," is activated by intracellular [Ca2+], indicating that BEST1 is controlled by ligands from both sides of the membrane. The studies demonstrate that BEST1, which shares no structural homology with GABAA receptors, is a GABA-activated chloride channel. The physiological implications of this finding remain to be studied.
Collapse
Affiliation(s)
- Swati Pant
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephanie W. Tam
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
| |
Collapse
|
7
|
Al Tannir R, Pautrat A, Soutrenon R, Overton P, Coizet V. Brainstem Neuroadaptations in Rodent Models of Parkinson's Disease. Eur J Neurosci 2025; 61:e70068. [PMID: 40162834 PMCID: PMC11956546 DOI: 10.1111/ejn.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/11/2025] [Accepted: 01/22/2025] [Indexed: 04/02/2025]
Abstract
A classical theory of a key pathophysiological change in Parkinson's disease (PD) is that GABAergic neurons in the substantia nigra pars reticulata (SNr), an output structure of the basal ganglia, become hyperactive following the dopaminergic loss. Increased GABA release from the SNr neurons is thus likely to induce neuroadaptations in structures receiving a direct projection from the SNr, including the parabrachial nucleus (PBN), superior colliculus (SC), and periaqueductal gray (PAG). We have shown that the PBN indeed exhibits cellular and molecular changes in PD rat models. We thus expected the SC and the PAG to likewise show neuroplasticity. The objective of the present work was to evaluate the cellular and molecular plasticity in both the SC (lateral and medial) and the PAG in PD rats with a partial or total dopaminergic lesion. We used Golgi-Cox to measure the spine density and spine morphology and Western blot to analyze GABAA receptor expression in both PD rat models compared to sham animals. We found an increase in spine density (thin and stubby types) following total dopaminergic lesions in the SC and the PAG. Additionally, increased GABAA receptor expression was observed in the lateral SC in the total lesion group only. These results suggest compensatory mechanisms in PD that may delay disease onset and contribute to both motor and nonmotor symptoms. Further investigation should be performed to fully understand the functional impact of the plasticity revealed in this work.
Collapse
Affiliation(s)
- Racha Al Tannir
- Université Grenoble Alpes, Inserm U1216, CHU Grenoble Alpes, Grenoble Institut NeurosciencesGrenobleFrance
| | - Arnaud Pautrat
- Université Grenoble Alpes, Inserm U1216, CHU Grenoble Alpes, Grenoble Institut NeurosciencesGrenobleFrance
| | - Remi Soutrenon
- Université Grenoble Alpes, Inserm U1216, CHU Grenoble Alpes, Grenoble Institut NeurosciencesGrenobleFrance
| | | | - Veronique Coizet
- Université Grenoble Alpes, Inserm U1216, CHU Grenoble Alpes, Grenoble Institut NeurosciencesGrenobleFrance
| |
Collapse
|
8
|
Zhou B, Li Q, Su M, Liao P, Luo Y, Luo R, Yu Y, Luo M, Lei F, Li X, Jiao J, Yi L, Wang J, Yang L, Liao D, Zhou C, Zhang X, Xiao H, Zuo Y, Liu J, Zhu T, Jiang R. Astrocyte morphological remodeling regulates consciousness state transitions induced by inhaled general anesthesia. Mol Psychiatry 2025:10.1038/s41380-025-02978-2. [PMID: 40169801 DOI: 10.1038/s41380-025-02978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
General anesthetics (GAs) are conventionally thought to induce loss of consciousness (LOC) by acting on pre- and post-synaptic targets. However, the mechanism underlying the involvement of astrocytes in LOC remains unclear. Here we report that inhaled GAs cause reversible impairments in the fine processes of astrocytes within the somatosensory cortex, mediated by regulating the phosphorylation level of Ezrin, a protein critical for the fine morphology of astrocytes. Genetically deleting Ezrin or disrupting its phosphorylation was sufficient to decrease astrocyte-synapse interaction and enhance sensitivity to sevoflurane (Sevo) in vivo. Moreover, we show that disrupting astrocytic Ezrin phosphorylation boosted the inhibitory effect of Sevo on pyramidal neurons by enhancing tonic GABA and lowering excitability under anesthesia. Our work reveals previously unappreciated phosphorylation-dependent morphological dynamics, which enable astrocytes to regulate neuronal activity during the transition between two brain consciousness states.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingran Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengchan Su
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuncheng Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunqing Yu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meiyan Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Lei
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Xin Li
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Jiao Jiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Limei Yi
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xia Zhang
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Xiao
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Xiong S, Sun M, Zhang Y, Kong PR, Gan L, Gao L, Xu K, Wu HY, Zhu DY, Lin YH, Li R, Luo CX. Astrocytic BEST1 can serve as a target for functional recovery after ischemic stroke. Mol Ther 2025:S1525-0016(25)00196-0. [PMID: 40119514 DOI: 10.1016/j.ymthe.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/23/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Solid evidence from animal experiments supported the concept of peri-infarct tonic inhibition. Related drug targets have the potential to be translated for clinical stroke treatment. Recently, we reported the contribution of neuronal bestrophin-1 (BEST1)-mediated glutamate release to acute ischemic damage exacerbation in rodents. Now, we found a switch of abnormal BEST1 expression and function from neurons to astrocytes in the peri-infarct cortex following astrocytic activation. Excessive GABA was released through astrocytic BEST1 channel during the subacute phase of stroke, leading to sustained tonic inhibition. Astrocyte-specific knockdown of BEST1 promoted motor functional recovery, depending on reduced tonic inhibition. Moreover, we prepared self-assembled nanoparticles encapsulating siBest1 (SNP-siBest1), which displayed high brain accumulation and long circulation and knocked down astrocytic BEST1 effectively and safely. Systemic treatment with SNP-siBest1 after ischemic stroke showed a therapeutic effect in mice. Therefore, BEST1 is a potential target for stroke therapy from acute to subacute phase, and selective BEST1 blockers beyond nanoparticles are worth developing.
Collapse
Affiliation(s)
- Shuai Xiong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meng Sun
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Pei-Ran Kong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lu Gan
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Gao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Xu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Rui Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
10
|
Gu J, Wang J, Fan H, Wei Y, Li Y, Ma C, Xing K, Wang P, Wu Z, Wu T, Li X, Zhang L, Han Y, Chen T, Qu J, Yan X. Decoding the mechanism of proanthocyanidins in central analgesia: redox regulation and KCNK3 blockade. Exp Mol Med 2025; 57:567-583. [PMID: 40025170 PMCID: PMC11958645 DOI: 10.1038/s12276-025-01412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 03/04/2025] Open
Abstract
Neuropathic pain causes enduring physical discomfort and emotional distress. Conventional pharmacological treatments often provide restricted relief and may result in undesirable side effects, posing a substantial clinical challenge. Peripheral and spinal redox homeostasis plays an important role in pain processing and perception. However, the roles of oxidative stress and antioxidants in pain and analgesia on the cortical region during chronic pain remains obscure. Here we focus on the ventrolateral orbital cortex (VLO), a brain region associated with pain severity and involved in pain inhibition. Using a spared nerve injury mouse model, we observed the notable reactive oxygen species (ROS)-mediated suppression of the excitability of pyramidal cells (PYRVLO) in the VLO. Nasal application or microinjection of the natural antioxidants proanthocyanidins (PACs) to the VLO specifically increased the activity of PYRVLO and induced a significant analgesic effect. Mechanistically, PACs activate PYRVLO by inhibiting distinct potassium channels in different ways: (1) by scavenging ROS to reduce ROS-sensitive voltage-gated potassium currents and (2) by acting as a channel blocker through direct binding to the cap structure of KCNK3 to inhibit the leak potassium current (Ileak). These results reveal the role of cortical oxidative stress in central hyperalgesia and elucidate the mechanism and potential translational significance of PACs in central analgesia. These findings suggest that the effects of PACs extend beyond their commonly assumed antioxidant or anti-inflammatory effects.
Collapse
Affiliation(s)
- Junxiang Gu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Jian Wang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongwei Fan
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wei
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- School of Medicine, Northwest University, Xi'an, China
| | - Yan Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chengwen Ma
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Keke Xing
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Pan Wang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Zhenyu Wu
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Teng Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.
| | - Jianqiang Qu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xianxia Yan
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
11
|
Chiou LC, Sieghart W. IUPHAR Review: Alpha6-containing GABA A receptors - Novel targets for the treatment of schizophrenia. Pharmacol Res 2025; 213:107613. [PMID: 39848349 DOI: 10.1016/j.phrs.2025.107613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
α6-containing GABAA receptors (α6GABAARs) are strongly expressed in cerebellar granule cells and are of central importance for cerebellar functions. The cerebellum not only is involved in regulation of motor activity, but also in regulation of thought, cognition, emotion, language, and social behavior. Activation of α6GABAARs enhances the precision of sensory inputs, enables rapid and coordinated movement and adequate responses to the environment, and protects the brain from information overflow. The cerebellum has strong connections to multiple brain regions via closed loop circuits and is also extensively connected with the dopamine system in the prefrontal cortex, that initiates the execution of behavior. Patients suffering from schizophrenia exhibit an impaired structure and function of the cerebellum and an impaired GABAergic transmission at α6GABAARs. This also impairs the function of the dopamine system, can explain a variety of schizophrenia symptoms observed, and might be one of the pathophysiological causes of schizophrenia. Enhancing GABAergic transmission at α6GABAARs should thus reduce the symptoms of schizophrenia. This recently has been confirmed by demonstrating that positive allosteric modulators with high selectivity for α6GABAARs can reduce positive and negative symptoms and cognitive impairment of schizophrenia in several animal models of this disorder. So far, the beneficial actions of these modulators have been demonstrated in animal models of neuropsychiatric disorders, only. Future human studies have to investigate the safety and possible side effects of these modulators and to clarify, to which extent individual symptoms of schizophrenia can be reduced by these drugs in patients during acute and chronic dosing.
Collapse
Affiliation(s)
- Lih-Chu Chiou
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
12
|
Kim SH, Lee J, Jang M, Roh SE, Kim S, Lee JH, Seo J, Baek J, Hwang JY, Baek IS, Lee YS, Shigetomi E, Lee CJ, Koizumi S, Kim SK, Kim SJ. Cerebellar Bergmann glia integrate noxious information and modulate nocifensive behaviors. Nat Neurosci 2025; 28:336-345. [PMID: 39748107 DOI: 10.1038/s41593-024-01807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/27/2024] [Indexed: 01/04/2025]
Abstract
The cerebellum is activated by noxious stimuli and pathological pain but its role in noxious information processing remains unknown. Here, we show that in mice, cutaneous noxious electrical stimuli induced noradrenaline (NA) release from locus coeruleus (LC) terminals in the cerebellar cortex. Bergmann glia (BG) accumulated these LC-NA signals by increasing intracellular calcium in an integrative manner ('flares'). BG flares were also elicited in response to an intraplantar capsaicin injection. Chemogenetic inactivation of LC terminals or BG in the cerebellar cortex or BG-specific knockdown of α1-adrenergic receptors suppressed BG flares, reduced nocifensive licking and had analgesic effects in nerve injury-induced chronic neuropathic pain. Moreover, chemogenetic activation of BG or an intraplantar capsaicin injection reduced Purkinje cell firing, which may disinhibit the output activity of the deep cerebellar nuclei. These results suggest a role for BG in computing noxious information from the LC and in modulating pain-related behaviors by regulating cerebellar output.
Collapse
Affiliation(s)
- Seung Ha Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jaegeon Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Mirae Jang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Eon Roh
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soobin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hwan Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jewoo Seo
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jinhee Baek
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Yoon Hwang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - In Seon Baek
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Memory Network Medical Research Center, Neuroscience Research Institute, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Korea
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea.
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Memory Network Medical Research Center, Neuroscience Research Institute, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
13
|
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
|
14
|
Juvenal G, Higa GSV, Bonfim Marques L, Tessari Zampieri T, Costa Viana FJ, Britto LR, Tang Y, Illes P, di Virgilio F, Ulrich H, de Pasquale R. Regulation of GABAergic neurotransmission by purinergic receptors in brain physiology and disease. Purinergic Signal 2025; 21:149-177. [PMID: 39046648 PMCID: PMC11958915 DOI: 10.1007/s11302-024-10034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Purinergic receptors regulate the processing of neural information in the hippocampus and cerebral cortex, structures related to cognitive functions. These receptors are activated when astrocytic and neuronal populations release adenosine triphosphate (ATP) in an autocrine and paracrine manner, following sustained patterns of neuronal activity. The modulation by these receptors of GABAergic transmission has only recently been studied. Through their ramifications, astrocytes and GABAergic interneurons reach large groups of excitatory pyramidal neurons. Their inhibitory effect establishes different synchronization patterns that determine gamma frequency rhythms, which characterize neural activities related to cognitive processes. During early life, GABAergic-mediated synchronization of excitatory signals directs the experience-driven maturation of cognitive development, and dysfunctions concerning this process have been associated with neurological and neuropsychiatric diseases. Purinergic receptors timely modulate GABAergic control over ongoing neural activity and deeply affect neural processing in the hippocampal and neocortical circuitry. Stimulation of A2 receptors increases GABA release from presynaptic terminals, leading to a considerable reduction in neuronal firing of pyramidal neurons. A1 receptors inhibit GABAergic activity but only act in the early postnatal period when GABA produces excitatory signals. P2X and P2Y receptors expressed in pyramidal neurons reduce the inhibitory tone by blocking GABAA receptors. Finally, P2Y receptor activation elicits depolarization of GABAergic neurons and increases GABA release, thus favoring the emergence of gamma oscillations. The present review provides an overall picture of purinergic influence on GABAergic transmission and its consequences on neural processing, extending the discussion to receptor subtypes and their involvement in the onset of brain disorders, including epilepsy and Alzheimer's disease.
Collapse
Affiliation(s)
- Guilherme Juvenal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Guilherme Shigueto Vilar Higa
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lucas Bonfim Marques
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Thais Tessari Zampieri
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe José Costa Viana
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luiz R Britto
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Yong Tang
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany
| | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Roberto de Pasquale
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
15
|
Palacios ER, Houghton C, Chadderton P. GlyT2-Positive Interneurons Regulate Timing and Variability of Information Transfer in a Cerebellar-Behavioral Loop. J Neurosci 2025; 45:e1568242024. [PMID: 39658258 PMCID: PMC11780355 DOI: 10.1523/jneurosci.1568-24.2024] [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: 08/13/2024] [Revised: 10/24/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
GlyT2-positive interneurons, Golgi and Lugaro cells, reside in the input layer of the cerebellar cortex in a key position to influence information processing. Here, we examine the contribution of GlyT2-positive interneurons to network dynamics in Crus 1 of mouse lateral cerebellar cortex during free whisking. We recorded neuronal population activity using Neuropixels probes before and after chemogenetic downregulation of GlyT2-positive interneurons in male and female mice. Under resting conditions, cerebellar population activity reliably encoded whisker movements. Reductions in the activity of GlyT2-positive cells produced mild increases in neural activity which did not significantly impair these sensorimotor representations. However, reduced Golgi and Lugaro cell inhibition did increase the temporal alignment of local population network activity at the initiation of movement. These network alterations had variable impacts on behavior, producing both increases and decreases in whisking velocity. Our results suggest that inhibition mediated by GlyT2-positive interneurons primarily governs the temporal patterning of population activity, which in turn is required to support downstream cerebellar dynamics and behavioral coordination.
Collapse
Affiliation(s)
- Ensor Rafael Palacios
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
- School of Engineering Mathematics and Technology, University of Bristol, Bristol BS8 1UB, United Kingdom
| | - Conor Houghton
- School of Engineering Mathematics and Technology, University of Bristol, Bristol BS8 1UB, United Kingdom
| | - Paul Chadderton
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
16
|
Bhalla M, Joo J, Kim D, Shin JI, Park YM, Ju YH, Park U, Yoo S, Hyeon SJ, Lee H, Lee J, Ryu H, Lee CJ. SIRT2 and ALDH1A1 as critical enzymes for astrocytic GABA production in Alzheimer's disease. Mol Neurodegener 2025; 20:6. [PMID: 39815261 PMCID: PMC11734448 DOI: 10.1186/s13024-024-00788-8] [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: 07/10/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disease with drastically altered astrocytic metabolism. Astrocytic GABA and H2O2 are associated with memory impairment in AD and synthesized through the Monoamine Oxidase B (MAOB)-mediated multi-step degradation of putrescine. However, the enzymes downstream to MAOB in this pathway remain unidentified. METHODS Using transcriptomics analysis, we identified two candidate enzymes, Aldehyde Dehydrogenase 1 family member A1 (ALDH1A1) and Sirtuin 2 (SIRT2) for the steps following MAOB in the astrocytic GABA production pathway. We used immunostaining, metabolite analysis and electrophysiology, both in vitro and in vivo, to confirm the participation of these enzymes in astrocytic GABA production. We checked for the presence of SIRT2 in human AD patients as well as the mouse model APP/PS1 and finally, we selectively ablated SIRT2 in the astrocytes of APP/PS1 mice to observe its effects on pathology. RESULTS Immunostaining, metabolite analysis, and electrophysiology recapitulated the participation of ALDH1A1 and SIRT2 in GABA production. Inhibition of SIRT2 reduced the production of astrocytic GABA but not H2O2, a key molecule in neurodegeneration. Elevated expression of these enzymes was found in hippocampal astrocytes of AD patients and APP/PS1 mice. Astrocyte-specific gene-silencing of SIRT2 in APP/PS1 mice restored GABA production and partially improved memory function. CONCLUSIONS Our study is the first to identify the specific role of SIRT2 in reactive astrogliosis and determine the specific pathway and metabolic step catalyzed by the enzyme. We determine the partial, yet significant role of ALDH1A1 in this process, thereby highlighting 2 new players the astrocytic GABA production pathway. Our findings therefore, offer SIRT2 as a new tool to segregate GABA from H2O2 production, aiding future research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mridula Bhalla
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jinhyeong Joo
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Daeun Kim
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jeong Im Shin
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- Division of Bio-Medical Science & Technology, Department of Neuroscience, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Yongmin Mason Park
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yeon Ha Ju
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Uiyeol Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seonguk Yoo
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyunbeom Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, USA.
- VA Boston Healthcare System, Boston, USA.
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea.
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
17
|
Park GY, Lee G, Yoon J, Han J, Choi P, Kim M, Lee S, Park C, Wu Z, Li Y, Choi M. Glia-like taste cells mediate an intercellular mode of peripheral sweet adaptation. Cell 2025; 188:141-156.e16. [PMID: 39561773 DOI: 10.1016/j.cell.2024.10.041] [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: 09/06/2023] [Revised: 06/30/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024]
Abstract
The sense of taste generally shows diminishing sensitivity to prolonged sweet stimuli, referred to as sweet adaptation. Yet, its mechanistic landscape remains incomplete. Here, we report that glia-like type I cells provide a distinct mode of sweet adaptation via intercellular crosstalk with chemosensory type II cells. Using the microfluidic-based intravital tongue imaging system, we found that sweet adaptation is facilitated along the synaptic transduction from type II cells to gustatory afferent nerves, while type I cells display temporally delayed and prolonged activities. We identified that type I cells receive purinergic input from adjacent type II cells via P2RY2 and provide inhibitory feedback to the synaptic transduction of sweet taste. Aligning with our cellular-level findings, purinergic activation of type I cells attenuated sweet licking behavior, and P2RY2 knockout mice showed decelerated adaptation behavior. Our study highlights a veiled intercellular mode of sweet adaptation, potentially contributing to the efficient encoding of prolonged sweetness.
Collapse
Affiliation(s)
- Gha Yeon Park
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Geehyun Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Jongmin Yoon
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Jisoo Han
- Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Pyonggang Choi
- Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Minjae Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Sungho Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Chaeri Park
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Myunghwan Choi
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea; The Institute of Molecular Biology and Genetics, Seoul 08826, Republic of Korea.
| |
Collapse
|
18
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
19
|
Zinsmaier AK, Nestler EJ, Dong Y. Astrocytic G Protein-Coupled Receptors in Drug Addiction. ENGINEERING (BEIJING, CHINA) 2025; 44:256-265. [PMID: 40109668 PMCID: PMC11922559 DOI: 10.1016/j.eng.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Understanding the cellular mechanisms of drug addiction remains a key task in current brain research. While neuron-based mechanisms have been extensively explored over the past three decades, recent evidence indicates a critical involvement of astrocytes, the main type of non-neuronal cells in the brain. In response to extracellular stimuli, astrocytes modulate the activity of neurons, synaptic transmission, and neural network properties, collectively influencing brain function. G protein-coupled receptors (GPCRs) expressed on astrocyte surfaces respond to neuron- and environment-derived ligands by activating or inhibiting astrocytic signaling, which in turn regulates adjacent neurons and their circuitry. In this review, we focus on the dopamine D1 receptors (D1R) and metabotropic glutamate receptor 5 (mGLUR5 or GRM5)-two GPCRs that have been critically implicated in the acquisition and maintenance of addiction-related behaviors. Positioned as an introductory-level review, this article briefly discusses astrocyte biology, outlines earlier discoveries about the role of astrocytes in substance-use disorders (SUDs), and provides detailed discussion about astrocytic D1Rs and mGLUR5s in regulating synapse and network functions in the nucleus accumbens (NAc)-a brain region that mediates addiction-related emotional and motivational responses. This review serves as a stepping stone for readers of Engineering to explore links between astrocytic GPCRs and drug addiction and other psychiatric disorders.
Collapse
Affiliation(s)
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
20
|
Correa-da-Silva F, Yi CX. Neuroglia in eating disorders (obesity, Prader-Willi syndrome and anorexia nervosa). HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:313-324. [PMID: 40148052 DOI: 10.1016/b978-0-443-19102-2.00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The hypothalamus is widely recognized as one of the most extensively studied brain regions involved in the central regulation of energy homeostasis. Within the hypothalamus, peptidergic neurons play a crucial role in monitoring peripheral concentrations of metabolites and hormones, and they finely adjust the sensing of these factors, leading to the activation of either anorexigenic (appetite-suppressing) or orexigenic (appetite-stimulating) pathways. While cortical innervation of the hypothalamus does influence these processes, it is generally considered of secondary importance. Eating-related disorders, such as obesity and anorexia nervosa, are strongly associated with imbalances in energy intake and expenditure. The phenotypes of these disorders can be attributed to dysfunctions in the hypothalamus. Traditionally, it has been believed that hypothalamic dysfunction in these disorders primarily stems from defects in neural pathways. However, recent evidence challenges this perception, highlighting the active participation of neuroglial cells in shaping both physiologic and behavioral characteristics. This review aims to provide an overview of the latest insights into glial biology in three specific eating disorders: obesity, Prader-Willi syndrome, and anorexia. In these disorders, neural dysfunction coincides with glial malfunction, suggesting that neuroglia actively contribute to the development and progression of various neurologic disorders. These findings underscore the importance of glial cells and open up potential new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Ness N, Díaz-Clavero S, Hoekstra MMB, Brancaccio M. Rhythmic astrocytic GABA production synchronizes neuronal circadian timekeeping in the suprachiasmatic nucleus. EMBO J 2025; 44:356-381. [PMID: 39623138 PMCID: PMC11731042 DOI: 10.1038/s44318-024-00324-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 01/15/2025] Open
Abstract
Astrocytes of the suprachiasmatic nucleus (SCN) can regulate sleep-wake cycles in mammals. However, the nature of the information provided by astrocytes to control circadian patterns of behavior is unclear. Neuronal circadian activity across the SCN is organized into spatiotemporal waves that govern seasonal adaptations and timely engagement of behavioral outputs. Here, we show that astrocytes across the mouse SCN exhibit instead a highly uniform, pulse-like nighttime activity. We find that rhythmic astrocytic GABA production via polyamine degradation provides an inhibitory nighttime tone required for SCN circuit synchrony, thereby acting as an internal astrocyte zeitgeber (or "astrozeit"). We further identify synaptic GABA and astrocytic GABA as two key players underpinning coherent spatiotemporal circadian patterns of SCN neuronal activity. In describing a new mechanism by which astrocytes contribute to circadian timekeeping, our work provides a general blueprint for understanding how astrocytes encode temporal information underlying complex behaviors in mammals.
Collapse
Affiliation(s)
- Natalie Ness
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Sandra Díaz-Clavero
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Marieke M B Hoekstra
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Marco Brancaccio
- Department of Brain Science, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| |
Collapse
|
22
|
Cheong E, Lee CJ. Gliotransmission in physiologic and pathologic conditions. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:93-116. [PMID: 40122634 DOI: 10.1016/b978-0-443-19104-6.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter explores the roles of gliotransmission in physiologic and pathologic conditions, including psychiatric and neurologic disorders. Gliotransmission, facilitated by astrocytes through the release of gliotransmitters such as glutamate, d-serine, and GABA, regulates neuronal activity and synaptic transmission. Under physiologic conditions, astrocytic gliotransmission maintains the balance of tonic excitation and inhibition, influencing synaptic plasticity and cognitive functions. In psychiatric disorders, the chapter examines how dysregulated gliotransmission contributes to major depression and schizophrenia. In major depression, changes in astrocytic glutamate and adenosine signaling impact mood regulation and cognitive functions. Schizophrenia involves complex astrocyte-neuron interactions, with dysregulated astrocytic activity affecting synaptic function and contributing to symptoms. The chapter also delves into neurologic disorders. In Alzheimer disease, aberrant GABA release from reactive astrocytes impairs memory and cognitive functions. Parkinson disease features alterations in glutamatergic and GABAergic systems, affecting motor and nonmotor symptoms. Epilepsy involves a disruption in the balance between excitatory and inhibitory neurotransmission, with astrocytic GABA accumulation helping to maintain neuronal stability. Autism spectrum disorder (ASD) is linked to imbalances in glutamatergic and GABAergic neurotransmission, underlying sensory, cognitive, and social impairments. Overall, the chapter underscores the pivotal role of gliotransmission in maintaining neural homeostasis and highlights its potential as a therapeutic target in various disorders.
Collapse
Affiliation(s)
- 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
|
23
|
Semyanov A, Verkhratsky A. Neuroglia in aging. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:49-67. [PMID: 40122631 DOI: 10.1016/b978-0-443-19104-6.00002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Aging is associated with morphologic and functional decline of the brain active milieu and, in particular, of the neuroglia, which compromises homeostatic support and neuroprotection. Astrocytes in aging undergo complex and region specific changes, manifested by morphologic atrophy and widespread functional asthenia. Aging leads to mitochondrial malfunction and reduced protein/lipid ratio in human astrocytes. Oligodendrocyte lineage cells are the most affected cells by the aging process, which limits myelinating capacity, thus leading to a substantial reduction in the white matter and deficient brain connectome. Finally, microglia undergo a morphologic functional dystrophy in the aged human brain which curtails brain defenses and increases brain vulnerability to neuropathology and especially to age-dependent neurodegenerative disorders. Lifestyle modifications, such as enriched environment, physical exercise, and healthy dieting, boost neuroglial support, thus improving cognitive longevity.
Collapse
Affiliation(s)
- Alexey Semyanov
- Department of Physiology, Jiaxing University College of Medicine, Jiaxing, Zhejiang, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
24
|
Di Palma M, Koh W, Lee CJ, Conti F. A quantitative analysis of bestrophin 1 cellular localization in mouse cerebral cortex. Acta Physiol (Oxf) 2025; 241:e14245. [PMID: 39466647 DOI: 10.1111/apha.14245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
AIM Calcium-activated ligand-gated chloride channels, beyond their role in maintaining anion homeostasis, modulate neuronal excitability by facilitating nonvesicular neurotransmitter release. BEST1, a key member of this family, is permeable to γ-aminobutyric acid (GABA) and glutamate. While astrocytic BEST1 is well-studied and known to regulate neurotransmitter levels, its distribution and role in other brain cell types remain unclear. This study aimed to reassess the localization of BEST1 in the mouse cerebral cortex. METHODS We examined the localization and distribution of BEST1 in the mouse parietal cortex using light microscopy, confocal double-labeling with markers for astrocytes, neurons, microglia, and oligodendrocyte precursor cells, and 3D reconstruction techniques. RESULTS In the cerebral cortex, BEST1 is more broadly distributed than previously thought. Neurons are the second most abundant BEST1+ cell type in the cerebral cortex, following astrocytes. BEST1 is diffusely expressed in neuronal somatic and neuropilar domains and is present at glutamatergic and GABAergic terminals, with a prevalence at GABAergic terminals. We also confirmed that BEST1 is expressed in cortical microglia and identified it in oligodendrocyte precursor cells, albeit to a lesser extent. CONCLUSIONS Together, these findings suggest that BEST1's role in controlling neurotransmission may extend beyond astrocytes to include other brain cells. Understanding BEST1's function in these cells could offer new insights into the molecular mechanisms shaping cortical circuitry. Further research is needed to clarify the diverse roles of BEST1 in both normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
25
|
Wu H, Dong Y, Meng Q, Jiang J, Gao B, Ren Y, Liu Y, Li H, Wang C, Zhang H. Best1 mitigates ER stress induced by the increased cellular microenvironment stiffness in epilepsy. Neurobiol Dis 2025; 204:106767. [PMID: 39674551 DOI: 10.1016/j.nbd.2024.106767] [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: 09/13/2024] [Revised: 11/06/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024] Open
Abstract
Changes in brain tissue stiffness are closely linked to the development and diseases of the nervous system. Endoplasmic reticulum (ER) stress plays a role in various pathological processes related to epilepsy. However, the relationship between stiffness changes, ER stress, and epilepsy remains unclear. This study aimed to investigate the impact of Best1 upregulation on alleviating ER stress and the underlying mechanism. Additionally, we proposed a protective strategy to prevent cell death resulting from ER stress in epilepsy. This study investigated the expression levels of ER stress-related proteins in epileptic tissues of varying stiffness. Atomic force microscopy revealed differences in stiffness across various lesion regions in patients with epilepsy. The expression levels of ECM and ER stress-related proteins were elevated in tissues with higher stiffness. Polypropionamide hydrogels were used to simulate extracellular matrix (ECM) with varying stiffness levels. Basal ER stress increased in the stiffer hydrogel substrates. Furthermore, the calcium-activated anion channel Bestrophin 1 (Best1) mitigated ER stress induced by both the stiffer substrate and thapsigargin. The loss-of-function mutations in Best1 inhibited this activity. The underlying mechanism involves the upregulation of the endosomal sorting complex required for the transport (ESCRT) components by Best1, which helps mitigate ER stress. These findings suggest that increased stiffness of the cellular microenvironment may contribute to neuronal death during epileptogenesis. Additionally, Best1 upregulation may serve as a protective strategy against excessive ER stress-induced neuronal damage in epilepsy.
Collapse
Affiliation(s)
- Hao Wu
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yicong Dong
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiang Meng
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Jiang
- The Second Clinical School, Lanzhou University, Lanzhou, China
| | - Bojian Gao
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, China
| | - Yutao Ren
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huanfa Li
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Changhe Wang
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Hua Zhang
- Department of Neurosurgery, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
26
|
Owji AP, Dong J, Kittredge A, Wang J, Zhang Y, Yang T. Neurotransmitter-bound bestrophin channel structures reveal small molecule drug targeting sites for disease treatment. Nat Commun 2024; 15:10766. [PMID: 39737942 PMCID: PMC11685958 DOI: 10.1038/s41467-024-54938-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Best1 and Best2 are two members of the bestrophin family of anion channels critically involved in the prevention of retinal degeneration and maintenance of intraocular pressure, respectively. Here, we solved glutamate- and γ-aminobutyric acid (GABA)-bound Best2 structures, which delineate an intracellular glutamate binding site and an extracellular GABA binding site on Best2, respectively, identified extracellular GABA as a permeable activator of Best2, and elucidated the co-regulation of Best2 by glutamate, GABA and glutamine synthetase in vivo. We further identified multiple small molecules as activators of the bestrophin channels. Extensive analyses were carried out for a potent activator, 4-aminobenzoic acid (PABA): PABA-bound Best1 and Best2 structures are solved and illustrate the same binding site as in GABA-bound Best2; PABA treatment rescues the functional deficiency of patient-derived Best1 mutations. Together, our results demonstrate the mechanism and potential of multiple small molecule candidates as clinically applicable drugs for bestrophin-associated diseases/conditions.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Jingyun Dong
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
27
|
Pant S, Tam SW, Long SB. The pentameric chloride channel BEST1 is activated by extracellular GABA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624909. [PMID: 39605608 PMCID: PMC11601618 DOI: 10.1101/2024.11.22.624909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Bestrophin 1 (BEST1) is chloride channel expressed in the eye, central nervous system (CNS), and other tissues in the body. A link between BEST1 and the principal inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been proposed. The most appreciated receptors for extracellular GABA are the GABAB G-protein coupled receptors and the pentameric GABAA chloride channels, both of which have fundamental roles in the CNS. Here, we demonstrate that BEST1 is directly activated by GABA. Through functional studies and atomic-resolution structures of human and chicken BEST1, we identify a GABA binding site on the channel's extracellular side and determine the mechanism by which GABA binding induces opening of the channel's central gate. This same gate is activated by intracellular [Ca2+], indicating that BEST1 is controlled by ligands from both sides of the membrane. The studies demonstrate that BEST1, which shares no structural homology with GABAA, is a GABA-activated chloride channel. The physiological implications of this finding remain to be studied.
Collapse
Affiliation(s)
- Swati Pant
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA
| | - Stephanie W. Tam
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Graduate Program in Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
28
|
Baron M, Devor M. Neurosteroids foster sedation by engaging tonic GABA A-Rs within the mesopontine tegmental anesthesia area (MPTA). Neurosci Lett 2024; 843:138030. [PMID: 39490574 DOI: 10.1016/j.neulet.2024.138030] [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: 06/04/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Neurosteroids are endogenous molecules with anxiolytic, anticonvulsant, sleep-promoting and sedative effects. They are biosynthesized de novo within the brain, among other tissues, and are thought to act primarily as positive allosteric modulators of high-affinity extrasynaptic GABAAδ-receptors. The location of action of neurosteroids in the brain, however, remains unknown. We have demonstrated that GABAergic anesthetics act within the brainstem mesopontine tegmental anesthesia area (MPTA) to induce and maintain anesthetic loss-of-consciousness. Here we asked whether endogenous and synthetic neurosteroids might also act in the MPTA to induce their suppressive effects. Direct exposure of the MPTA to the endogenous neurosteroids pregnenolone and progesterone, their metabolites testosterone, allopregnanolone and 3α5α-THDOC, and the synthetic neurosteroids ganaxolone and alphaxalone, was found to be pro-anesthetic. Although we cannot rule out additional sites of action, results of this study suggest that the suppressive effects of neurosteroids are due, at least in part, to actions within the MPTA, presumably by recruitment of dedicated neuronal circuitry. This undermines the usual presumption that neurosteroids, like other sedatives, endogenous somnogens and anesthetics, act by nonspecific global distribution.
Collapse
Affiliation(s)
- Mark Baron
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Marshall Devor
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
29
|
Wang C, Vidal B, Sural S, Loer C, Aguilar GR, Merritt DM, Toker IA, Vogt MC, Cros CC, Hobert O. A neurotransmitter atlas of C. elegans males and hermaphrodites. eLife 2024; 13:RP95402. [PMID: 39422452 PMCID: PMC11488851 DOI: 10.7554/elife.95402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Mapping neurotransmitter identities to neurons is key to understanding information flow in a nervous system. It also provides valuable entry points for studying the development and plasticity of neuronal identity features. In the Caenorhabditis elegans nervous system, neurotransmitter identities have been largely assigned by expression pattern analysis of neurotransmitter pathway genes that encode neurotransmitter biosynthetic enzymes or transporters. However, many of these assignments have relied on multicopy reporter transgenes that may lack relevant cis-regulatory information and therefore may not provide an accurate picture of neurotransmitter usage. We analyzed the expression patterns of 16 CRISPR/Cas9-engineered knock-in reporter strains for all main types of neurotransmitters in C. elegans (glutamate, acetylcholine, GABA, serotonin, dopamine, tyramine, and octopamine) in both the hermaphrodite and the male. Our analysis reveals novel sites of expression of these neurotransmitter systems within both neurons and glia, as well as non-neural cells, most notably in gonadal cells. The resulting expression atlas defines neurons that may be exclusively neuropeptidergic, substantially expands the repertoire of neurons capable of co-transmitting multiple neurotransmitters, and identifies novel sites of monoaminergic neurotransmitter uptake. Furthermore, we also observed unusual co-expression patterns of monoaminergic synthesis pathway genes, suggesting the existence of novel monoaminergic transmitters. Our analysis results in what constitutes the most extensive whole-animal-wide map of neurotransmitter usage to date, paving the way for a better understanding of neuronal communication and neuronal identity specification in C. elegans.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Curtis Loer
- Department of Biology, University of San DiegoSan DiegoUnited States
| | - G Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Daniel M Merritt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Itai Antoine Toker
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Merly C Vogt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Cyril C Cros
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| |
Collapse
|
30
|
Chandra S, Vassar R. The role of the gut microbiome in the regulation of astrocytes in Alzheimer's disease. Neurotherapeutics 2024; 21:e00425. [PMID: 39054180 PMCID: PMC11585888 DOI: 10.1016/j.neurot.2024.e00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and is the most common cause of dementia. AD is characterized pathologically by proteinaceous aggregates composed of amyloid beta (Aβ) and tau as well as progressive neurodegeneration. Concurrently with the buildup of protein aggregates, a strong neuroinflammatory response, in the form of reactive astrocytosis and microgliosis, occurs in the AD brain. It has recently been shown that the gut microbiome (GMB), composed of trillions of bacteria in the human intestine, can regulate both reactive astrocytosis and microgliosis in the context of both amyloidosis and tauopathy. Many studies have implicated microglia in these processes. However, growing evidence suggests that interactions between the GMB and astrocytes have a much larger role than previously thought. In this review, we summarize evidence regarding the gut microbiome in the control of reactive astrocytosis in AD.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA; Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
31
|
Wang J, Owji AP, Kittredge A, Clark Z, Zhang Y, Yang T. GAD65 tunes the functions of Best1 as a GABA receptor and a neurotransmitter conducting channel. Nat Commun 2024; 15:8051. [PMID: 39277606 PMCID: PMC11401937 DOI: 10.1038/s41467-024-52039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/23/2024] [Indexed: 09/17/2024] Open
Abstract
Bestrophin-1 (Best1) is an anion channel genetically linked to vision-threatening retinal degenerative channelopathies. Here, we identify interactions between Best1 and both isoforms of glutamic acid decarboxylases (GAD65 and GAD67), elucidate the distinctive influences of GAD65 and GAD67 on Best1's permeability to various anions/neurotransmitters, discover the functionality of Best1 as a γ-Aminobutyric acid (GABA) type A receptor, and solve the structure of GABA-bound Best1. GAD65 and GAD67 both promote Best1-mediated Cl- currents, but only GAD65 drastically enhances the permeability of Best1 to glutamate and GABA, for which GAD67 has no effect. GABA binds to Best1 on an extracellular site and stimulates Best1-mediated Cl- currents at the nano-molar concentration level. The physiological role of GAD65 as a cell type-specific binding partner and facilitator of Best1 is demonstrated in retinal pigment epithelial cells. Together, our results reveal critical regulators of Best1 and inform a network of membrane transport metabolons formed between bestrophin channels and glutamate metabolic enzymes.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Zada Clark
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
32
|
Wang Y, Wang Z. Effects and Safety of Monoamine Oxidase-B Inhibitors for Early Parkinson's Disease: A Network Meta-Analysis. Eur Neurol 2024; 87:273-290. [PMID: 39278214 DOI: 10.1159/000541315] [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: 02/29/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024]
Abstract
INTRODUCTION The objective of this study was to evaluate the effects and safety of monoamine oxidase-B inhibitors (MAO-B inhibitors) for early Parkinson's disease (PD). METHODS All studies that assessed the efficacy of MAO-B inhibitors in patients with early PD were searched. Publications were screened, and data were extracted according to predefined criteria. Rev Man 5.4 and Stata 14.0 software were used for statistical analysis. Outcomes assessed included change of Unified Parkinson's Disease Rating Scale (UPDRS) total score, UPDRS part II score, UPDRS part III score, and the incidence of adverse events. RESULTS Thirty trials were identified and included in this meta-analysis. Compared with placebo, rasagiline, selegiline, safinamide, and zonisamide were significantly more effective, with a standardized mean difference (SMD) of -0.41 (95% confidence interval (CI) = -0.64 to -0.18), SMD = -0.38 (95% CI = -0.51 to -0.24), SMD = -0.37 (95% CI = -0.54 to -0.21), and SMD = -0.31 (95% CI = -0.57 to -0.05) on the UPDRS III score change, respectively. The surface under the cumulative ranking results showed that rasagiline ranked first in improving UPDRS II and UPDRS III, respectively. For safety outcomes, safinamide combination with dopaminergic treatment had lower risk of incurring any adverse events (risk ratio = 0.1, 95% CI = 0.01-0.2), and no statistical difference in incidence of adverse events was observed among other MAO-B inhibitor regimes and placebo. CONCLUSION Rasagiline, selegiline, safinamide, and zonisamide were effective compared to placebo in the treatment of early PD, but rasagiline was the most effective drug. As for safety, safinamide combination with dopaminergic treatment had lower risk of incurring any adverse events.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
33
|
Abdelaziz MA, Chen WH, Chang YW, Mindaye SA, Chen CC. Exploring the role of spinal astrocytes in the onset of hyperalgesic priming signals in acid-induced chronic muscle pain. PNAS NEXUS 2024; 3:pgae362. [PMID: 39228816 PMCID: PMC11370897 DOI: 10.1093/pnasnexus/pgae362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024]
Abstract
Hyperalgesic priming, a form of pain plasticity initiated by initial injury, leads to heightened sensitivity to subsequent noxious stimuli, contributing to chronic pain development in animals. While astrocytes play active roles in modulating synaptic transmission in various pain models, their specific involvement in hyperalgesic priming remains elusive. Here, we show that spinal astrocytes are essential for hyperalgesic priming formation in a mouse model of acid-induced muscle pain. We observed spinal astrocyte activation 4 h after initial acid injection, and inhibition of this activation prevented chronic pain development upon subsequent acid injection. Chemogenetic activation of spinal astrocytes mimicked the first acid-induced hyperalgesic priming. We also demonstrated that spinal phosphorylated extracellular regulated kinase (pERK)-positive neurons were mainly vesicular glutamate transporter-2 positive (Vglut2+) neurons after the first acid injection, and inhibition of spinal pERK prevented astrocyte activation. Furthermore, pharmacological inhibition of astrocytic glutamate transporters glutamate transporter-1 and glutamate-aspartate transporter abolished the hyperalgesic priming. Collectively, our results suggest that pERK activation in Vglut2+ neurons activate astrocytes through astrocytic glutamate transporters. This process eventually establishes hyperalgesic priming through spinal D-serine. We conclude that spinal astrocytes play a crucial role in the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Mohamed Abbas Abdelaziz
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Zoology Department, Faculty of Science, Al-Azhar University Assiut Branch, Assiut 71524, Egypt
| | - Wei-Hsin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Selomon Assefa Mindaye
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
34
|
Joo J, Kim KJ, Lim J, Choi SY, Koh W, Lee CJ. Generation of Astrocyte-specific BEST1 Conditional Knockout Mouse with Reduced Tonic GABA Inhibition in the Brain. Exp Neurobiol 2024; 33:180-192. [PMID: 39266474 PMCID: PMC11411089 DOI: 10.5607/en24019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/14/2024] Open
Abstract
Bestrophin-1 (BEST1) is a Ca2+-activated anion channel known for its role in astrocytes. Best1 is permeable to gliotransmitters, including GABA, to contribute to tonic GABA inhibition and modulate synaptic transmission in neighboring neurons. Despite the crucial functions of astrocytic BEST1, there is an absence of genetically engineered cell-type specific conditional mouse models addressing these roles. In this study, we developed an astrocyte-specific BEST1 conditional knock-out (BEST1 aKO) mouse line. Using the embryonic stem cell (ES cell) targeting method, we developed Best1 floxed mice (C57BL/6JCya-Best1em1flox/Cya), which have exon 3, 4, 5, and 6 of Best1 flanked by two loxP sites. By crossing with hGFAP-CreERT2 mice, we generated Best1 floxed/hGFAP-CreERT2 mice, which allowed for the tamoxifen-inducible deletion of Best1 under the human GFAP promoter. We characterized its features across various brain regions, including the striatum, hippocampal dentate gyrus (HpDG), and Parafascicular thalamic nucleus (Pf). Compared to the Cre-negative control, we observed significantly reduced BEST1 protein expression in immunohistochemistry (IHC) and tonic GABA inhibition in patch clamp recordings. The reduction in tonic GABA inhibition was 66.7% in the striatum, 46.4% in the HpDG, and 49.6% in the Pf. Our findings demonstrate that the BEST1 channel in astrocytes significantly contributes to tonic inhibition in the local brain areas. These mice will be valuable for future studies not only on tonic GABA release but also on tonic release of gliotransmitters mediated by astrocytic BEST1.
Collapse
Affiliation(s)
- Jinhyeong Joo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Ki Jung Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Sun Yeong Choi
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
35
|
Calì C, Cantando I, Veloz Castillo MF, Gonzalez L, Bezzi P. Metabolic Reprogramming of Astrocytes in Pathological Conditions: Implications for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:8922. [PMID: 39201607 PMCID: PMC11354244 DOI: 10.3390/ijms25168922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Astrocytes play a pivotal role in maintaining brain energy homeostasis, supporting neuronal function through glycolysis and lipid metabolism. This review explores the metabolic intricacies of astrocytes in both physiological and pathological conditions, highlighting their adaptive plasticity and diverse functions. Under normal conditions, astrocytes modulate synaptic activity, recycle neurotransmitters, and maintain the blood-brain barrier, ensuring a balanced energy supply and protection against oxidative stress. However, in response to central nervous system pathologies such as neurotrauma, stroke, infections, and neurodegenerative diseases like Alzheimer's and Huntington's disease, astrocytes undergo significant morphological, molecular, and metabolic changes. Reactive astrocytes upregulate glycolysis and fatty acid oxidation to meet increased energy demands, which can be protective in acute settings but may exacerbate chronic inflammation and disease progression. This review emphasizes the need for advanced molecular, genetic, and physiological tools to further understand astrocyte heterogeneity and their metabolic reprogramming in disease states.
Collapse
Affiliation(s)
- Corrado Calì
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, 10143 Orbassano, Italy
| | - Iva Cantando
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
| | - Maria Fernanda Veloz Castillo
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, 10143 Orbassano, Italy
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Laurine Gonzalez
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
| | - Paola Bezzi
- Department of Fundamental Neurosciences (DNF), University of Lausanne (UNIL), 1005 Lausanne, Switzerland; (I.C.); (L.G.)
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy
| |
Collapse
|
36
|
Tan W, Ikoma Y, Takahashi Y, Konno A, Hirai H, Hirase H, Matsui K. Anxiety control by astrocytes in the lateral habenula. Neurosci Res 2024; 205:1-15. [PMID: 38311032 DOI: 10.1016/j.neures.2024.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/06/2024]
Abstract
The potential role of astrocytes in lateral habenula (LHb) in modulating anxiety was explored in this study. The habenula are a pair of small nuclei located above the thalamus, known for their involvement in punishment avoidance and anxiety. Herein, we observed an increase in theta-band oscillations of local field potentials (LFPs) in the LHb when mice were exposed to anxiety-inducing environments. Electrical stimulation of LHb at theta-band frequency promoted anxiety-like behavior. Calcium (Ca2+) levels and pH in the cytosol of astrocytes and local brain blood volume changes were studied in mice expressing either a Ca2+ or a pH sensor protein specifically in astrocytes and mScarlet fluorescent protein in the blood plasma using fiber photometry. An acidification response to anxiety was observed. Photoactivation of archaerhopsin-T (ArchT), an optogenetic tool that acts as an outward proton pump, results in intracellular alkalinization. Photostimulation of LHb in astrocyte-specific ArchT-expressing mice resulted in dissipation of theta-band LFP oscillation in an anxiogenic environment and suppression of anxiety-like behavior. These findings provide evidence that LHb astrocytes modulate anxiety and may offer a new target for treatment of anxiety disorders.
Collapse
Affiliation(s)
- Wanqin Tan
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yusuke Takahashi
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan; Systems Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai 980-8579 Japan
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan.
| |
Collapse
|
37
|
Wang C, Vidal B, Sural S, Loer C, Aguilar GR, Merritt DM, Toker IA, Vogt MC, Cros C, Hobert O. A neurotransmitter atlas of C. elegans males and hermaphrodites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.24.573258. [PMID: 38895397 PMCID: PMC11185579 DOI: 10.1101/2023.12.24.573258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mapping neurotransmitter identities to neurons is key to understanding information flow in a nervous system. It also provides valuable entry points for studying the development and plasticity of neuronal identity features. In the C. elegans nervous system, neurotransmitter identities have been largely assigned by expression pattern analysis of neurotransmitter pathway genes that encode neurotransmitter biosynthetic enzymes or transporters. However, many of these assignments have relied on multicopy reporter transgenes that may lack relevant cis-regulatory information and therefore may not provide an accurate picture of neurotransmitter usage. We analyzed the expression patterns of 16 CRISPR/Cas9-engineered knock-in reporter strains for all main types of neurotransmitters in C. elegans (glutamate, acetylcholine, GABA, serotonin, dopamine, tyramine, and octopamine) in both the hermaphrodite and the male. Our analysis reveals novel sites of expression of these neurotransmitter systems within both neurons and glia, as well as non-neural cells. The resulting expression atlas defines neurons that may be exclusively neuropeptidergic, substantially expands the repertoire of neurons capable of co-transmitting multiple neurotransmitters, and identifies novel neurons that uptake monoaminergic neurotransmitters. Furthermore, we also observed unusual co-expression patterns of monoaminergic synthesis pathway genes, suggesting the existence of novel monoaminergic transmitters. Our analysis results in what constitutes the most extensive whole-animal-wide map of neurotransmitter usage to date, paving the way for a better understanding of neuronal communication and neuronal identity specification in C. elegans.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Curtis Loer
- Department of Biology, University of San Diego, San Diego, California, USA
| | - G. Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Daniel M. Merritt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Itai Antoine Toker
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Merly C. Vogt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Cyril Cros
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| |
Collapse
|
38
|
Graziano B, Wang L, White OR, Kaplan DH, Fernandez-Abascal J, Bianchi L. Glial KCNQ K + channels control neuronal output by regulating GABA release from glia in C. elegans. Neuron 2024; 112:1832-1847.e7. [PMID: 38460523 PMCID: PMC11156561 DOI: 10.1016/j.neuron.2024.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/11/2024]
Abstract
KCNQs are voltage-gated K+ channels that control neuronal excitability and are mutated in epilepsy and autism spectrum disorder (ASD). KCNQs have been extensively studied in neurons, but their function in glia is unknown. Using voltage, calcium, and GABA imaging, optogenetics, and behavioral assays, we show here for the first time in Caenorhabditis elegans (C. elegans) that glial KCNQ channels control neuronal excitability by mediating GABA release from glia via regulation of the function of L-type voltage-gated Ca2+ channels. Further, we show that human KCNQ channels have the same role when expressed in nematode glia, underscoring conservation of function across species. Finally, we show that pathogenic loss-of-function and gain-of-function human KCNQ2 mutations alter glia-to-neuron GABA signaling in distinct ways and that the KCNQ channel opener retigabine exerts rescuing effects. This work identifies glial KCNQ channels as key regulators of neuronal excitability via control of GABA release from glia.
Collapse
Affiliation(s)
- Bianca Graziano
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lei Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Olivia R White
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daryn H Kaplan
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
39
|
Gonçalves-Ribeiro J, Savchak OK, Costa-Pinto S, Gomes JI, Rivas-Santisteban R, Lillo A, Sánchez Romero J, Sebastião AM, Navarrete M, Navarro G, Franco R, Vaz SH. Adenosine receptors are the on-and-off switch of astrocytic cannabinoid type 1 (CB1) receptor effect upon synaptic plasticity in the medial prefrontal cortex. Glia 2024; 72:1096-1116. [PMID: 38482984 DOI: 10.1002/glia.24518] [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: 08/23/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 04/12/2024]
Abstract
The medial prefrontal cortex (mPFC) is involved in cognitive functions such as working memory. Astrocytic cannabinoid type 1 receptor (CB1R) induces cytosolic calcium (Ca2+) concentration changes with an impact on neuronal function. mPFC astrocytes also express adenosine A1 and A2A receptors (A1R, A2AR), being unknown the crosstalk between CB1R and adenosine receptors in these cells. We show here that a further level of regulation of astrocyte Ca2+ signaling occurs through CB1R-A2AR or CB1R-A1R heteromers that ultimately impact mPFC synaptic plasticity. CB1R-mediated Ca2+ transients increased and decreased when A1R and A2AR were activated, respectively, unveiling adenosine receptors as modulators of astrocytic CB1R. CB1R activation leads to an enhancement of long-term potentiation (LTP) in the mPFC, under the control of A1R but not of A2AR. Notably, in IP3R2KO mice, that do not show astrocytic Ca2+ level elevations, CB1R activation decreases LTP, which is not modified by A1R or A2AR. The present work suggests that CB1R has a homeostatic role on mPFC LTP, under the control of A1R, probably due to physical crosstalk between these receptors in astrocytes that ultimately alters CB1R Ca2+ signaling.
Collapse
Affiliation(s)
- Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Oksana K Savchak
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Costa-Pinto
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Joana I Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rafael Rivas-Santisteban
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Javier Sánchez Romero
- Instituto Cajal, CSIC, Madrid, Spain
- PhD Program in Neuroscience, Universidad Autónoma de Madrid-Instituto Cajal, Madrid, Spain
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Gemma Navarro
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
40
|
Won D, Lee EH, Chang JE, Nam MH, Park KD, Oh SJ, Hwang JY. The role of astrocytic γ-aminobutyric acid in the action of inhalational anesthetics. Eur J Pharmacol 2024; 970:176494. [PMID: 38484926 DOI: 10.1016/j.ejphar.2024.176494] [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: 12/04/2023] [Revised: 01/24/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Inhalational anesthetics target the inhibitory extrasynaptic γ-aminobutyric acid type A (GABAA) receptors. Both neuronal and glial GABA mediate tonic inhibition of the extrasynaptic GABAA receptors. However, the role of glial GABA during inhalational anesthesia remains unclear. This study aimed to evaluate whether astrocytic GABA contributes to the action of different inhalational anesthetics. METHODS Gene knockout of monoamine oxidase B (MAOB) was used to reduce astrocytic GABA levels in mice. The hypnotic and immobilizing effects of isoflurane, sevoflurane, and desflurane were assessed by evaluating the loss of righting reflex (LORR) and tail-pinch withdrawal response (LTWR) in MAOB knockout and wild-type mice. Minimum alveolar concentration (MAC) for LORR, time to LORR, MAC for LTWR and time to LTWR of isoflurane, sevoflurane, and desflurane were assessed. RESULTS Time to LORR and time to LTWR with isoflurane were significantly longer in MAOB knockout mice than in wild-type mice (P < 0.001 and P = 0.032, respectively). Time to LORR with 0.8 MAC of sevoflurane was significantly longer in MAOB knockout mice than in wild-type mice (P < 0.001), but not with 1.0 MAC of sevoflurane (P=0.217). MAC for LTWR was significantly higher in MAOB knockout mice exposed to sevoflurane (P < 0.001). With desflurane, MAOB knockout mice had a significantly higher MAC for LORR (P = 0.003) and higher MAC for LTWR (P < 0.001) than wild-type mice. CONCLUSIONS MAOB knockout mice showed reduced sensitivity to the hypnotic and immobilizing effects of isoflurane, sevoflurane, and desflurane. Behavioral tests revealed that the hypnotic and immobilizing effects of inhalational anesthetics would be mediated by astrocytic GABA.
Collapse
Affiliation(s)
- Dongwook Won
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Elliot H Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jee-Eun Chang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| | - Jin-Young Hwang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Wang DS, Ju L, Pinguelo AG, Kaneshwaran K, Haffey SC, Lecker I, Gohil H, Wheeler MB, Kaustov L, Ariza A, Yu M, Volchuk A, Steinberg BE, Goldenberg NM, Orser BA. Crosstalk between GABA A receptors in astrocytes and neurons triggered by general anesthetic drugs. Transl Res 2024; 267:39-53. [PMID: 38042478 DOI: 10.1016/j.trsl.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/17/2023] [Accepted: 11/29/2023] [Indexed: 12/04/2023]
Abstract
General anesthetic drugs cause cognitive deficits that persist after the drugs have been eliminated. Astrocytes may contribute to such cognition-impairing effects through the release of one or more paracrine factors that increase a tonic inhibitory conductance generated by extrasynaptic γ-aminobutyric acid type A (GABAA) receptors in hippocampal neurons. The mechanisms underlying this astrocyte-to-neuron crosstalk remain unknown. Interestingly, astrocytes express anesthetic-sensitive GABAA receptors. Here, we tested the hypothesis that anesthetic drugs activate astrocytic GABAA receptors to initiate crosstalk leading to a persistent increase in extrasynaptic GABAA receptor function in neurons. We also investigated the signaling pathways in neurons and aimed to identify the paracrine factors released from astrocytes. Astrocytes and neurons from mice were grown in primary cell cultures and studied using in vitro electrophysiological and biochemical assays. We discovered that the commonly used anesthetics etomidate (injectable) and sevoflurane (inhaled) stimulated astrocytic GABAA receptors, which in turn promoted the release paracrine factors, that increased the tonic current in neurons via a p38 MAPK-dependent signaling pathway. The increase in tonic current was mimicked by exogenous IL-1β and abolished by blocking IL-1 receptors; however, unexpectedly, IL-1β and other cytokines were not detected in astrocyte-conditioned media. In summary, we have identified a novel form of crosstalk between GABAA receptors in astrocytes and neurons that engages a p38 MAPK-dependent pathway. Brief commentary BACKGROUND: Many older patients experience cognitive deficits after surgery. Anesthetic drugs may be a contributing factor as they cause a sustained increase in the function of "memory blocking" extrasynaptic GABAA receptors in neurons. Interestingly, astrocytes are required for this increase; however, the mechanisms underlying the astrocyte-to-neuron crosstalk remain unknown. TRANSLATIONAL SIGNIFICANCE: We discovered that commonly used general anesthetic drugs stimulate GABAA receptors in astrocytes, which in turn release paracrine factors that trigger a persistent increase in extrasynaptic GABAA receptor function in neurons via p38 MAPK. This novel form of crosstalk may contribute to persistent cognitive deficits after general anesthesia and surgery.
Collapse
Affiliation(s)
- Dian-Shi Wang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Li Ju
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arsène G Pinguelo
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kirusanthy Kaneshwaran
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sean C Haffey
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Irene Lecker
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Himaben Gohil
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lilia Kaustov
- Department of Anesthesia, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Anthony Ariza
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - MeiFeng Yu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allen Volchuk
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Benjamin E Steinberg
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Anesthesiology & Pain Medicine, Temerty Faculty of Medicine, University of Toronto, Room 3318, Medical Sciences Building, 1 King's College Circle, Ontario M5S 1A8, Canada
| | - Neil M Goldenberg
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Anesthesiology & Pain Medicine, Temerty Faculty of Medicine, University of Toronto, Room 3318, Medical Sciences Building, 1 King's College Circle, Ontario M5S 1A8, Canada; Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Beverley A Orser
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Department of Anesthesiology & Pain Medicine, Temerty Faculty of Medicine, University of Toronto, Room 3318, Medical Sciences Building, 1 King's College Circle, Ontario M5S 1A8, Canada.
| |
Collapse
|
42
|
Rothman DL, Behar KL, Dienel GA. Mechanistic stoichiometric relationship between the rates of neurotransmission and neuronal glucose oxidation: Reevaluation of and alternatives to the pseudo-malate-aspartate shuttle model. J Neurochem 2024; 168:555-591. [PMID: 36089566 DOI: 10.1111/jnc.15619] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
The ~1:1 stoichiometry between the rates of neuronal glucose oxidation (CMRglc-ox-N) and glutamate (Glu)/γ-aminobutyric acid (GABA)-glutamine (Gln) neurotransmitter (NT) cycling between neurons and astrocytes (VNTcycle) has been firmly established. However, the mechanistic basis for this relationship is not fully understood, and this knowledge is critical for the interpretation of metabolic and brain imaging studies in normal and diseased brain. The pseudo-malate-aspartate shuttle (pseudo-MAS) model established the requirement for glycolytic metabolism in cultured glutamatergic neurons to produce NADH that is shuttled into mitochondria to support conversion of extracellular Gln (i.e., astrocyte-derived Gln in vivo) into vesicular neurotransmitter Glu. The evaluation of this model revealed that it could explain half of the 1:1 stoichiometry and it has limitations. Modifications of the pseudo-MAS model were, therefore, devised to address major knowledge gaps, that is, submitochondrial glutaminase location, identities of mitochondrial carriers for Gln and other model components, alternative mechanisms to transaminate α-ketoglutarate to form Glu and shuttle glutamine-derived ammonia while maintaining mass balance. All modified models had a similar 0.5 to 1.0 predicted mechanistic stoichiometry between VNTcycle and the rate of glucose oxidation. Based on studies of brain β-hydroxybutyrate oxidation, about half of CMRglc-ox-N may be linked to glutamatergic neurotransmission and localized in pre-synaptic structures that use pseudo-MAS type mechanisms for Glu-Gln cycling. In contrast, neuronal compartments that do not participate in transmitter cycling may use the MAS to sustain glucose oxidation. The evaluation of subcellular compartmentation of neuronal glucose metabolism in vivo is a critically important topic for future studies to understand glutamatergic and GABAergic neurotransmission.
Collapse
Affiliation(s)
- Douglas L Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Kevin L Behar
- Magnetic Resonance Research Center and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
43
|
Asano Y, Sasaki D, Ikoma Y, Matsui K. Glial tone of aggression. Neurosci Res 2024; 202:39-51. [PMID: 38007191 DOI: 10.1016/j.neures.2023.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Anger transition is often abrupt. In this study, we investigated the mechanisms responsible for switching and modulating aggression levels. The cerebellum is considered a center for motor coordination and learning; however, its connection to social behavior has long been observed. Here, we used the resident-intruder paradigm in male mice and examined local field potential (LFP) changes, glial cytosolic ion fluctuations, and vascular dynamics in the cerebellar vermis throughout various phases of a combat sequence. Notably, we observed the emergence of theta band oscillations in the LFP and sustained elevations in glial Ca2+ levels during combat breakups. When astrocytes, including Bergmann glial cells, were photoactivated using channelrhodopsin-2, the theta band emerged and an early combat breakup occurred. Within a single combat sequence, rapid alteration of offensive (fight) and passive (flight) responses were observed, which roughly correlated with decreases and increases in glial Ca2+, respectively. Neuron-glial interactions in the cerebellar vermis may play a role in adjusting Purkinje cell excitability and setting the tone of aggression. Future anger management strategies and clinical control of excessive aggression and violent behavior may be realized by developing a therapeutic strategy that adjusts glial activity in the cerebellum.
Collapse
Affiliation(s)
- Yuki Asano
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Daichi Sasaki
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan.
| |
Collapse
|
44
|
Djama D, Zirpel F, Ye Z, Moore G, Chue C, Edge C, Jager P, Delogu A, Brickley SG. The type of inhibition provided by thalamic interneurons alters the input selectivity of thalamocortical neurons. CURRENT RESEARCH IN NEUROBIOLOGY 2024; 6:100130. [PMID: 38694514 PMCID: PMC11061260 DOI: 10.1016/j.crneur.2024.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
A fundamental problem in neuroscience is how neurons select for their many inputs. A common assumption is that a neuron's selectivity is largely explained by differences in excitatory synaptic input weightings. Here we describe another solution to this important problem. We show that within the first order visual thalamus, the type of inhibition provided by thalamic interneurons has the potential to alter the input selectivity of thalamocortical neurons. To do this, we developed conductance injection protocols to compare how different types of synchronous and asynchronous GABA release influence thalamocortical excitability in response to realistic patterns of retinal ganglion cell input. We show that the asynchronous GABA release associated with tonic inhibition is particularly efficient at maintaining information content, ensuring that thalamocortical neurons can distinguish between their inputs. We propose a model where alterations in GABA release properties results in rapid changes in input selectivity without requiring structural changes in the network.
Collapse
Affiliation(s)
- Deyl Djama
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Florian Zirpel
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Zhiwen Ye
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Gerald Moore
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Charmaine Chue
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Edge
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Polona Jager
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - Alessio Delogu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | | |
Collapse
|
45
|
Zheng Z, Zhou H, Yang L, Zhang L, Guo M. Selective disruption of mTORC1 and mTORC2 in VTA astrocytes induces depression and anxiety-like behaviors in mice. Behav Brain Res 2024; 463:114888. [PMID: 38307148 DOI: 10.1016/j.bbr.2024.114888] [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: 12/11/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Dysfunction of the mechanistic target of rapamycin (mTOR) signaling pathway is implicated in neuropsychiatric disorders including depression and anxiety. Most studies have been focusing on neurons, and the function of mTOR signaling pathway in astrocytes is less investigated. mTOR forms two distinct complexes, mTORC1 and mTORC2, with key scaffolding protein Raptor and Rictor, respectively. The ventral tegmental area (VTA), a vital component of the brain reward system, is enrolled in regulating both depression and anxiety. In the present study, we aimed to examine the regulation effect of VTA astrocytic mTOR signaling pathway on depression and anxiety. We specifically deleted Raptor or Rictor in VTA astrocytes in mice and performed a series of behavioral tests for depression and anxiety. Deletion of Raptor and Rictor both decreased the immobility time in the tail suspension test and the latency to eat in the novelty suppressed feeding test, and increased the horizontal activity and the movement time in locomotor activity. Deletion of Rictor decreased the number of total arm entries in the elevated plus-maze test and the vertical activity in locomotor activity. These data suggest that VTA astrocytic mTORC1 plays a role in regulating depression-related behaviors and mTORC2 is involved in both depression and anxiety-related behaviors. Our results indicate that VTA astrocytic mTOR signaling pathway might be new targets for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Ziteng Zheng
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Han Zhou
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Lu Yang
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Lanlan Zhang
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Ming Guo
- Department of Psychology, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, the First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong 256603, China.
| |
Collapse
|
46
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Mishima T, Komano K, Tabaru M, Kofuji T, Saito A, Ugawa Y, Terao Y. Repetitive pulsed-wave ultrasound stimulation suppresses neural activity by modulating ambient GABA levels via effects on astrocytes. Front Cell Neurosci 2024; 18:1361242. [PMID: 38601023 PMCID: PMC11004293 DOI: 10.3389/fncel.2024.1361242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Ultrasound is highly biopermeable and can non-invasively penetrate deep into the brain. Stimulation with patterned low-intensity ultrasound can induce sustained inhibition of neural activity in humans and animals, with potential implications for research and therapeutics. Although mechanosensitive channels are involved, the cellular and molecular mechanisms underlying neuromodulation by ultrasound remain unknown. To investigate the mechanism of action of ultrasound stimulation, we studied the effects of two types of patterned ultrasound on synaptic transmission and neural network activity using whole-cell recordings in primary cultured hippocampal cells. Single-shot pulsed-wave (PW) or continuous-wave (CW) ultrasound had no effect on neural activity. By contrast, although repetitive CW stimulation also had no effect, repetitive PW stimulation persistently reduced spontaneous recurrent burst firing. This inhibitory effect was dependent on extrasynaptic-but not synaptic-GABAA receptors, and the effect was abolished under astrocyte-free conditions. Pharmacological activation of astrocytic TRPA1 channels mimicked the effects of ultrasound by increasing the tonic GABAA current induced by ambient GABA. Pharmacological blockade of TRPA1 channels abolished the inhibitory effect of ultrasound. These findings suggest that the repetitive PW low-intensity ultrasound used in our study does not have a direct effect on neural function but instead exerts its sustained neuromodulatory effect through modulation of ambient GABA levels via channels with characteristics of TRPA1, which is expressed in astrocytes.
Collapse
Affiliation(s)
- Tatsuya Mishima
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| | - Kenta Komano
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Marie Tabaru
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takefumi Kofuji
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
- Radioisotope Laboratory, Kyorin University School of Medicine, Mitaka, Japan
| | - Ayako Saito
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuo Terao
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| |
Collapse
|
48
|
Bhusal A, Kim JH, Kim SC, Hwang EM, Ryu H, Ali MS, Park SC, Lee WH, Suk K. The microglial innate immune protein PGLYRP1 mediates neuroinflammation and consequent behavioral changes. Cell Rep 2024; 43:113813. [PMID: 38393947 DOI: 10.1016/j.celrep.2024.113813] [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: 03/25/2023] [Revised: 01/05/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Peptidoglycan recognition protein 1 (PGLYRP1) is a pattern-recognition protein that mediates antibacterial actions and innate immune responses. Its expression and role in neuroinflammatory conditions remain unclear. We observed the upregulation of PGLYRP1 in inflamed human and mouse spinal cord and brain, with microglia being the primary cellular source. Experiments using a recombinant PGLYRP1 protein show that PGLYRP1 potentiates reactive gliosis, neuroinflammation, and consequent behavioral changes in multiple animal models of neuroinflammation. Furthermore, shRNA-mediated knockdown of Pglyrp1 gene expression attenuates this inflammatory response. In addition, we identify triggering receptor expressed on myeloid cell-1 (TREM1) as an interaction partner of PGLYRP1 and demonstrate that PGLYRP1 promotes neuroinflammation through the TREM1-Syk-Erk1/2-Stat3 axis in cultured glial cells. Taken together, our results reveal a role for microglial PGLYRP1 as a neuroinflammation mediator. Finally, we propose that PGLYRP1 is a potential biomarker and therapeutic target in various neuroinflammatory diseases.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Korea 21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Seung-Chan Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hoon Ryu
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Md Sekendar Ali
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
49
|
Patel JC, Sherpa AD, Melani R, Witkovsky P, Wiseman MR, O'Neill B, Aoki C, Tritsch NX, Rice ME. GABA co-released from striatal dopamine axons dampens phasic dopamine release through autoregulatory GABA A receptors. Cell Rep 2024; 43:113834. [PMID: 38431842 PMCID: PMC11089423 DOI: 10.1016/j.celrep.2024.113834] [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: 10/05/2022] [Revised: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Striatal dopamine axons co-release dopamine and gamma-aminobutyric acid (GABA), using GABA provided by uptake via GABA transporter-1 (GAT1). Functions of GABA co-release are poorly understood. We asked whether co-released GABA autoinhibits dopamine release via axonal GABA type A receptors (GABAARs), complementing established inhibition by dopamine acting at axonal D2 autoreceptors. We show that dopamine axons express α3-GABAAR subunits in mouse striatum. Enhanced dopamine release evoked by single-pulse optical stimulation in striatal slices with GABAAR antagonism confirms that an endogenous GABA tone limits dopamine release. Strikingly, an additional inhibitory component is seen when multiple pulses are used to mimic phasic axonal activity, revealing the role of GABAAR-mediated autoinhibition of dopamine release. This autoregulation is lost in conditional GAT1-knockout mice lacking GABA co-release. Given the faster kinetics of ionotropic GABAARs than G-protein-coupled D2 autoreceptors, our data reveal a mechanism whereby co-released GABA acts as a first responder to dampen phasic-to-tonic dopamine signaling.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | - Ang D Sherpa
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Riccardo Melani
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Madeline R Wiseman
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Brian O'Neill
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Chiye Aoki
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Nicolas X Tritsch
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
50
|
Berndt A, Lee J, Won W, Kimball K, Neiswanger C, Schattauer S, Wang Y, Yeboah F, Ruiz M, Evitts K, Rappleye M, Bremner S, Chun C, Smith N, Mack D, Young J, Lee CJ, Chavkin C. Ultra-fast genetically encoded sensor for precise real-time monitoring of physiological and pathophysiological peroxide dynamics. RESEARCH SQUARE 2024:rs.3.rs-4048855. [PMID: 38585715 PMCID: PMC10996778 DOI: 10.21203/rs.3.rs-4048855/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Hydrogen Peroxide (H2O2) is a central oxidant in redox biology due to its pleiotropic role in physiology and pathology. However, real-time monitoring of H2O2 in living cells and tissues remains a challenge. We address this gap with the development of an optogenetic hydRogen perOxide Sensor (oROS), leveraging the bacterial peroxide binding domain OxyR. Previously engineered OxyR-based fluorescent peroxide sensors lack the necessary sensitivity and response speed for effective real-time monitoring. By structurally redesigning the fusion of Escherichia coli (E. coli) ecOxyR with a circularly permutated green fluorescent protein (cpGFP), we created a novel, green-fluorescent peroxide sensor oROS-G. oROS-G exhibits high sensitivity and fast on-and-off kinetics, ideal for monitoring intracellular H2O2 dynamics. We successfully tracked real-time transient and steady-state H2O2 levels in diverse biological systems, including human stem cell-derived neurons and cardiomyocytes, primary neurons and astrocytes, and mouse brain ex vivo and in vivo. These applications demonstrate oROS's capabilities to monitor H2O2 as a secondary response to pharmacologically induced oxidative stress and when adapting to varying metabolic stress. We showcased the increased oxidative stress in astrocytes via Aβ-putriscine-MAOB axis, highlighting the sensor's relevance in validating neurodegenerative disease models. Lastly, we demonstrated acute opioid-induced generation of H2O2 signal in vivo which highlights redox-based mechanisms of GPCR regulation. oROS is a versatile tool, offering a window into the dynamic landscape of H2O2 signaling. This advancement paves the way for a deeper understanding of redox physiology, with significant implications for understanding diseases associated with oxidative stress, such as cancer, neurodegenerative, and cardiovascular diseases.
Collapse
|