1
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2026; 21:248-264. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
2
|
Mulero-Franco J, Jimenez-Herrera R, Contreras A, Djebari S, Jiménez-Díaz L, Navarro-López JD. VU0810464, a selective GIRK channel activator, improves hippocampal-dependent synaptic plasticity and memory disrupted by amyloid-β oligomers. Biomed Pharmacother 2025; 189:118247. [PMID: 40516330 DOI: 10.1016/j.biopha.2025.118247] [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: 11/11/2024] [Revised: 05/30/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025] Open
Abstract
Selective pharmacological activation of G protein-gated inwardly rectifying K⁺ (GIRK) channels -a key physiological determinant of neuronal excitability control- has proven highly effective in counteracting amyloid-β oligomer (oAβ)-induced hyperexcitability and hippocampal dysfunction in early Alzheimer's disease (AD). However, GIRK gain-of-function in healthy animals disrupts learning, memory and underlying synaptic plasticity, greatly limiting its therapeutic potential in preclinical asymptomatic AD patients. Therefore, GIRK-based pharmacological treatment needs further investigation to overcome these limitations. Here we tested two doses of a novel, more potent, and neuronal selective GIRK activator, VU0810464, in healthy and intracerebroventricular (icv.) oAβ1-42-induced AD-like male and female mice. Both doses normalized: 1) at the synaptic level, CA3-CA1 long-term synaptic potentiation (LTP) in hippocampal slices, and 2) at the behavioral level, object location memory (OLM), a hippocampal-dependent spatial contextual recognition memory task. However, in healthy mice, while a low VU0810464 dose had no significant effect on hippocampal LTP and OLM, the higher dose impaired both processes. Importantly, these effects were consistent across sexes, as no sex differences were observed in any condition. Finally, VU0810464 reduced oAβ-induced hippocampal hyperexcitability, providing direct functional evidence of a mechanistic link between GIRK activation and restoration of network excitability. Our results suggest that the precise tuning of neural excitability with low dosing of VU0810464 might be a promising strategy to safely treat and prevent hippocampal overexcitation and upstreaming memory deficits in early preclinical asymptomatic phases of AD.
Collapse
Affiliation(s)
- Jaime Mulero-Franco
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Raquel Jimenez-Herrera
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Ana Contreras
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Souhail Djebari
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Lydia Jiménez-Díaz
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan D Navarro-López
- Neurophysiology & Behavior Lab, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
3
|
Wu B, Liu Y, Li H, Zhu L, Zeng L, Zhang Z, Peng W. Liver as a new target organ in Alzheimer's disease: insight from cholesterol metabolism and its role in amyloid-beta clearance. Neural Regen Res 2025; 20:695-714. [PMID: 38886936 PMCID: PMC11433892 DOI: 10.4103/1673-5374.391305] [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: 05/31/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease, the primary cause of dementia, is characterized by neuropathologies, such as amyloid plaques, synaptic and neuronal degeneration, and neurofibrillary tangles. Although amyloid plaques are the primary characteristic of Alzheimer's disease in the central nervous system and peripheral organs, targeting amyloid-beta clearance in the central nervous system has shown limited clinical efficacy in Alzheimer's disease treatment. Metabolic abnormalities are commonly observed in patients with Alzheimer's disease. The liver is the primary peripheral organ involved in amyloid-beta metabolism, playing a crucial role in the pathophysiology of Alzheimer's disease. Notably, impaired cholesterol metabolism in the liver may exacerbate the development of Alzheimer's disease. In this review, we explore the underlying causes of Alzheimer's disease and elucidate the role of the liver in amyloid-beta clearance and cholesterol metabolism. Furthermore, we propose that restoring normal cholesterol metabolism in the liver could represent a promising therapeutic strategy for addressing Alzheimer's disease.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Lingfeng Zeng
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Zhen Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
- Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei Province, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
4
|
Zhang X, Zhang Y, Zhang T, Wang J, Liu C, Shang Q, Wei X, Zhu H, Shen H, Sun B. HCN2 deficiency correlates with memory deficits and hyperexcitability of dCA1 pyramidal neurons in Alzheimer's disease. Alzheimers Res Ther 2025; 17:55. [PMID: 40016780 PMCID: PMC11866685 DOI: 10.1186/s13195-025-01704-y] [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: 09/18/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Abnormal excitability of hippocampal neurons may lead to dysfunction of neural circuits and then causes cognitive impairments in Alzheimer's disease (AD). However, the underlying mechanisms remain to be fully elucidated. METHODS Electrophysiology was performed to examine the intrinsic excitability of CA1 neurons and the activity of the hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs) of CA1 neurons in wild type (WT) and hAPP-J20 mice. The activity of CA1 pyramidal neurons (PNs) was modulated with chemogenetics. The activity of HCNs was regulated with nonselective facilitator (cAMP) or inhibitor (ZD7288) of HCNs. Immunohistochemical staining or western blotting were performed to examine the expression of HCN1 and HCN2 in the hippocampus of WT and hAPP-J20 mice, or AD patients and non-AD controls. AAVs were injected to specifically modulate the expression of HCN2 in dorsal CA1 (dCA1) PNs. Cognitive performance of mice was assessed with behavioral tests. RESULTS dCA1 PNs were more excitable in hAPP-J20 mice, but the excitability of PNs in the ventral CA1 (vCA1) or PV neurons was comparable between WT and hAPP-J20 mice. The activity of the HCNs was reduced in dCA1 PNs of hAPP-J20 mice, and pharmacologically increasing the activity of HCNs attenuated the hyperexcitability of dCA1 PNs in hAPP-J20 mice, suggesting that the reduced activity of HCNs is associated with the hyperexcitability of dCA1 PNs in hAPP-J20 mice. The expression of HCN2 but not HCN1 was reduced in the hippocampus of hAPP-J20 mice, and the expression of HCN2 was also reduced in the hippocampus of AD patients, suggesting that dysregulation of HCN2 is associated with the reduced activity of HCNs in AD. Overexpressing HCN2 rescued the activity of HCNs, attenuated the hyperexcitability of dCA1 PNs and improved memory of hAPP-J20 mice, and knocking down HCN2 impaired the function of HCNs, increased the excitability of dCA1 PNs and led to memory deficits in WT mice. CONCLUSIONS Our data suggest that dysregulation of HCNs, particularly HCN2, contributes to the abnormal excitability of CA1 PNs in AD mice and probably in AD patients as well, and thus provide new insights into the mechanisms underlying the aberrant activity or excitability of hippocampal neurons in AD.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Yiping Zhang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Ting Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jing Wang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Chang Liu
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang Province, 315211, China
| | - Xiaojie Wei
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Huaqiang Zhu
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang Province, 315100, China
| | - Haowei Shen
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Binggui Sun
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
5
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
6
|
Yu W, Zhang R, Zhang A, Mei Y. Deciphering the Functions of Raphe-Hippocampal Serotonergic and Glutamatergic Circuits and Their Deficits in Alzheimer's Disease. Int J Mol Sci 2025; 26:1234. [PMID: 39941002 PMCID: PMC11818420 DOI: 10.3390/ijms26031234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Subcortical innervation of the hippocampus by the raphe nucleus is essential for emotional and cognitive control. The two major afferents from raphe to hippocampus originate from serotonergic and glutamatergic neurons, of which the serotonergic control of hippocampal inhibitory network, theta activity, and synaptic plasticity have been extensively explored in the growing body of literature, whereas those of glutamatergic circuits have received little attention. Notably, both serotonergic and glutamatergic circuits between raphe and hippocampus are disrupted in Alzheimer's disease (AD), which may contribute to initiation and progression of behavioral and psychological symptoms of dementia. Thus, deciphering the mechanism underlying abnormal raphe-hippocampal circuits in AD is crucial to prevent dementia-associated emotional and cognitive symptoms. In this review, we summarize the anatomical, neurochemical, and electrophysiological diversity of raphe nuclei as well as the architecture of raphe-hippocampal circuitry. We then elucidate subcortical control of hippocampal activity by raphe nuclei and their role in regulation of emotion and cognition. Additionally, we present an overview of disrupted raphe-hippocampal circuits in AD pathogenesis and analyze the available therapies that can potentially be used clinically to alleviate the neuropsychiatric symptoms and cognitive decline in AD course.
Collapse
Affiliation(s)
| | | | | | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer’s Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
7
|
Zou Y, Wang C, Li H, Zhong M, Lin J, Hu Y, Chen Z, Gan CL. Epileptic seizures induced by pentylenetetrazole kindling accelerate Alzheimer-like neuropathology in 5×FAD mice. Front Pharmacol 2024; 15:1500105. [PMID: 39545066 PMCID: PMC11560768 DOI: 10.3389/fphar.2024.1500105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
Clinical studies have shown that epileptic seizures worsen Alzheimer's disease (AD) pathology and related cognitive deficits; however, the underlying mechanism is unclear. To assess the effects of seizures on the progression of AD, chronic temporal lobe epilepsy was induced in five familial AD mutation (5×FAD) mice by kindling with the chemoconvulsant pentylenetetrazole (PTZ) at 3-3.5 months of age. The amyloidogenic pathway, tauopathy, synaptic damage, neuronal death, neurological inflammatory response and associated kinase signaling pathway dysregulation were examined at 9 months of age. We found that APP, p-APP, BACE1, Aβ and kinase-associated p-tau levels were elevated after PTZ kindling in 5×FAD mice. In addition, PTZ kindling exacerbated hippocampal synaptic damage and neuronal cell death, as determined by scanning electron microscopy and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining, respectively. Finally, the levels of the neuroinflammation markers GFAP and Iba1, as well as the inflammatory cytokine IL-1β, were increased after PTZ insult. PTZ kindling profoundly exacerbated extracellular regulated kinase (ERK)-death-associated protein kinase (DAPK) signaling pathway overactivation, and acute ERK inhibitor treatment downregulated Aβ production and p-APP and p-tau levels in epileptic 5×FAD mice. In addition, long-term use of the antiseizure drug carbamazepine (CBZ) alleviated seizure-induced accelerated amyloid and tau pathology and ERK-DAPK overactivation in 5×FAD mice. Collectively, these results demonstrate that seizure-induced increases in AD-like neuropathology in 5×FAD mice are partially regulated by the ERK-DAPK pathway, suggesting that the ERK-DAPK axis could be a new therapeutic target for the treatment of AD patients with comorbid seizures.
Collapse
Affiliation(s)
- Yulian Zou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China
| | - Chengyan Wang
- Institute of Laboratory Animal Center, Fujian Medical University, Fuzhou, China
| | - Huang Li
- Department of Pharmacy of Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China
| | - Meihua Zhong
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Jin Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Hu
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhou Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Chen-Ling Gan
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
8
|
Stinson HE, Ninan I. Median raphe glutamatergic neuron-mediated enhancement of GABAergic transmission and suppression of long-term potentiation in the hippocampus. Heliyon 2024; 10:e38192. [PMID: 39386853 PMCID: PMC11462361 DOI: 10.1016/j.heliyon.2024.e38192] [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: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
The ascending neuromodulatory pathway from the median raphe nucleus (MRN) extends widely throughout midline/para-midline regions and robustly innervates the hippocampus. This neuromodulatory pathway is believed to be critical for regulating emotional and affective behaviors. Although the MRN primarily contains serotoninergic (5-HTergic), GABAergic, and glutamatergic neurons, glutamatergic neurons expressing vesicular glutamate transporter 3 (VGLUT3) form the primary MRN input to the hippocampus. Despite the earlier demonstration of the robust MRN VGLUT3 innervation of the hippocampus, little is known about how this MRN glutamatergic input modulates synaptic transmission and plasticity in the hippocampus. Our studies show that MRN VGLUT3 neurons activate serotonin 3a receptor (5-HT3aR)-expressing GABAergic neurons, including VGLUT3-expressing neurons, at the stratum radiatum (SR)/stratum lacunosum moleculare (SLM) border. This MRN VGLUT3 neuron-mediated glutamatergic transmission onto SR/SLM 5-HT3aR neurons is negatively regulated by 5-HT through 5-HT1B receptors. In agreement with the MRN VGLUT3 neuron-mediated activation of the 5-HT3aR GABAergic neurons, activation of MRN VGLUT3 projections induces a long-lasting increase in GABAergic transmission but not glutamatergic transmission in CA1 pyramidal neurons from male but not female mice. Consistent with the MRN VGLUT3 neuron-mediated enhancement of GABAergic transmission in male mice, activation of MRN VGLUT3 projections suppresses Schaffer collateral (SC)-CA1 long-term potentiation (LTP) in male but not female mice. Thus, our results show that MRN VGLUT3 neurons modulate the dorsal hippocampus by augmenting synaptic inhibition of CA1 pyramidal neurons and by suppressing SC-CA1 LTP in a sex-specific manner.
Collapse
Affiliation(s)
- Hannah E. Stinson
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ipe Ninan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
9
|
Yu CC, Wang XF, Wang J, Li C, Xiao J, Wang XS, Han R, Wang SQ, Lin YF, Kong LH, Du YJ. Electroacupuncture Alleviates Memory Deficits in APP/PS1 Mice by Targeting Serotonergic Neurons in Dorsal Raphe Nucleus. Curr Med Sci 2024; 44:987-1000. [PMID: 38990450 DOI: 10.1007/s11596-024-2908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Alzheimer's disease (AD) has become a significant global concern, but effective drugs able to slow down AD progression is still lacked. Electroacupuncture (EA) has been demonstrated to ameliorate cognitive impairment in individuals with AD. However, the underlying mechanisms remains poorly understood. This study aimed at examining the neuroprotective properties of EA and its potential mechanism of action against AD. METHODS APP/PS1 transgenic mice were employed to evaluate the protective effects of EA on Shenshu (BL 23) and Baihui (GV 20). Chemogenetic manipulation was used to activate or inhibit serotonergic neurons within the dorsal raphe nucleus (DRN). Learning and memory abilities were assessed by the novel object recognition and Morris water maze tests. Golgi staining, western blot, and immunostaining were utilized to determine EA-induced neuroprotection. RESULTS EA at Shenshu (BL 23) and Baihui (GV 20) effectively ameliorated learning and memory impairments in APP/PS1 mice. EA attenuated dendritic spine loss, increased the expression levels of PSD95, synaptophysin, and brain-derived neurotrophic factor in hippocampus. Activation of serotonergic neurons within the DRN can ameliorate cognitive deficits in AD by activating glutamatergic neurons mediated by 5-HT1B. Chemogenetic inhibition of serotonergic neurons in the DRN reversed the effects of EA on synaptic plasticity and memory. CONCLUSION EA can alleviate cognitive dysfunction in APP/PS1 mice by activating serotonergic neurons in the DRN. Further study is necessary to better understand how the serotonergic neurons-related neural circuits involves in EA-induced memory improvement in AD.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Xiao-Fei Wang
- Department of Rehabilitation, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430074, China
| | - Jia Wang
- Department of Acupuncture and Moxibustion, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, 430030, China
| | - Chu Li
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Juan Xiao
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xue-Song Wang
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, 050299, China
| | - Rui Han
- Department of Child Rehabilitation Medicine, Qujing Hospital of Maternity and Childcare, Qujing, 655002, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
10
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Wu X, Zhu X, Pan Y, Gu X, Liu X, Chen S, Zhang Y, Xu T, Xu N, Sun S. Amygdala neuronal dyshomeostasis via 5-HT receptors mediates mood and cognitive defects in Alzheimer's disease. Aging Cell 2024; 23:e14187. [PMID: 38716507 PMCID: PMC11320345 DOI: 10.1111/acel.14187] [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: 02/01/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 08/15/2024] Open
Abstract
Behavioral changes or neuropsychiatric symptoms (NPSs) are common features in dementia and are associated with accelerated cognitive impairment and earlier deaths. However, how NPSs are intertwined with cognitive decline remains elusive. In this study, we identify that the basolateral amygdala (BLA) is a key brain region that is associated with mood disorders and memory decline in the AD course. During the process from pre- to post-onset in AD, the dysfunction of parvalbumin (PV) interneurons and pyramidal neurons in the amygdala leads to hyperactivity of pyramidal neurons in the basal state and insensitivity to external stimuli. We further demonstrate that serotonin (5-HT) receptors in distinct neurons synergistically regulate the BLA microcircuit of AD rather than 5-HT levels, in which both restrained inhibitory inputs by excessive 5-HT1AR signaling in PV interneurons and depolarized pyramidal neurons via upregulated 5-HT2AR contribute to aberrant neuronal hyperactivity. Downregulation of these two 5-HT receptors simultaneously enables neurons to resist β-amyloid peptides (Aβ) neurotoxicity and ameliorates the mood and cognitive defects. Therefore, our study reveals a crucial role of 5-HT receptors for regulating neuronal homeostasis in AD pathogenesis, and this would provide early intervention and potential targets for AD cognitive decline.
Collapse
Affiliation(s)
- Xin‐Rong Wu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao‐Na Zhu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuan‐Bo Pan
- Department of Neurosurgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Gu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xian‐Dong Liu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Si Chen
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Zhang
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tian‐Le Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Nan‐Jie Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
- Songjiang Hospital and Songjiang Research InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Suya Sun
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
12
|
Chen M, Wang C, Lin Y, Chen Y, Xie W, Huang X, Zhang F, Fu C, Zhuang K, Zou T, Can D, Li H, Wu S, Luo C, Zhang J. Dorsal raphe nucleus-hippocampus serotonergic circuit underlies the depressive and cognitive impairments in 5×FAD male mice. Transl Neurodegener 2024; 13:34. [PMID: 39044270 PMCID: PMC11267773 DOI: 10.1186/s40035-024-00425-w] [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: 12/04/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Depressive symptoms often occur in patients with Alzheimer's disease (AD) and exacerbate the pathogenesis of AD. However, the neural circuit mechanisms underlying the AD-associated depression remain unclear. The serotonergic system plays crucial roles in both AD and depression. METHODS We used a combination of in vivo trans-synaptic circuit-dissecting anatomical approaches, chemogenetic manipulations, optogenetic manipulations, pharmacological methods, behavioral testing, and electrophysiological recording to investigate dorsal raphe nucleus serotonergic circuit in AD-associated depression in AD mouse model. RESULTS We found that the activity of dorsal raphe nucleus serotonin neurons (DRN5-HT) and their projections to the dorsal hippocampal CA1 (dCA1) terminals (DRN5-HT-dCA1CaMKII) both decreased in brains of early 5×FAD mice. Chemogenetic or optogenetic activation of the DRN5-HT-dCA1CaMKII neural circuit attenuated the depressive symptoms and cognitive impairments in 5×FAD mice through serotonin receptor 1B (5-HT1BR) and 4 (5-HT4R). Pharmacological activation of 5-HT1BR or 5-HT4R attenuated the depressive symptoms and cognitive impairments in 5×FAD mice by regulating the DRN5-HT-dCA1CaMKII neural circuit to improve synaptic plasticity. CONCLUSIONS These findings provide a new mechanistic connection between depression and AD and provide potential pharmaceutical prevention targets for AD.
Collapse
Affiliation(s)
- Meiqin Chen
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Chenlu Wang
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yinan Lin
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yanbing Chen
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wenting Xie
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xiaoting Huang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Fan Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Congrui Fu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Kai Zhuang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Tingting Zou
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Dan Can
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Huifang Li
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China.
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China.
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Institute of Neuroscience, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
13
|
Ogelman R, Gomez Wulschner LE, Hoelscher VM, Hwang IW, Chang VN, Oh WC. Serotonin modulates excitatory synapse maturation in the developing prefrontal cortex. Nat Commun 2024; 15:1368. [PMID: 38365905 PMCID: PMC10873381 DOI: 10.1038/s41467-024-45734-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Serotonin (5-HT) imbalances in the developing prefrontal cortex (PFC) are linked to long-term behavioral deficits. However, the synaptic mechanisms underlying 5-HT-mediated PFC development are unknown. We found that chemogenetic suppression and enhancement of 5-HT release in the PFC during the first two postnatal weeks decreased and increased the density and strength of excitatory spine synapses, respectively, on prefrontal layer 2/3 pyramidal neurons in mice. 5-HT release on single spines induced structural and functional long-term potentiation (LTP), requiring both 5-HT2A and 5-HT7 receptor signals, in a glutamatergic activity-independent manner. Notably, LTP-inducing 5-HT stimuli increased the long-term survival of newly formed spines ( ≥ 6 h) via 5-HT7 Gαs activation. Chronic treatment of mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first two weeks, but not the third week of postnatal development, increased the density and strength of excitatory synapses. The effect of fluoxetine on PFC synaptic alterations in vivo was abolished by 5-HT2A and 5-HT7 receptor antagonists. Our data describe a molecular basis of 5-HT-dependent excitatory synaptic plasticity at the level of single spines in the PFC during early postnatal development.
Collapse
Affiliation(s)
- Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria M Hoelscher
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria N Chang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
14
|
Wei X, Wang J, Yang E, Zhang Y, Qian Q, Li X, Huang F, Sun B. Efr3b is essential for social recognition by modulating the excitability of CA2 pyramidal neurons. Proc Natl Acad Sci U S A 2024; 121:e2314557121. [PMID: 38190534 PMCID: PMC10801834 DOI: 10.1073/pnas.2314557121] [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: 08/29/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
CA2 pyramidal neurons (PNs) are associated with social behaviors. The mechanisms, however, remain to be fully investigated. Here, we report that Efr3b, a protein essential for phospholipid metabolism at the plasma membrane, is widely expressed in the brain, especially in the hippocampal CA2/CA3 areas. To assess the functional significance of Efr3b in the brain, we generated Efr3bf/f mice and crossed them with Nestin-cre mice to delete Efr3b specifically in the brain. We find that Efr3b deficiency in the brain leads to deficits of social novelty recognition and hypoexcitability of CA2 PNs. We then knocked down the expression of Efr3b specifically in CA2 PNs of C57BL/6J mice, and our results showed that reducing Efr3b in CA2 PNs also resulted in deficits of social novelty recognition and hypoexcitability of CA2 PNs. More interestingly, restoring the expression of Efr3b in CA2 PNs enhances their excitability and improves social novelty recognition in Efr3b-deficient mice. Furthermore, direct activation of CA2 PNs with chemogenetics improves social behaviors in Efr3b-deficient mice. Together, our data suggest that Efr3b is essential for social novelty by modulating the excitability of CA2 PNs.
Collapse
Affiliation(s)
- Xiaojie Wei
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
- Children’s Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310020, China
| | - Jing Wang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
- School of Medicine, Shaoxing University, Shaoxing312000, China
| | - Enlu Yang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Yiping Zhang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Qi Qian
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Xuekun Li
- Children’s Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310020, China
| | - Fude Huang
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing100190, China
- Nuo-Beta Pharmaceutical Technology, Co. Ltd., Shanghai201210, China
| | - Binggui Sun
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| |
Collapse
|