1
|
Deng F, Yang D, Qing L, Chen Y, Zou J, Jia M, Wang Q, Jiang R, Huang L. Exploring the interaction between the gut microbiota and cyclic adenosine monophosphate-protein kinase A signaling pathway: a potential therapeutic approach for neurodegenerative diseases. Neural Regen Res 2025; 20:3095-3112. [PMID: 39589173 PMCID: PMC11881707 DOI: 10.4103/nrr.nrr-d-24-00607] [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: 06/01/2024] [Revised: 08/07/2024] [Accepted: 09/10/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut microbiota and cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling pathway in the host's central nervous system plays a crucial role in neurological diseases and enhances communication along the gut-brain axis. The gut microbiota influences the cAMP-PKA signaling pathway through its metabolites, which activates the vagus nerve and modulates the immune and neuroendocrine systems. Conversely, alterations in the cAMP-PKA signaling pathway can affect the composition of the gut microbiota, creating a dynamic network of microbial-host interactions. This reciprocal regulation affects neurodevelopment, neurotransmitter control, and behavioral traits, thus playing a role in the modulation of neurological diseases. The coordinated activity of the gut microbiota and the cAMP-PKA signaling pathway regulates processes such as amyloid-β protein aggregation, mitochondrial dysfunction, abnormal energy metabolism, microglial activation, oxidative stress, and neurotransmitter release, which collectively influence the onset and progression of neurological diseases. This study explores the complex interplay between the gut microbiota and cAMP-PKA signaling pathway, along with its implications for potential therapeutic interventions in neurological diseases. Recent pharmacological research has shown that restoring the balance between gut flora and cAMP-PKA signaling pathway may improve outcomes in neurodegenerative diseases and emotional disorders. This can be achieved through various methods such as dietary modifications, probiotic supplements, Chinese herbal extracts, combinations of Chinese herbs, and innovative dosage forms. These findings suggest that regulating the gut microbiota and cAMP-PKA signaling pathway may provide valuable evidence for developing novel therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Fengcheng Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lingxi Qing
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yifei Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jilian Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Meiling Jia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Qian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Runda Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihua Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Jin S, Bellier J, Wells A, LIopis PM, Anekal PV, Tresback JS, Caldarone BJ, Liu L, Li S, Dettmer U, Ramalingam N, Selkoe DJ. Inhibition of hippocampal mossy fiber plasticity and episodic memory by human Aβ oligomers is prevented by enhancing cAMP signaling in Alzheimer's mice. Alzheimers Dement 2025; 21:e70194. [PMID: 40302031 PMCID: PMC12040739 DOI: 10.1002/alz.70194] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION Early episodic memory impairment in Alzheimer's disease (AD) is linked to synaptic dysfunction from amyloid β-protein oligomers (oAβ), particularly affecting the dentate gyrus mossy fiber-CA3 pathway. The APPNL-G-F mouse model exhibits early deficits in mossy fiber long-term potentiation (mf-LTP). METHODS We administered the β-adrenergic receptor agonist isoproterenol (ISO) in vivo and phosphodiesterase type 4 inhibitor GSK356278 in vitro to assess their impact on mf-LTP and contextual fear memory. Fluorescence lifetime imaging (FLIM)-Förster resonance energy transfer (FRET) microscopy was used to visualize impaired and rescued cyclic adenosine monophosphate (cAMP) signaling in dentate gyrus neurons. RESULTS ISO prevented mf-LTP impairment at 3-4 mo and improved memory by 7 mo. GSK356278 inhibited mf-LTP deficits in a dose-dependent manner. ISO also reduced hyperphosphorylation of synapsin I and microgliosis. DISCUSSION These findings suggest that β-AR activation and phosphodiesterase 4 (PDE4) inhibition mitigate oAβ-induced memory deficits, supporting enhanced cAMP signaling as a therapeutic target for early AD. HIGHLIGHTS Early episodic memory deficits in AD linked to oAβ-induced synaptic dysfunction. Isoproterenol and GSK356278 improve mossy fiber-LTP and fear memory deficits. FLIM-FRET shows treatments restore cAMP signaling in dentate gyrus neurons. Isoproterenol reduces synapsin I hyperphosphorylation and microgliosis. Enhancing cAMP signaling may help mitigate early memory deficits in AD.
Collapse
Affiliation(s)
- Shan‐Xue Jin
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Jean‐Pierre Bellier
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | | | | | | | - Jason S. Tresback
- Center for Nanoscale SystemsHarvard UniversityCambridgeMassachusettsUSA
| | | | - Lei Liu
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Shaomin Li
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Dennis J. Selkoe
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
3
|
Kumari S, Bagri K, Deshmukh R. Connecting dots: Preclinical foundations to clinical realities of PDE4 inhibitors in Alzheimer's disease. Inflammopharmacology 2025; 33:593-603. [PMID: 39808238 DOI: 10.1007/s10787-024-01638-1] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.
Collapse
Affiliation(s)
- Shilpa Kumari
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Kajal Bagri
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Rahul Deshmukh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India.
| |
Collapse
|
4
|
Pan G, Chai L, Chen R, Yuan Q, Song Z, Feng W, Wei J, Yang Z, Zhang Y, Xie G, Yan A, Lv Q, Wang C, Zhao Y, Wang Y. Potential mechanism of Qinggong Shoutao pill alleviating age-associated memory decline based on integration strategy. PHARMACEUTICAL BIOLOGY 2024; 62:105-119. [PMID: 38145345 PMCID: PMC10763866 DOI: 10.1080/13880209.2023.2291689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 11/30/2023] [Indexed: 12/26/2023]
Abstract
CONTEXT Qinggong Shoutao Wan (QGSTW) is a pill used as a traditional medicine to treat age-associated memory decline (AAMI). However, its potential mechanisms are unclear. OBJECTIVE This study elucidates the possible mechanisms of QGSTW in treating AAMI. MATERIALS AND METHODS Network pharmacology and molecular docking approaches were utilized to identify the potential pathway by which QGSTW alleviates AAMI. C57BL/6J mice were divided randomly into control, model, and QGSTW groups. A mouse model of AAMI was established by d-galactose, and the pathways that QGSTW acts on to ameliorate AAMI were determined by ELISA, immunofluorescence staining and Western blotting after treatment with d-gal (100 mg/kg) and QGSTW (20 mL/kg) for 12 weeks. RESULTS Network pharmacology demonstrated that the targets of the active components were significantly enriched in the cAMP signaling pathway. AKT1, FOS, GRIN2B, and GRIN1 were the core target proteins. QGSTW treatment increased the discrimination index from -16.92 ± 7.06 to 23.88 ± 15.94% in the novel location test and from -19.54 ± 5.71 to 17.55 ± 6.73% in the novel object recognition test. ELISA showed that QGSTW could increase the levels of cAMP. Western blot analysis revealed that QGSTW could upregulate the expression of PKA, CREB, c-Fos, GluN1, GluA1, CaMKII-α, and SYN. Immunostaining revealed that the expression of SYN was decreased in the CA1 and DG. DISCUSSION AND CONCLUSIONS This study not only provides new insights into the mechanism of QGSTW in the treatment of AAMI but also provides important information and new research ideas for the discovery of traditional Chinese medicine compounds that can treat AAMI.
Collapse
Affiliation(s)
- Guiyun Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijuan Chai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanying Feng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinna Wei
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhang Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - An Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Lv
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Caijun Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingqiang Zhao
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
5
|
Yao W, Hou X, Zhou H, You S, Lv T, Chen H, Yang Z, Chen C, Bai F. Associations between the multitrajectory neuroplasticity of neuronavigated rTMS-mediated angular gyrus networks and brain gene expression in AD spectrum patients with sleep disorders. Alzheimers Dement 2024; 20:7885-7901. [PMID: 39324544 PMCID: PMC11567849 DOI: 10.1002/alz.14255] [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: 04/24/2024] [Accepted: 08/18/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The multifactorial influence of repetitive transcranial magnetic stimulation (rTMS) on neuroplasticity in neural networks is associated with improvements in cognitive dysfunction and sleep disorders. The mechanisms of rTMS and the transcriptional-neuronal correlation in Alzheimer's disease (AD) patients with sleep disorders have not been fully elucidated. METHODS Forty-six elderly participants with cognitive impairment (23 patients with low sleep quality and 23 patients with high sleep quality) underwent 4-week periods of neuronavigated rTMS of the angular gyrus and neuroimaging tests, and gene expression data for six post mortem brains were collected from another database. Transcription-neuroimaging association analysis was used to evaluate the effects on cognitive dysfunction and the underlying biological mechanisms involved. RESULTS Distinct variable neuroplasticity in the anterior and posterior angular gyrus networks was detected in the low sleep quality group. These interactions were associated with multiple gene pathways, and the comprehensive effects were associated with improvements in episodic memory. DISCUSSION Multitrajectory neuroplasticity is associated with complex biological mechanisms in AD-spectrum patients with sleep disorders. HIGHLIGHTS This was the first transcription-neuroimaging study to demonstrate that multitrajectory neuroplasticity in neural circuits was induced via neuronavigated rTMS, which was associated with complex gene expression in AD-spectrum patients with sleep disorders. The interactions between sleep quality and neuronavigated rTMS were coupled with multiple gene pathways and improvements in episodic memory. The present strategy for integrating neuroimaging, rTMS intervention, and genetic data provide a new approach to comprehending the biological mechanisms involved in AD.
Collapse
Affiliation(s)
- Weina Yao
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Xinle Hou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Huijuan Zhou
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Shengqi You
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Tingyu Lv
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Haifeng Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Zhiyuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Chang Chen
- School of Elderly Care Services and ManagementNanjing University of Chinese MedicineNanjingChina
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Geriatric Medicine Center, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Geriatric MedicineMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
6
|
Marneffe C, Moreira-de-Sá A, Lecomte S, Erhardt A, Mulle C. Short term plasticity at hippocampal mossy fiber synapses. Neuroscience 2024:S0306-4522(24)00497-4. [PMID: 39332701 DOI: 10.1016/j.neuroscience.2024.09.044] [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: 02/28/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Short-term synaptic plasticity refers to the regulation of synapses by their past activity on time scales of milliseconds to minutes. Hippocampal mossy fiber synapses onto CA3 pyramidal cells (Mf-CA3 synapses) are endowed with remarkable forms of short-term synaptic plasticity expressed as facilitation of synaptic release by a factor of up to ten-fold. Three main forms of short-term plasticity are distinguished: 1) Frequency facilitation, which includes low frequency facilitation and train facilitation, operating in the range of tens of milliseconds to several seconds; 2) Post-tetanic potentiation triggered by trains of high frequency stimulation, which lasts several minutes; 3) Finally, depolarization-induced potentiation of excitation, based on retrograde signaling, with an onset and offset of several minutes. Here we describe the proposed mechanisms for short-term plasticity, mainly based on the kinetics of presynaptic Ca2+ transients and the Ca2+ sensor synaptotagmin 7, on cAMP-dependent mechanisms and readily releasable vesicle pool, and on the regulation of presynaptic K+ channels. We then review evidence for a physiological function of short-term plasticity of Mf-CA3 synapses in information transfer between the dentate gyrus and CA3 in conditions of natural spiking, and discuss how short-term plasticity counteracts robust feedforward inhibition in a frequency-dependent manner. Although DG-CA3 connections have long been proposed to play a role in memory, direct evidence for an implication of short-term plasticity at Mf-CA3 synapses is mostly lacking. The mechanistic knowledge gained on short-term plasticity at Mf-CA3 synapses should help in designing future experiments to directly test how this evolutionary conserved feature controls hippocampal circuit function in behavioural conditions.
Collapse
Affiliation(s)
- Catherine Marneffe
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Ana Moreira-de-Sá
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Simon Lecomte
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Anaël Erhardt
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France.
| |
Collapse
|
7
|
Mitias S, Schaffer N, Nair S, Hook C, Lindberg I. ProSAAS is preferentially up-regulated during homeostatic scaling and reduces amyloid plaque burden in the 5xFAD mouse hippocampus. J Neurochem 2024; 168:3235-3249. [PMID: 39115041 PMCID: PMC11449639 DOI: 10.1111/jnc.16193] [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: 12/20/2023] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 09/21/2024]
Abstract
The accumulation of β-amyloid in Alzheimer's disease greatly impacts neuronal health and synaptic function. To maintain network stability in the face of altered synaptic activity, neurons engage a feedback mechanism termed homeostatic scaling; however, this process is thought to be disrupted during disease progression. Previous proteomics studies have shown that one of the most highly regulated proteins in cell culture models of homeostatic scaling is the small secretory chaperone proSAAS. Our prior work has shown that proSAAS exhibits anti-aggregant behavior against alpha-synuclein and β-amyloid fibrillation in vitro and is up-regulated in cell models of proteostatic stress. However, the specific role that this protein might play in homeostatic scaling, and its anti-aggregant role in Alzheimer's progression, is not clear. To learn more about the role of proSAAS in maintaining hippocampal proteostasis, we compared its expression in a primary neuron model of homeostatic scaling to other synaptic components using western blotting and qPCR, revealing that proSAAS protein responses to homeostatic up- and down-regulation were significantly higher than those of two other synaptic vesicle components, 7B2 and carboxypeptidase E. However, proSAAS mRNA expression was static, suggesting translational control and/or altered protein degradation. ProSAAS was readily released upon depolarization of differentiated hippocampal cultures, supporting its synaptic localization. Immunohistochemical analysis demonstrated abundant proSAAS within the mossy fiber layer of the hippocampus in both wild-type and 5xFAD mice; in the latter, proSAAS was also concentrated around amyloid plaques. Importantly, overexpression of proSAAS in the CA1 region via stereotaxic injection of proSAAS-encoding AAV2/1 significantly decreased amyloid plaque burden in 5xFAD mice. We hypothesize that dynamic changes in proSAAS expression play a critical role in hippocampal proteostatic processes, both in the context of normal homeostatic plasticity and in the control of protein aggregation during Alzheimer's disease progression.
Collapse
Affiliation(s)
- Samira Mitias
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Nicholas Schaffer
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Saaya Nair
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Chelsea Hook
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Iris Lindberg
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
8
|
Donders Z, Skorupska IJ, Willems E, Mussen F, Broeckhoven JV, Carlier A, Schepers M, Vanmierlo T. Beyond PDE4 inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed Pharmacother 2024; 177:117009. [PMID: 38908196 DOI: 10.1016/j.biopha.2024.117009] [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: 03/28/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.
Collapse
Affiliation(s)
- Zoë Donders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Iga Joanna Skorupska
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Emily Willems
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Femke Mussen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
9
|
Mitias S, Schaffer N, Nair S, Hook C, Lindberg I. ProSAAS is Preferentially Secreted from Neurons During Homeostatic Scaling and Reduces Amyloid Plaque Size in the 5xFAD Mouse Hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590133. [PMID: 38712265 PMCID: PMC11071301 DOI: 10.1101/2024.04.18.590133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The accumulation of β-amyloid in Alzheimer's disease greatly impacts neuronal health and synaptic function. To maintain network stability in the face of altered synaptic activity, neurons engage a feedback mechanism termed homeostatic scaling; however, this process is thought to be disrupted during disease progression. Previous proteomics studies have shown that one of the most highly regulated proteins in cell culture models of homeostatic scaling is the small secretory chaperone proSAAS. Our prior work has shown that proSAAS exhibits anti-aggregant behavior against alpha synuclein and β-amyloid fibrillation in vitro, and is upregulated in cell models of proteostatic stress. However, the specific role that this protein might play in homeostatic scaling, and its anti-aggregant role in Alzheimer's progression, is not clear. To learn more about the role of proSAAS in maintaining hippocampal proteostasis, we compared its expression in a primary neuron model of homeostatic scaling to other synaptic components using Western blotting and qPCR, revealing that proSAAS protein responses to homeostatic up- and down-regulation were significantly higher than those of two other synaptic vesicle components, 7B2 and carboxypeptidase E. However, proSAAS mRNA expression was static, suggesting translational control (and/or reduced degradation). ProSAAS was readily released upon depolarization of differentiated hippocampal cultures, supporting its synaptic localization. Immunohistochemical analysis demonstrated abundant proSAAS within the mossy fiber layer of the hippocampus in both wild-type and 5xFAD mice; in the latter, proSAAS was also concentrated around amyloid plaques. Interestingly, overexpression of proSAAS in the CA1 region via stereotaxic injection of proSAAS-encoding AAV2/1 significantly decreased amyloid plaque burden in 5xFAD mice. We hypothesize that dynamic changes in proSAAS expression play a critical role in hippocampal proteostatic processes, both in the context of normal homeostatic plasticity and in the control of protein aggregation during Alzheimer's disease progression.
Collapse
Affiliation(s)
- Samira Mitias
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Nicholas Schaffer
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Saaya Nair
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Chelsea Hook
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| | - Iris Lindberg
- Dept. of Neurobiology, Univ. of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
Vandael D, Jonas P. Structure, biophysics, and circuit function of a "giant" cortical presynaptic terminal. Science 2024; 383:eadg6757. [PMID: 38452088 DOI: 10.1126/science.adg6757] [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: 10/19/2023] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells and dendrites of CA3 pyramidal neurons, is a key synapse in the trisynaptic circuitry of the hippocampus. Because of its comparatively large size, this synapse is accessible to direct presynaptic recording, allowing a rigorous investigation of the biophysical mechanisms of synaptic transmission and plasticity. Furthermore, because of its placement in the very center of the hippocampal memory circuit, this synapse seems to be critically involved in several higher network functions, such as learning, memory, pattern separation, and pattern completion. Recent work based on new technologies in both nanoanatomy and nanophysiology, including presynaptic patch-clamp recording, paired recording, super-resolution light microscopy, and freeze-fracture and "flash-and-freeze" electron microscopy, has provided new insights into the structure, biophysics, and network function of this intriguing synapse. This brings us one step closer to answering a fundamental question in neuroscience: how basic synaptic properties shape higher network computations.
Collapse
Affiliation(s)
- David Vandael
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| |
Collapse
|
11
|
Ding J, Wu J, Hou X, Yang L, Gao Y, Zheng J, Jia N, He Z, Zhang H, Wang C, Qi X, Huang J, Pei X, Wang J. α-synuclein-lack expression rescues methamphetamine-induced mossy fiber degeneration in dorsal hippocampal CA3. Neurotoxicology 2024; 101:36-45. [PMID: 38311184 DOI: 10.1016/j.neuro.2024.01.005] [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: 04/24/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Methamphetamine (METH) - induced cognitive impairments may be related to synaptic degeneration at mossy fiber terminals, critical for spatial memory formation in hippocampal circuits. We have previously found METH-induced neurodegeneration in the striatum by increasing the α-synuclein (α-SYN) level. However, whether and how the METH-induced mossy fiber degeneration is also blamed for the abnormal accumulation of α-SYN remains to be elucidated. Chronic METH exposure decreased mossy fiber density but upregulatedα-SYN and phosphorylated TAU (TAU-pSer396) in hippocampal CA3, associated with glial cell overactivation, axonal neuropathies, and memory impairment. Notably, the knockout of the α-SYN gene significantly alleviated the METH-induced mossy fiber degeneration and memory impairment. Meanwhile, the TAU-pSer396 accumulation and glial activation were ameliorated by α-SYN knockout. Our findings suggest an essential role of α-SYN in mediating METH-induced mossy fiber degeneration, providing promising therapeutic and prophylactic targets for METH-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China; Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jun Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaotao Hou
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China; Guangdong Provincial Key Laboratory of Genetic Disease Diagnostic, Guangzhou, China
| | - Li Yang
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Yingdong Gao
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Juan Zheng
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Nannan Jia
- Neonatal Screening Center, Taian Maternity and Child Health Hospital, Taian, China
| | - Zheng He
- Neonatal Screening Center, Taian Maternity and Child Health Hospital, Taian, China
| | - Hui Zhang
- Department of Reproductive Medicine, Taian Maternity and Child Health Hospital, Taian, China
| | - Chengfei Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xianglin Pei
- School of Materials and Architectural Engineering, Guizhou Normal University, Guiyang China.
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
12
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
13
|
Pluta R, Kocki J, Bogucki J, Bogucka-Kocka A, Czuczwar SJ. LRP1 and RAGE Genes Transporting Amyloid and Tau Protein in the Hippocampal CA3 Area in an Ischemic Model of Alzheimer's Disease with 2-Year Survival. Cells 2023; 12:2763. [PMID: 38067191 PMCID: PMC10706460 DOI: 10.3390/cells12232763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Explaining changes at the gene level that occur during neurodegeneration in the CA3 area is crucial from the point of view of memory impairment and the development of post-ischemic dementia. An ischemic model of Alzheimer's disease was used to evaluate changes in the expression of genes related to amyloid transport in the CA3 region of the hippocampus after 10 min of brain ischemia with survival of 2, 7 and 30 days and 12, 18 and 24 months. The quantitative reverse transcriptase PCR assay revealed that the expression of the LRP1 and RAGE genes involved in amyloid transport was dysregulated from 2 days to 24 months post-ischemia in the CA3 area of the hippocampus. LRP1 gene expression 2 and 7 days after ischemia was below control values. However, its expression from day 30 to 24 months, survival after an ischemic episode was above control values. RAGE gene expression 2 days after ischemia was below control values, reaching a maximum increase 7 and 30 days post-ischemia. Then, after 12, 18 and 24 months, it was again below the control values. The data indicate that in the CA3 area of the hippocampus, an episode of brain ischemia causes the increased expression of the RAGE gene for 7-30 days during the acute phase and that of LRP1 from 1 to 24 months after ischemia during the chronic stage. In other words, in the early post-ischemic stage, the expression of the gene that transport amyloid to the brain increases (7-30 days). Conversely, in the late post-ischemic stage, amyloid scavenging/cleaning gene activity increases, reducing and/or preventing further neuronal damage or facilitating the healing of damaged sites. This is how the new phenomenon of pyramidal neuronal damage in the CA3 area after ischemia is defined. In summary, post-ischemic modification of the LRP1 and RAGE genes is useful in the study of the ischemic pathways and molecular factors involved in the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Jacek Bogucki
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, 20-093 Lublin, Poland;
| | | |
Collapse
|
14
|
Yang J, Yuan S, Jian Y, Lei Y, Hu Z, Yang Q, Yan X, Zheng L, Li J, Liu W. Aerobic exercise regulates GPR81 signal pathway and mediates complement- microglia axis homeostasis on synaptic protection in the early stage of Alzheimer's disease. Life Sci 2023; 331:122042. [PMID: 37634815 DOI: 10.1016/j.lfs.2023.122042] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
AIMS Memory impairment is a major clinical manifestation in Alzheimer's disease (AD) patients, while regular exercise may prevent and delay degenerative changes in memory functions, and our aim is to explore the influence and molecular mechanisms of aerobic exercise on the early stages of Alzheimer's disease. MAIN METHODS 3-month-old male APP/PS1 transgenic AD mice and C57BL/6J wild-type mice were randomly divided into four groups: wild-type and APP/PS1 mice with sedentary (WT-SED, AD-SED), and running (WT-RUN, AD-RUN) for 12-weeks. The spatial learning and memory function, RNA-sequencing, spine density, synaptic associated protein, mRNA and protein expression involved in G protein-coupled receptor 81 (GPR81) signaling pathway, and complement factors in brain were measured. KEY FINDINGS Aerobic exercise improved spatial learning and memory in APP/PS1 mice, potentially attributed to increased dendritic spine density. Subsequently, potential underlying mechanisms were identified through RNA sequencing: regular aerobic exercise could activate the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) cAMP/PKA signaling pathway and upregulate synaptic function-related proteins to promote synaptic growth, possibly by modulating GPR81. Notably, regular aerobic exercise inhibited microglial activation, reversed the microglial phenotype, reduced the production of initiation factor C1q and central factor C3 in the complement cascade in the brain, prevented the colocalization of microglia and PSD-95, and thus prevented synaptic loss. SIGNIFICANCE Physical exercise could play a critical role in improving cognitive function in AD by promoting synaptic growth and preventing synaptic loss, which may be related to the regulation of the GPR81/cAMP/PKA signaling pathway and inhibition of complement-mediated microglial phagocytosis of synapses.
Collapse
Affiliation(s)
- Jialun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yong Lei
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zelin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Qiming Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xinjun Yan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Lan Zheng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Jianghua Li
- College of Physical Education, Jiangxi Normal University, Nanchang 330022, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
15
|
Tian L, Zhao T, Dong L, Liu Q, Zheng Y. Passive array micro-magnetic stimulation device based on multi-carrier wireless flexible control for magnetic neuromodulation. J Neural Eng 2023; 20:056020. [PMID: 37714145 DOI: 10.1088/1741-2552/acfa23] [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: 05/25/2023] [Accepted: 09/15/2023] [Indexed: 09/17/2023]
Abstract
Objective.The passive micro-magnetic stimulation (µMS) devices typically consist of an external transmitting coil and a single internal micro-coil, which enables a point-to-point energy supply from the external coil to the internal coil and the realization of magnetic neuromodulation via wireless energy transmission. The internal array of micro coils can achieve multi-target stimulation without movement, which improves the focus and effectiveness of magnetic stimulations. However, achieving a free selection of an appropriate external coil to deliver energy to a particular internal array of micro-coils for multiple stimulation targets has been challenging. To address this challenge, this study uses a multi-carrier modulation technique to transmit the energy of the external coil.Approach.In this study, a theoretical model of a multi-carrier resonant compensation network for the arrayµMS is established based on the principle of magnetically coupled resonance. The resonant frequency coupling parameter corresponding to each micro-coil of the arrayµMS is determined, and the magnetic field interference between the external coil and its non-resonant micro-coils is eliminated. Therefore, an effective magnetic stimulation threshold for a micro-coil corresponding to the target is determined, and wireless free control of the internal micro-coil array is achieved by using an external transmitting coil.Main results.The passiveµMS array model is designed using a multi-carrier wireless modulation method, and its synergistic modulation of the magnetic stimulation of synaptic plasticity long-term potentiation in multiple hippocampal regions is investigated using hippocampal isolated brain slices.Significance.The results presented in this study could provide theoretical and experimental bases for implantable micro-magnetic device-targeted therapy, introducing an efficient method for diagnosis and treatment of neurological diseases and providing innovative ideas for in-depth application of micro-magnetic stimulation in the neuroscience field.
Collapse
Affiliation(s)
- Lei Tian
- Department of Biomedical Engineering, Tiangong University, Tian Jin, People's Republic of China
| | - Tong Zhao
- Department of Biomedical Engineering, Tiangong University, Tian Jin, People's Republic of China
| | - Lei Dong
- Department of Biomedical Engineering, Tiangong University, Tian Jin, People's Republic of China
| | - Qiwen Liu
- Department of Biomedical Engineering, Tiangong University, Tian Jin, People's Republic of China
| | - Yu Zheng
- Department of Biomedical Engineering, Tiangong University, Tian Jin, People's Republic of China
| |
Collapse
|
16
|
Stern AM, Yang Y, Jin S, Yamashita K, Meunier AL, Liu W, Cai Y, Ericsson M, Liu L, Goedert M, Scheres SHW, Selkoe DJ. Abundant Aβ fibrils in ultracentrifugal supernatants of aqueous extracts from Alzheimer's disease brains. Neuron 2023; 111:2012-2020.e4. [PMID: 37167969 PMCID: PMC10330525 DOI: 10.1016/j.neuron.2023.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/21/2023] [Accepted: 04/07/2023] [Indexed: 05/13/2023]
Abstract
Soluble oligomers of amyloid β-protein (Aβ) have been defined as aggregates in supernatants following ultracentrifugation of aqueous extracts from Alzheimer's disease (AD) brains and are believed to be upstream initiators of synaptic dysfunction, but little is known about their structures. We now report the unexpected presence of Aβ fibrils in synaptotoxic high-speed supernatants from AD brains extracted by soaking in an aqueous buffer. The fibrils did not appear to form during preparation, and their counts by EM correlated with Aβ ELISA quantification. Cryo-EM structures of aqueous Aβ fibrils were identical to those from sarkosyl-insoluble homogenates. The fibrils in aqueous extracts were labeled by lecanemab, an Aβ aggregate-directed antibody reported to improve AD cognitive outcomes. Lecanemab provided protection against aqueous fibril synaptotoxicity. We conclude that fibrils are abundant in aqueous extracts from AD brains and have the same structures as those from plaques. These findings have implications for AD pathogenesis and drug design.
Collapse
Affiliation(s)
- Andrew M Stern
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yang Yang
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Shanxue Jin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Keitaro Yamashita
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Angela L Meunier
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yuqi Cai
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Maria Ericsson
- Harvard Medical School Electron Microscopy Facility, Boston, MA 02115, USA
| | - Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Sjors H W Scheres
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|