1
|
Cai Y, Wang T. Regulation of presynaptic homeostatic plasticity by glial signalling in Alzheimer's disease. J Physiol 2024. [PMID: 39705214 DOI: 10.1113/jp286751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/04/2024] [Indexed: 12/22/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia among the elderly, affects numerous individuals worldwide. Despite advances in understanding the molecular underpinnings of AD pathology, effective treatments to prevent or cure the disease remain elusive. AD is characterized not only by pathological hallmarks such as amyloid plaques and neurofibrillary tangles but also by impairments in synaptic physiology, circuit activity and cognitive function. Synaptic homeostatic plasticity plays a vital role in maintaining the stability of synaptic and neural functions amid genetic and environmental disturbances. A key component of this regulation is presynaptic homeostatic potentiation, where increased presynaptic neurotransmitter release compensates for reduced postsynaptic glutamate receptor functionality, thereby stabilizing neuronal excitability. The role of presynaptic homeostatic plasticity in synapse stabilization in AD, however, remains unclear. Moreover, recent advances in transcriptomics have illuminated the complex roles of glial cells in regulating synaptic function in ageing brains and in the progression of neurodegenerative diseases. Yet, the impact of AD-related abnormalities in glial signalling on synaptic homeostatic plasticity has not been fully delineated. This review discusses recent findings on how glial dysregulation in AD affects presynaptic homeostatic plasticity. There is increasing evidence that disrupted glial signalling, particularly through aberrant histone acetylation and transcriptomic changes in glia, compromises this plasticity in AD. Notably, the sphingosine signalling pathway has been identified as being protective in stabilizing synaptic physiology through epigenetic and homeostatic mechanisms, presenting potential therapeutic targets for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Yimei Cai
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Tingting Wang
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA
| |
Collapse
|
2
|
Rao NR, DeGulis O, Nomura T, Lee S, Hark TJ, Dynes JC, Dexter EX, Dulewicz M, Ge J, Upadhyay A, Fornasiero EF, Vassar R, Hanrieder J, Contractor A, Savas JN. Levetiracetam prevents Aβ 42 production through SV2a-dependent modulation of App processing in Alzheimer's disease models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620698. [PMID: 39554163 PMCID: PMC11565754 DOI: 10.1101/2024.10.28.620698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
In Alzheimer's disease (AD), amyloid-beta (Aβ) peptides are produced by proteolytic cleavage of the amyloid precursor protein (APP), which can occur during synaptic vesicle (SV) cycling at presynapses. Precisely how amyloidogenic APP processing may impair presynaptic proteostasis and how to therapeutically target this process remains poorly understood. Using App knock-in mouse models of early Aβ pathology, we found proteins with hampered degradation accumulate at presynaptic sites. At this mild pathological stage, amyloidogenic processing leads to accumulation of Aβ42 inside SVs. To explore if targeting SVs modulates Aβ accumulation, we investigated levetiracetam (Lev), a SV-binding small molecule drug that has shown promise in mitigating AD-related pathologies despite its mechanism of action being unclear. We discovered Lev reduces Aβ42 levels by decreasing amyloidogenic processing of APP in a SV2a-dependent manner. Lev corrects SV protein levels and cycling, which results in increased surface localization of APP, where it favors processing via the non-amyloidogenic pathway. Using metabolic stable isotopes and mass spectrometry we confirmed that Lev prevents the production of Aβ42 in vivo. In transgenic mice with aggressive pathology, electrophysiological and immunofluorescent microscopy analyses revealed that Lev treatment reduces SV cycling and minimizes synapse loss. Finally, we found that human Down syndrome brains with early Aβ pathology, have elevated levels of presynaptic proteins, confirming a comparable presynaptic deficit in human brains. Taken together, we report a mechanism that highlights the therapeutic potential of Lev to modify the early stages of AD and represent a promising strategy to prevent Aβ42 pathology before irreversible damage occurs.
Collapse
Affiliation(s)
- Nalini R. Rao
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Olivia DeGulis
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Toshihiro Nomura
- Department of Neuroscience, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - SeungEun Lee
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Timothy J. Hark
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Justin C. Dynes
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Emily X. Dexter
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
| | - Junyue Ge
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
| | - Arun Upadhyay
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Eugenio F. Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Robert Vassar
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
- Department of Neurodegenerative disease, Queen Square Institute of Neurology, University College London, London, UK
| | - Anis Contractor
- Department of Neuroscience, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Jeffrey N. Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| |
Collapse
|
3
|
Barrantes FJ. Cognitive synaptopathy: synaptic and dendritic spine dysfunction in age-related cognitive disorders. Front Aging Neurosci 2024; 16:1476909. [PMID: 39420927 PMCID: PMC11484076 DOI: 10.3389/fnagi.2024.1476909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Cognitive impairment is a leading component of several neurodegenerative and neurodevelopmental diseases, profoundly impacting on the individual, the family, and society at large. Cognitive pathologies are driven by a multiplicity of factors, from genetic mutations and genetic risk factors, neurotransmitter-associated dysfunction, abnormal connectomics at the level of local neuronal circuits and broader brain networks, to environmental influences able to modulate some of the endogenous factors. Otherwise healthy older adults can be expected to experience some degree of mild cognitive impairment, some of which fall into the category of subjective cognitive deficits in clinical practice, while many neurodevelopmental and neurodegenerative diseases course with more profound alterations of cognition, particularly within the spectrum of the dementias. Our knowledge of the underlying neuropathological mechanisms at the root of this ample palette of clinical entities is far from complete. This review looks at current knowledge on synaptic modifications in the context of cognitive function along healthy ageing and cognitive dysfunction in disease, providing insight into differential diagnostic elements in the wide range of synapse alterations, from those associated with the mild cognitive changes of physiological senescence to the more profound abnormalities occurring at advanced clinical stages of dementia. I propose the term "cognitive synaptopathy" to encompass the wide spectrum of synaptic pathologies associated with higher brain function disorders.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, Pontifical Catholic University of Argentina (UCA), Argentine Scientific and Technological Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
4
|
López-Cerdán A, Andreu Z, Hidalgo MR, Soler-Sáez I, de la Iglesia-Vayá M, Mikozami A, Guerini FR, García-García F. An integrated approach to identifying sex-specific genes, transcription factors, and pathways relevant to Alzheimer's disease. Neurobiol Dis 2024; 199:106605. [PMID: 39009097 DOI: 10.1016/j.nbd.2024.106605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/06/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Age represents a significant risk factor for the development of Alzheimer's disease (AD); however, recent research has documented an influencing role of sex in several features of AD. Understanding the impact of sex on specific molecular mechanisms associated with AD remains a critical challenge to creating tailored therapeutic interventions. METHODS The exploration of the sex-based differential impact on disease (SDID) in AD used a systematic review to first select transcriptomic studies of AD with data regarding sex in the period covering 2002 to 2021 with a focus on the primary brain regions affected by AD - the cortex (CT) and the hippocampus (HP). A differential expression analysis for each study and two tissue-specific meta-analyses were then performed. Focusing on the CT due to the presence of significant SDID-related alterations, a comprehensive functional characterization was conducted: protein-protein network interaction and over-representation analyses to explore biological processes and pathways and a VIPER analysis to estimate transcription factor activity. RESULTS We selected 8 CT and 5 HP studies from the Gene Expression Omnibus (GEO) repository for tissue-specific meta-analyses. We detected 389 significantly altered genes in the SDID comparison in the CT. Generally, female AD patients displayed more affected genes than males; we grouped said genes into six subsets according to their expression profile in female and male AD patients. Only subset I (repressed genes in female AD patients) displayed significant results during functional profiling. Female AD patients demonstrated more significant impairments in biological processes related to the regulation and organization of synapsis and pathways linked to neurotransmitters (glutamate and GABA) and protein folding, Aβ aggregation, and accumulation compared to male AD patients. These findings could partly explain why we observe more pronounced cognitive decline in female AD patients. Finally, we detected 23 transcription factors with different activation patterns according to sex, with some associated with AD for the first time. All results generated during this study are readily available through an open web resource Metafun-AD (https://bioinfo.cipf.es/metafun-ad/). CONCLUSION Our meta-analyses indicate the existence of differences in AD-related mechanisms in female and male patients. These sex-based differences will represent the basis for new hypotheses and could significantly impact precision medicine and improve diagnosis and clinical outcomes in AD patients.
Collapse
Affiliation(s)
- Adolfo López-Cerdán
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain; Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012, Valencia, Spain
| | - Zoraida Andreu
- Foundation Valencian Institute of Oncology (FIVO), 46009, Valencia, Spain
| | - Marta R Hidalgo
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Irene Soler-Sáez
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - María de la Iglesia-Vayá
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012, Valencia, Spain
| | - Akiko Mikozami
- Oral Health/Brain Health/Total health (OBT) Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | | - Francisco García-García
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain.
| |
Collapse
|
5
|
Thananthirige KPM, Chitranshi N, Basavarajappa D, Rajput R, Abbasi M, Palanivel V, Gupta VB, Paulo JA, Koronyo-Hamaoui M, Mirzaei M, Graham SL, Gupta V. Tau modulation through AAV9 therapy augments Akt/Erk survival signalling in glaucoma mitigating the retinal degenerative phenotype. Acta Neuropathol Commun 2024; 12:89. [PMID: 38845058 PMCID: PMC11158005 DOI: 10.1186/s40478-024-01804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
The microtubule-associated protein Tau is a key player in various neurodegenerative conditions, including Alzheimer's disease (AD) and Tauopathies, where its hyperphosphorylation disrupts neuronal microtubular lattice stability. Glaucoma, a neurodegenerative disorder affecting the retina, leads to irreversible vision loss by damaging retinal ganglion cells and the optic nerve, often associated with increased intraocular pressure. Prior studies have indicated Tau expression and phosphorylation alterations in the retina in both AD and glaucoma, yet the causative or downstream nature of Tau protein changes in these pathologies remains unclear. This study investigates the impact of Tau protein modulation on retinal neurons under normal and experimental glaucoma conditions. Employing AAV9-mediated gene therapy for Tau overexpression and knockdown, both manipulations were found to adversely affect retinal structural and functional measures as well as neuroprotective Akt/Erk survival signalling in healthy conditions. In the experimental glaucoma model, Tau overexpression intensified inner retinal degeneration, while Tau silencing provided significant protection against these degenerative changes. These findings underscore the critical role of endogenous Tau protein levels in preserving retinal integrity and emphasize the therapeutic potential of targeting Tau in glaucoma pathology.
Collapse
Affiliation(s)
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Rashi Rajput
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Mojdeh Abbasi
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| | - Viswanthram Palanivel
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Veer Bala Gupta
- School of Medicine, Deakin University, Melbourne, VIC, 3220, Australia
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Research Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
- Division of Applied Cell Biology and Physiology, Departments of Neurology and Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Stuart L Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
6
|
L'esperance OJ, McGhee J, Davidson G, Niraula S, Smith AS, Sosunov A, Yan SS, Subramanian J. Functional connectivity favors aberrant visual network c-Fos expression accompanied by cortical synapse loss in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.05.522900. [PMID: 36712054 PMCID: PMC9881957 DOI: 10.1101/2023.01.05.522900] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, sensory network dysfunction has received comparatively less attention despite compelling evidence of its significance in both Alzheimer's disease patients and mouse models. We recently found that neurons in the primary visual cortex of an AD mouse model expressing human amyloid protein precursor with the Swedish and Indiana mutations (hAPP mutations) exhibit aberrant c-Fos expression and altered synaptic structures at a pre-amyloid plaque stage. However, it is unclear whether aberrant c-Fos expression and synaptic pathology vary across the broader visual network and to what extent c-Fos abnormality in the cortex is inherited through functional connectivity. Using both sexes of 4-6-month AD model mice with hAPP mutations (J20[PDGF-APPSw, Ind]), we found that cortical regions of the visual network show aberrant c-Fos expression and impaired experience-dependent modulation while subcortical regions do not. Interestingly, the average network-wide functional connectivity strength of a brain region in wild type (WT) mice significantly predicts its aberrant c-Fos expression, which in turn correlates with impaired experience-dependent modulation in the AD model. Using in vivo two-photon and ex vivo imaging of presynaptic termini, we observed a subtle yet selective weakening of excitatory cortical synapses in the visual cortex. Intriguingly, the change in the size distribution of cortical boutons in the AD model is downscaled relative to those in WT mice, suggesting that synaptic weakening may reflect an adaptation to aberrant activity. Our observations suggest that cellular and synaptic abnormalities in the AD model represent a maladaptive transformation of the baseline physiological state seen in WT conditions rather than entirely novel and unrelated manifestations.
Collapse
|
7
|
Cai H, Pang Y, Ren Z, Fu X, Jia L. Delivering synaptic protein mRNAs via extracellular vesicles ameliorates cognitive impairment in a mouse model of Alzheimer's disease. BMC Med 2024; 22:138. [PMID: 38528511 PMCID: PMC10964680 DOI: 10.1186/s12916-024-03359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Synaptic dysfunction with reduced synaptic protein levels is a core feature of Alzheimer's disease (AD). Synaptic proteins play a central role in memory processing, learning, and AD pathogenesis. Evidence suggests that synaptic proteins in plasma neuronal-derived extracellular vesicles (EVs) are reduced in patients with AD. However, it remains unclear whether levels of synaptic proteins in EVs are associated with hippocampal atrophy of AD and whether upregulating the expression of these synaptic proteins has a beneficial effect on AD. METHODS In this study, we included 57 patients with AD and 56 healthy controls. We evaluated their brain atrophy through magnetic resonance imaging using the medial temporal lobe atrophy score. We measured the levels of four synaptic proteins, including synaptosome-associated protein 25 (SNAP25), growth-associated protein 43 (GAP43), neurogranin, and synaptotagmin 1 in both plasma neuronal-derived EVs and cerebrospinal fluid (CSF). We further examined the association of synaptic protein levels with brain atrophy. We also evaluated the levels of these synaptic proteins in the brains of 5×FAD mice. Then, we loaded rabies virus glycoprotein-engineered EVs with messenger RNAs (mRNAs) encoding GAP43 and SNAP25 and administered these EVs to 5×FAD mice. After treatment, synaptic proteins, dendritic density, and cognitive function were evaluated. RESULTS The results showed that GAP43, SNAP25, neurogranin, and synaptotagmin 1 were decreased in neuronal-derived EVs but increased in CSF in patients with AD, and the changes corresponded to the severity of brain atrophy. GAP43 and SNAP25 were decreased in the brains of 5×FAD mice. The engineered EVs efficiently and stably delivered these synaptic proteins to the brain, where synaptic protein levels were markedly upregulated. Upregulation of synaptic protein expression could ameliorate cognitive impairment in AD by promoting dendritic density. This marks the first successful delivery of synaptic protein mRNAs via EVs in AD mice, yielding remarkable therapeutic effects. CONCLUSIONS Synaptic proteins are closely related to AD processes. Delivery of synaptic protein mRNAs via EVs stands as a promising effective precision treatment strategy for AD, which significantly advances the current understanding of therapeutic approaches for the disease.
Collapse
Affiliation(s)
- Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yana Pang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Gomez‐Arboledas A, Fonseca MI, Kramar E, Chu S, Schartz ND, Selvan P, Wood MA, Tenner AJ. C5aR1 signaling promotes region- and age-dependent synaptic pruning in models of Alzheimer's disease. Alzheimers Dement 2024; 20:2173-2190. [PMID: 38278523 PMCID: PMC10984438 DOI: 10.1002/alz.13682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Synaptic loss is a hallmark of Alzheimer's disease (AD) that correlates with cognitive decline in AD patients. Complement-mediated synaptic pruning has been associated with this excessive loss of synapses in AD. Here, we investigated the effect of C5aR1 inhibition on microglial and astroglial synaptic pruning in two mouse models of AD. METHODS A combination of super-resolution and confocal and tridimensional image reconstruction was used to assess the effect of genetic ablation or pharmacological inhibition of C5aR1 on the Arctic48 and Tg2576 models of AD. RESULTS Genetic ablation or pharmacological inhibition of C5aR1 partially rescues excessive pre-synaptic pruning and synaptic loss in an age and region-dependent fashion in two mouse models of AD, which correlates with improved long-term potentiation (LTP). DISCUSSION Reduction of excessive synaptic pruning is an additional beneficial outcome of the suppression of C5a-C5aR1 signaling, further supporting its potential as an effective targeted therapy to treat AD. HIGHLIGHTS C5aR1 ablation restores long-term potentiation in the Arctic model of AD. C5aR1 ablation rescues region specific excessive pre-synaptic loss. C5aR1 antagonist, PMX205, rescues VGlut1 loss in the Tg2576 model of AD. C1q tagging is not sufficient to induce VGlut1 microglial ingestion. Astrocytes contribute to excessive pre-synaptic loss at late stages of the disease.
Collapse
Affiliation(s)
- Angela Gomez‐Arboledas
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Maria I. Fonseca
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Enikö Kramar
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shu‐Hui Chu
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Nicole D. Schartz
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Purnika Selvan
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaSchool of MedicineIrvineCaliforniaUSA
| |
Collapse
|
9
|
Lu L, Shi Y, Wei B, Li W, Yu X, Zhao Y, Yu D, Sun M. YTHDF3 modulates the Cbln1 level by recruiting BTG2 and is implicated in the impaired cognition of prenatal hypoxia offspring. iScience 2024; 27:108703. [PMID: 38205248 PMCID: PMC10776956 DOI: 10.1016/j.isci.2023.108703] [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: 04/13/2023] [Revised: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
The "Fetal Origins of Adult Disease (FOAD)" hypothesis holds that adverse factors during pregnancy can increase the risk of chronic diseases in offspring. Here, we investigated the effects of prenatal hypoxia (PH) on brain structure and function in adult offspring and explored the role of the N6-methyladenosine (m6A) pathway. The results suggest that abnormal cognition in PH offspring may be related to the dysregulation of the m6A pathway, specifically increased levels of YTHDF3 in the hippocampus. YTHDF3 interacts with BTG2 and is involved in the decay of Cbln1 mRNA, leading to the down-regulation of Cbln1 expression. Deficiency of Cbln1 may contribute to abnormal synaptic function, which in turn causes cognitive impairment in PH offspring. This study provides a scientific clues for understanding the mechanisms of impaired cognition in PH offspring and provides a theoretical basis for the treatment of cognitive impairment in offspring exposed to PH.
Collapse
Affiliation(s)
- Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Weisheng Li
- Department of Gynaecology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic, Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
10
|
L'Esperance OJ, McGhee J, Davidson G, Niraula S, Smith AS, Sosunov AA, Yan SS, Subramanian J. Functional Connectivity Favors Aberrant Visual Network c-Fos Expression Accompanied by Cortical Synapse Loss in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 101:111-131. [PMID: 39121131 DOI: 10.3233/jad-240776] [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] [Indexed: 08/11/2024]
Abstract
Background While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, visual network dysfunction has received less attention despite compelling evidence of its significance in AD patients and mouse models. We recently reported c-Fos and synaptic dysregulation in the primary visual cortex of a pre-amyloid plaque AD-model. Objective We test whether c-Fos expression and presynaptic density/dynamics differ in cortical and subcortical visual areas in an AD-model. We also examine whether aberrant c-Fos expression is inherited through functional connectivity and shaped by light experience. Methods c-Fos+ cell density, functional connectivity, and their experience-dependent modulation were assessed for visual and whole-brain networks in both sexes of 4-6-month-old J20 (AD-model) and wildtype (WT) mice. Cortical and subcortical differences in presynaptic vulnerability in the AD-model were compared using ex vivo and in vivo imaging. Results Visual cortical, but not subcortical, networks show aberrant c-Fos expression and impaired experience-dependent modulation. The average functional connectivity of a brain region in WT mice significantly predicts aberrant c-Fos expression, which correlates with impaired experience-dependent modulation in the AD-model. We observed a subtle yet selective weakening of excitatory visual cortical synapses. The size distribution of cortical boutons in the AD-model is downscaled relative to those in WT mice, suggesting a synaptic scaling-like adaptation of bouton size. Conclusions Visual network structural and functional disruptions are biased toward cortical regions in pre-plaque J20 mice, and the cellular and synaptic dysregulation in the AD-model represents a maladaptive modification of the baseline physiology seen in WT conditions.
Collapse
Affiliation(s)
- Oliver J L'Esperance
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Joshua McGhee
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Garett Davidson
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Adam S Smith
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Alexandre A Sosunov
- Department of Neurosurgery, Columbia University Medical Center, New York, NY, USA
| | - Shirley Shidu Yan
- Department of Neurosurgery, Columbia University Medical Center, New York, NY, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
11
|
Yeo S, Jang J, Jung HJ, Lee H, Choe Y. Primary cilia-mediated regulation of microglial secretion in Alzheimer's disease. Front Mol Biosci 2023; 10:1250335. [PMID: 37942288 PMCID: PMC10627801 DOI: 10.3389/fmolb.2023.1250335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023] Open
Abstract
Alzheimer's disease (AD) is a brain disorder manifested by a gradual decline in cognitive function due to the accumulation of extracellular amyloid plaques, disruptions in neuronal substance transport, and the degeneration of neurons. In affected neurons, incomplete clearance of toxic proteins by neighboring microglia leads to irreversible brain inflammation, for which cellular signaling is poorly understood. Through single-cell transcriptomic analysis, we discovered distinct regional differences in the ability of microglia to clear damaged neurites. Specifically, microglia in the septal region of wild type mice exhibited a transcriptomic signature resembling disease-associated microglia (DAM). These lateral septum (LS)-enriched microglia were associated with dense axonal bundles originating from the hippocampus. Further transcriptomic and proteomic approaches revealed that primary cilia, small hair-like structures found on cells, played a role in the regulation of microglial secretory function. Notably, primary cilia were transiently observed in microglia, and their presence was significantly reduced in microglia from AD mice. We observed significant changes in the secretion and proteomic profiles of the secretome after inhibiting the primary cilia gene intraflagellar transport particle 88 (Ift88) in microglia. Intriguingly, inhibiting primary cilia in the septal microglia of AD mice resulted in the expansion of extracellular amyloid plaques and damage to adjacent neurites. These results indicate that DAM-like microglia are present in the LS, a critical target region for hippocampal nerve bundles, and that the primary ciliary signaling system regulates microglial secretion, affecting extracellular proteostasis. Age-related primary ciliopathy probably contributes to the selective sensitivity of microglia, thereby exacerbating AD. Targeting the primary ciliary signaling system could therefore be a viable strategy for modulating neuroimmune responses in AD treatments.
Collapse
Affiliation(s)
- Seungeun Yeo
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyeyoung Lee
- Division of Applied Bioengineering, Dong-eui University, Busan, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
12
|
Chen H, Cai J, Wang A, Su W, Ji C, Zhao L. Treadmill exercise prevents the hyperexcitability of pyramidal neurons in medial entorhinal cortex in the 3xTg-AD mouse model of Alzheimer's disease. Exp Gerontol 2023; 182:112309. [PMID: 37832802 DOI: 10.1016/j.exger.2023.112309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Neuronal hyperactivity is a key abnormality in early stage Alzheimer's disease (AD). Medial entorhinal cortex (mEC) plays a vital role in memory function and is affected early in AD. Growing evidence indicates benefits of regular exercise on memory and cognitive function in humans with AD, although, the underlying mechanisms are not clear. Therefore, this study was designed to test the effects of 16 weeks treadmill exercise on spatial learning memory and the underlying cellular mechanisms in 6-month-old 3xTg-AD mice. Whole-cell patch clamp was used to examine neuronal intrinsic excitability, spontaneous excitatory postsynaptic currents (sEPSCs) and spontaneous inhibitory postsynaptic currents (sIPSCs) of mEC layer II/III pyramidal neurons in the following groups: wild type (WT + sham), 3xTg-AD (AD+sham), WT receiving exercise (WT + Ex), and AD receiving exercise (AD+Ex). We found that at a behavioral level, treadmill exercise decreased working memory errors in radial arm maze (RAM) test in 6-month-old AD mice. At a cellular level, we found that treadmill exercise prevented the abnormal increase in mEC pyramidal neuron input resistance and action potential firing in 6-month-old 3xTg-AD mice compared with WT + sham and AD+Ex mice; further, sEPSC amplitude and frequency were normal in AD+Ex but overactive in AD+sham; additionally, GABAergic inhibition was normal in AD+Ex mice but reduced in AD+sham. In conclusion, our results indicate that treadmill exercise improves spatial learning memory and prevents network hyperexcitability in mEC by reducing pyramidal neuronal intrinsic excitability and normalizing excitatory and inhibitory synaptic transmission in 3xTg-AD mice.
Collapse
Affiliation(s)
- Huimin Chen
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Aozhe Wang
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Wantang Su
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| | - Chunyan Ji
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China.
| |
Collapse
|
13
|
Gomez-Arboledas A, Fonseca MI, Kramar E, Chu SH, Schartz N, Selvan P, Wood MA, Tenner AJ. C5aR1 signaling promotes region and age dependent synaptic pruning in models of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560234. [PMID: 37873302 PMCID: PMC10592845 DOI: 10.1101/2023.09.29.560234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Synaptic loss is a hallmark of Alzheimer's disease (AD) that correlates with cognitive decline in AD patients. Complement-mediated synaptic pruning has been associated with this excessive loss of synapses in AD. Here, we investigated the effect of C5aR1 inhibition on microglial and astroglial synaptic pruning in two mouse models of AD. METHODS A combination of super-resolution and confocal and tridimensional image reconstruction was used to assess the effect of genetic ablation or pharmacological inhibition of C5aR1 on the Arctic48 and Tg2576 models of AD. RESULTS Genetic ablation or pharmacological inhibition of C5aR1 rescues the excessive pre-synaptic pruning and synaptic loss in an age and region dependent fashion in two mouse models of AD, which correlates with improved long-term potentiation (LTP). DISCUSSION Reduction of excessive synaptic pruning is an additional beneficial outcome of the suppression of C5a-C5aR1 signaling, further supporting its potential as an effective targeted therapy to treat AD.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Maria I. Fonseca
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Enikö Kramar
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Shu-Hui Chu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Nicole Schartz
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Purnika Selvan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| |
Collapse
|
14
|
Dunot J, Ribera A, Pousinha PA, Marie H. Spatiotemporal insights of APP function. Curr Opin Neurobiol 2023; 82:102754. [PMID: 37542943 DOI: 10.1016/j.conb.2023.102754] [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: 04/17/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/07/2023]
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous protein with a strong genetic link to Alzheimer's disease. Although the protein was identified more than forty years ago, its physiological function is still unclear. In recent years, advances in technology have allowed researchers to tackle APP functions in greater depth. In this review, we discuss the latest research pertaining to APP functions from development to aging. We also address the different roles that APP could play in specific types of cells of the central and peripheral nervous system and in other organs of the body. We argue that, until we fully identify the functions of APP in space and time, we will be missing important pieces of the puzzle to solve its pathological implication in Alzheimer's disease and beyond.
Collapse
Affiliation(s)
- Jade Dunot
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France. https://twitter.com/DunotJade
| | - Aurore Ribera
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France. https://twitter.com/aurore_et_al_
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France.
| | - Hélène Marie
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France.
| |
Collapse
|
15
|
Čarna M, Onyango IG, Katina S, Holub D, Novotny JS, Nezvedova M, Jha D, Nedelska Z, Lacovich V, Vyvere TV, Houbrechts R, Garcia-Mansfield K, Sharma R, David-Dirgo V, Vyhnalek M, Texlova K, Chaves H, Bakkar N, Pertierra L, Vinkler M, Markova H, Laczo J, Sheardova K, Hortova-Kohoutkova M, Frič J, Forte G, Kaňovsky P, Belaškova S, Damborsky J, Hort J, Seyfried NT, Bowser R, Sevlever G, Rissman RA, Smith RA, Hajduch M, Pirrotte P, Spačil Z, Dammer EB, Limbäck-Stokin C, Stokin GB. Pathogenesis of Alzheimer's disease: Involvement of the choroid plexus. Alzheimers Dement 2023; 19:3537-3554. [PMID: 36825691 PMCID: PMC10634590 DOI: 10.1002/alz.12970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/30/2022] [Accepted: 12/19/2022] [Indexed: 02/25/2023]
Abstract
The choroid plexus (ChP) produces and is bathed in the cerebrospinal fluid (CSF), which in aging and Alzheimer's disease (AD) shows extensive proteomic alterations including evidence of inflammation. Considering inflammation hampers functions of the involved tissues, the CSF abnormalities reported in these conditions are suggestive of ChP injury. Indeed, several studies document ChP damage in aging and AD, which nevertheless remains to be systematically characterized. We here report that the changes elicited in the CSF by AD are consistent with a perturbed aging process and accompanied by aberrant accumulation of inflammatory signals and metabolically active proteins in the ChP. Magnetic resonance imaging (MRI) imaging shows that these molecular aberrancies correspond to significant remodeling of ChP in AD, which correlates with aging and cognitive decline. Collectively, our preliminary post-mortem and in vivo findings reveal a repertoire of ChP pathologies indicative of its dysfunction and involvement in the pathogenesis of AD. HIGHLIGHTS: Cerebrospinal fluid changes associated with aging are perturbed in Alzheimer's disease Paradoxically, in Alzheimer's disease, the choroid plexus exhibits increased cytokine levels without evidence of inflammatory activation or infiltrates In Alzheimer's disease, increased choroid plexus volumes correlate with age and cognitive performance.
Collapse
Affiliation(s)
- Maria Čarna
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | - Isaac G. Onyango
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | - Stanislav Katina
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Institute of Mathematics and Statistics, Masaryk University, Brno, Czech Republic
| | - Dušan Holub
- Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jan Sebastian Novotny
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | - Marketa Nezvedova
- RECETOX Centre, Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - Durga Jha
- RECETOX Centre, Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - Zuzana Nedelska
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Memory Clinic, Department of Neurology, 2 Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Valentina Lacovich
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | | | | | - Krystine Garcia-Mansfield
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Ritin Sharma
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Victoria David-Dirgo
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Martin Vyhnalek
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Memory Clinic, Department of Neurology, 2 Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Kateřina Texlova
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | | | - Nadine Bakkar
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Mojmir Vinkler
- Institute of Mathematics and Statistics, Masaryk University, Brno, Czech Republic
| | - Hana Markova
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Memory Clinic, Department of Neurology, 2 Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jan Laczo
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Memory Clinic, Department of Neurology, 2 Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Kateřina Sheardova
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- 1 Department of Neurology, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | - Jan Frič
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Giancarlo Forte
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | - Petr Kaňovsky
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University Olomouc and Research and Science Department, University Hospital Olomouc, Olomouc, Czech Republic
| | - Silvie Belaškova
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
| | - Jiři Damborsky
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- RECETOX Centre, Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - Jakub Hort
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Memory Clinic, Department of Neurology, 2 Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Nicholas T. Seyfried
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA, USA
- Departments of Biochemistry and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert Bowser
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Robert A. Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | - Marian Hajduch
- Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Mass Spectrometry & Proteomics Core Facility, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zdeněk Spačil
- RECETOX Centre, Faculty of Sciences, Masaryk University, Brno, Czech Republic
| | - Eric B. Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA, USA
| | - Clara Limbäck-Stokin
- Department of Cellular Pathology, Imperial College Healthcare NHS Trust, London, UK
- Imperial College London, Faculty of Medicine, London, UK
| | - Gorazd B. Stokin
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Division of Neurology, University Medical Centre, Ljubljana, Slovenia
- Translational Aging and Neuroscience Program, Mayo Clinic, MN, Rochester, USA
| |
Collapse
|
16
|
Lenz M, Eichler A, Kruse P, Galanis C, Kleidonas D, Andrieux G, Boerries M, Jedlicka P, Müller U, Deller T, Vlachos A. The Amyloid Precursor Protein Regulates Synaptic Transmission at Medial Perforant Path Synapses. J Neurosci 2023; 43:5290-5304. [PMID: 37369586 PMCID: PMC10359033 DOI: 10.1523/jneurosci.1824-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The perforant path provides the primary cortical excitatory input to the hippocampus. Because of its important role in information processing and coding, entorhinal projections to the dentate gyrus have been studied in considerable detail. Nevertheless, synaptic transmission between individual connected pairs of entorhinal stellate cells and dentate granule cells remains to be characterized. Here, we have used mouse organotypic entorhino-hippocampal tissue cultures of either sex, in which the entorhinal cortex (EC) to dentate granule cell (GC; EC-GC) projection is present, and EC-GC pairs can be studied using whole-cell patch-clamp recordings. By using cultures of wild-type mice, the properties of EC-GC synapses formed by afferents from the lateral and medial entorhinal cortex were compared, and differences in short-term plasticity were identified. As the perforant path is severely affected in Alzheimer's disease, we used tissue cultures of amyloid precursor protein (APP)-deficient mice to examine the role of APP at this synapse. APP deficiency altered excitatory neurotransmission at medial perforant path synapses, which was accompanied by transcriptomic and ultrastructural changes. Moreover, presynaptic but not postsynaptic APP deletion through the local injection of Cre-expressing adeno-associated viruses in conditional APPflox/flox tissue cultures increased the neurotransmission efficacy at perforant path synapses. In summary, these data suggest a physiological role for presynaptic APP at medial perforant path synapses that may be adversely affected under altered APP processing conditions.SIGNIFICANCE STATEMENT The hippocampus receives input from the entorhinal cortex via the perforant path. These projections to hippocampal dentate granule cells are of utmost importance for learning and memory formation. Although there is detailed knowledge about perforant path projections, the functional synaptic properties at the level of individual connected pairs of neurons are not well understood. In this study, we investigated the role of APP in mediating functional properties and transmission rules in individually connected neurons using paired whole-cell patch-clamp recordings and genetic tools in organotypic tissue cultures. Our results show that presynaptic APP expression limits excitatory neurotransmission via the perforant path, which could be compromised in pathologic conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, 30625 Hannover, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Peter Jedlicka
- Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
| | - Ulrike Müller
- Institute of Pharmacy and Molecular Biotechnology, Functional Genomics, Ruprecht-Karls University Heidelberg, 69120 Heidelberg, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
17
|
Lumeij LB, van Huijstee AN, Cappaert NLM, Kessels HW. Variance analysis as a method to predict the locus of plasticity at populations of non-uniform synapses. Front Cell Neurosci 2023; 17:1232541. [PMID: 37528963 PMCID: PMC10388551 DOI: 10.3389/fncel.2023.1232541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Our knowledge on synaptic transmission in the central nervous system has often been obtained by evoking synaptic responses to populations of synapses. Analysis of the variance in synaptic responses can be applied as a method to predict whether a change in synaptic responses is a consequence of altered presynaptic neurotransmitter release or postsynaptic receptors. However, variance analysis is based on binomial statistics, which assumes that synapses are uniform. In reality, synapses are far from uniform, which questions the reliability of variance analysis when applying this method to populations of synapses. To address this, we used an in silico model for evoked synaptic responses and compared variance analysis outcomes between populations of uniform versus non-uniform synapses. This simulation revealed that variance analysis produces similar results irrespectively of the grade of uniformity of synapses. We put this variance analysis to the test with an electrophysiology experiment using a model system for which the loci of plasticity are well established: the effect of amyloid-β on synapses. Variance analysis correctly predicted that postsynaptically produced amyloid-β triggered predominantly a loss of synapses and a minor reduction of postsynaptic currents in remaining synapses with little effect on presynaptic release probability. We propose that variance analysis can be reliably used to predict the locus of synaptic changes for populations of non-uniform synapses.
Collapse
|
18
|
Tousley AR, Yeh PWL, Yeh HH. Precocious emergence of cognitive and synaptic dysfunction in 3xTg-AD mice exposed prenatally to ethanol. Alcohol 2023; 107:56-72. [PMID: 36038084 PMCID: PMC10183974 DOI: 10.1016/j.alcohol.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting approximately 50 million people worldwide. Early life risk factors for AD, including prenatal exposures, remain underexplored. Exposure of the fetus to alcohol (ethanol) is not uncommon during pregnancy, and may result in physical, behavioral, and cognitive changes that are first detected during childhood but result in lifelong challenges. Whether or not prenatal ethanol exposure may contribute to Alzheimer's disease risk is not yet known. Here we exposed a mouse model of Alzheimer's disease (3xTg-AD), bearing three dementia-associated transgenes, presenilin1 (PS1M146V), human amyloid precursor protein (APPSwe), and human tau (TauP301S), to ethanol on gestational days 13.5-16.5 using an established binge-type maternal ethanol exposure paradigm. We sought to investigate whether prenatal ethanol exposure resulted in a precocious onset or increased severity of AD progression, or both. We found that a brief binge-type gestational exposure to ethanol during a period of peak neuronal migration to the developing cortex resulted in an earlier onset of spatial memory deficits and behavioral inflexibility in the progeny, as assessed by performance on the modified Barnes maze task. The observed cognitive changes coincided with alterations to both GABAergic and glutamatergic synaptic transmission in layer V/VI neurons, diminished GABAergic interneurons, and increased β-amyloid accumulation in the medial prefrontal cortex. These findings provide the first preclinical evidence for prenatal ethanol exposure as a potential factor for modifying the onset of AD-like behavioral dysfunction and set the groundwork for more comprehensive investigations into the underpinnings of AD-like cognitive changes in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Adelaide R Tousley
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; MD-PhD Program, Geisel School of Medicine at Dartmouth; Integrative Neuroscience at Dartmouth Graduate Program, Hanover, NH, United States
| | - Pamela W L Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Hermes H Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| |
Collapse
|
19
|
Tian C, Stewart T, Hong Z, Guo Z, Aro P, Soltys D, Pan C, Peskind ER, Zabetian CP, Shaw LM, Galasko D, Quinn JF, Shi M, Zhang J. Blood extracellular vesicles carrying synaptic function- and brain-related proteins as potential biomarkers for Alzheimer's disease. Alzheimers Dement 2023; 19:909-923. [PMID: 35779041 PMCID: PMC9806186 DOI: 10.1002/alz.12723] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Objective and accessible markers for Alzheimer's disease (AD) and other dementias are critically needed. METHODS We identified NMDAR2A, a protein related to synaptic function, as a novel marker of central nervous system (CNS)-derived plasma extracellular vesicles (EVs) and developed a flow cytometry-based technology for detecting such plasma EVs readily. The assay was initially tested in our local cross-sectional study to distinguish AD patients from healthy controls (HCs) or from Parkinson's disease (PD) patients, followed by a validation study using an independent cohort collected from multiple medical centers (the Alzheimer's Disease Neuroimaging Initiative). Cerebrospinal fluid AD molecular signature was used to confirm diagnoses of all AD participants. RESULTS Likely CNS-derived EVs in plasma were significantly reduced in AD compared to HCs in both cohorts. Integrative models including CNS-derived EV markers and AD markers present on EVs reached area under the curve of 0.915 in discovery cohort and 0.810 in validation cohort. DISCUSSION This study demonstrated that robust and rapid analysis of individual neuron-derived synaptic function-related EVs in peripheral blood may serve as a helpful marker of synaptic dysfunction in AD and dementia.
Collapse
Affiliation(s)
- Chen Tian
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Tessandra Stewart
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Zhen Hong
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Guo
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Patrick Aro
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - David Soltys
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Catherine Pan
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elaine R Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Northwest (VISN-20) Mental Illness, Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Cyrus P. Zabetian
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Leslie M. Shaw
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Douglas Galasko
- Department of Neurology, University of California, San Diego, California, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology and Parkinson’s Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, USA
| | - Min Shi
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jing Zhang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
20
|
Rem PD, Sereikaite V, Fernández-Fernández D, Reinartz S, Ulrich D, Fritzius T, Trovo L, Roux S, Chen Z, Rondard P, Pin JP, Schwenk J, Fakler B, Gassmann M, Barkat TR, Strømgaard K, Bettler B. Soluble amyloid-β precursor peptide does not regulate GABA B receptor activity. eLife 2023; 12:82082. [PMID: 36688536 PMCID: PMC9917443 DOI: 10.7554/elife.82082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/22/2023] [Indexed: 01/24/2023] Open
Abstract
Amyloid-β precursor protein (APP) regulates neuronal activity through the release of secreted APP (sAPP) acting at cell surface receptors. APP and sAPP were reported to bind to the extracellular sushi domain 1 (SD1) of GABAB receptors (GBRs). A 17 amino acid peptide (APP17) derived from APP was sufficient for SD1 binding and shown to mimic the inhibitory effect of sAPP on neurotransmitter release and neuronal activity. The functional effects of APP17 and sAPP were similar to those of the GBR agonist baclofen and blocked by a GBR antagonist. These experiments led to the proposal that sAPP activates GBRs to exert its neuronal effects. However, whether APP17 and sAPP influence classical GBR signaling pathways in heterologous cells was not analyzed. Here, we confirm that APP17 binds to GBRs with nanomolar affinity. However, biochemical and electrophysiological experiments indicate that APP17 does not influence GBR activity in heterologous cells. Moreover, APP17 did not regulate synaptic GBR localization, GBR-activated K+ currents, neurotransmitter release, or neuronal activity in vitro or in vivo. Our results show that APP17 is not a functional GBR ligand and indicate that sAPP exerts its neuronal effects through receptors other than GBRs.
Collapse
Affiliation(s)
- Pascal Dominic Rem
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Vita Sereikaite
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | | | - Sebastian Reinartz
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Daniel Ulrich
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Thorsten Fritzius
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Luca Trovo
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Salomé Roux
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Ziyang Chen
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of FreiburgFreiburgGermany
- CIBSS Center for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulationFreiburgGermany
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | | | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| |
Collapse
|
21
|
Effects of Chronic Caffeine Consumption on Synaptic Function, Metabolism and Adenosine Modulation in Different Brain Areas. Biomolecules 2023; 13:biom13010106. [PMID: 36671491 PMCID: PMC9855869 DOI: 10.3390/biom13010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Adenosine receptors mainly control synaptic function, and excessive activation of adenosine receptors may worsen the onset of many neurological disorders. Accordingly, the regular intake of moderate doses of caffeine antagonizes adenosine receptors and affords robust neuroprotection. Although caffeine intake alters brain functional connectivity and multi-omics analyses indicate that caffeine intake modifies synaptic and metabolic processes, it is unclear how caffeine intake affects behavior, synaptic plasticity and its modulation by adenosine. We now report that male mice drinking caffeinated water (0.3 g/L) for 2 weeks were behaviorally indistinguishable (locomotion, mood, memory) from control mice (drinking water) and displayed superimposable synaptic plasticity (long-term potentiation) in different brain areas (hippocampus, prefrontal cortex, amygdala). Moreover, there was a general preservation of the efficiency of adenosine A1 and A2A receptors to control synaptic transmission and plasticity, although there was a tendency for lower levels of endogenous adenosine ensuring A1 receptor-mediated inhibition. In spite of similar behavioral and neurophysiological function, caffeine intake increased the energy charge and redox state of cortical synaptosomes. This increased metabolic competence likely involved a putative increase in the glycolytic rate in synapses and a prospective greater astrocyte-synapse lactate shuttling. It was concluded that caffeine intake does not trigger evident alterations of behavior or of synaptic plasticity but increases the metabolic competence of synapses, which might be related with the previously described better ability of animals consuming caffeine to cope with deleterious stimuli triggering brain dysfunction.
Collapse
|
22
|
Li Y, Wang JZ, Deng YM, Wang K, Yang L, Long C. Amyloid-β Protein Precursor Regulates Electrophysiological Properties in the Hippocampus via Altered Kv1.4 Expression and Function in Mice. J Alzheimers Dis 2023; 92:1241-1256. [PMID: 36872774 DOI: 10.3233/jad-220606] [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] [Indexed: 03/06/2023]
Abstract
BACKGROUND Amyloid-β protein precursor (AβPP) is enriched in neurons. However, the mechanism underlying AβPP regulation of neuronal activity is poorly understood. Potassium channels are critically involved in neuronal excitability. In hippocampus, A-type potassium channels are highly expressed and involved in determining neuronal spiking. OBJECTIVE We explored hippocampal local field potential (LFP) and spiking in the presence and absence of AβPP, and the potential involvement of an A-type potassium channel. METHODS We used in vivo extracellular recording and whole-cell patch-clamp recording to determine neuronal activity, current density of A-type potassium currents, and western blot to detect changes in related protein levels. RESULTS Abnormal LFP was observed in AβPP-/- mice, including reduced beta and gamma power, and increased epsilon and ripple power. The firing rate of glutamatergic neurons reduced significantly, in line with an increased action potential rheobase. Given that A-type potassium channels regulate neuronal firing, we measured the protein levels and function of two major A-type potassium channels and found that the post-transcriptional level of Kv1.4, but not Kv4.2, was significantly increased in the AβPP-/- mice. This resulted in a marked increase in the peak time of A-type transient outward potassium currents in both glutamatergic and gamma-aminobutyric acid-ergic (GABAergic) neurons. Furthermore, a mechanistic experiment using human embryonic kidney 293 (HEK293) cells revealed that the AβPP deficiency-induced increase in Kv1.4 may not involve protein-protein interaction between AβPP and Kv1.4. CONCLUSION This study suggests that AβPP modulates neuronal firing and oscillatory activity in the hippocampus, and Kv1.4 may be involved in mediating the modulation.
Collapse
Affiliation(s)
- Yi Li
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jin-Zhao Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yue-Ming Deng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Kun Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cheng Long
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
23
|
Li Q, Wang X, Wang ZH, Lin Z, Yang J, Chen J, Wang R, Ye W, Li Y, Wu Y, Xuan A. Changes in dendritic complexity and spine morphology following BCG immunization in APP/PS1 mice. Hum Vaccin Immunother 2022; 18:2121568. [PMID: 36113067 DOI: 10.1080/21645515.2022.2121568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bacillus Calmette - Guerin (BCG) is an immune regulator that can enhance hippocampal synaptic plasticity in rats; however, it is unclear whether it can improve synaptic function in a mouse model with Alzheimer's disease (AD). We hypothesized that BCG plays a protective role in AD mice and investigated its effect on dendritic morphology. The results obtained show that BCG immunization significantly increases dendritic complexity, as indicated by the increased number of dendritic intersections and branch points, as well as the increase in the fractal dimension. Furthermore, the number of primary neurites and dendritic length also increased following BCG immunization, which increased the number of spines and promoted maturation. IFN-γ and IL-4 levels increased, while TNF-α levels decreased following BCG immunization; expression levels of p-JAK2, P-STAT3, SYN, and PSD-95 also increased. Therefore, this study demonstrates that BCG immunization in APP/PS1 mice mitigated hippocampal dendritic spine pathology, especially after the third round of immunization. This effect could possibly be attributed to; changes in dendritic arborization and spine morphology or increases in SYN and PSD-95 expression levels. It could also be related to mechanisms of BCG-induced increases in IFN-γ or IL-4/JAK2/STAT3 levels.
Collapse
Affiliation(s)
| | | | | | - Zhenzong Lin
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Jieyi Yang
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Jichun Chen
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Rui Wang
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Wenfeng Ye
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Ya Li
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Yingying Wu
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| | - Aiguo Xuan
- Department of Anatomy and Neurobiology, Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
24
|
Jordà‐Siquier T, Petrel M, Kouskoff V, Smailovic U, Cordelières F, Frykman S, Müller U, Mulle C, Barthet G. APP accumulates with presynaptic proteins around amyloid plaques: A role for presynaptic mechanisms in Alzheimer's disease? Alzheimers Dement 2022; 18:2099-2116. [PMID: 35076178 PMCID: PMC9786597 DOI: 10.1002/alz.12546] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/21/2021] [Accepted: 10/25/2021] [Indexed: 01/31/2023]
Abstract
In Alzheimer's disease (AD), the distribution of the amyloid precursor protein (APP) and its fragments other than amyloid beta, has not been fully characterized. Here, we investigate the distribution of APP and its fragments in human AD brain samples and in mouse models of AD in reference to its proteases, synaptic proteins, and histopathological features characteristic of the AD brain, by combining an extensive set of histological and analytical tools. We report that the prominent somatic distribution of APP observed in control patients remarkably vanishes in human AD patients to the benefit of dense accumulations of extra-somatic APP, which surround dense-core amyloid plaques enriched in APP-Nter. These features are accentuated in patients with familial forms of the disease. Importantly, APP accumulations are enriched in phosphorylated tau and presynaptic proteins whereas they are depleted of post-synaptic proteins suggesting that the extra-somatic accumulations of APP are of presynaptic origin. Ultrastructural analyses unveil that APP concentrates in autophagosomes and in multivesicular bodies together with presynaptic vesicle proteins. Altogether, alteration of APP distribution and its accumulation together with presynaptic proteins around dense-core amyloid plaques is a key histopathological feature in AD, lending support to the notion that presynaptic failure is a strong physiopathological component of AD.
Collapse
Affiliation(s)
- Tomàs Jordà‐Siquier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Melina Petrel
- University Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4BordeauxFrance
| | - Vladimir Kouskoff
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Una Smailovic
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Fabrice Cordelières
- University Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4BordeauxFrance
| | - Susanne Frykman
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Ulrike Müller
- Institute for Pharmacy and Molecular BiotechnologyHeidelbergGermany
| | - Christophe Mulle
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Gaël Barthet
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| |
Collapse
|
25
|
Zhao DP, Lei X, Wang YY, Xue A, Zhao CY, Xu YM, Zhang Y, Liu GL, Geng F, Xu HD, Zhang N. Sagacious confucius’ pillow elixir ameliorates Dgalactose induced cognitive injury in mice via estrogenic effects and synaptic plasticity. Front Pharmacol 2022; 13:971385. [PMID: 36249769 PMCID: PMC9555387 DOI: 10.3389/fphar.2022.971385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a growing concern in modern society, and there is currently a lack of effective therapeutic drugs. Sagacious Confucius’ Pillow Elixir (SCPE) has been studied for the treatment of neurodegenerative diseases such as AD. This study aimed to reveal the key components and mechanisms of SCPE’s anti-AD effect by combining Ultra-high Performance Liquid Chromatography-electrostatic field Orbitrap combined high-resolution Mass Spectrometry (UPLC-LTQ/Orbitrap-MS) with a network pharmacology approach. And the mechanism was verified by in vivo experiments. Based on UPLC-LTQ/Orbitrap-MS technique identified 9 blood components from rat serum containing SCPE, corresponding to 113 anti-AD targets, and 15 of the 113 targets had high connectivity. KEGG pathway enrichment analysis showed that estrogen signaling pathway and synaptic signaling pathway were the most significantly enriched pathways in SCPE anti-AD, which has been proved by in vivo experiments. SCPE can exert estrogenic effects in the brain by increasing the amount of estrogen in the brain and the expression of ERα receptors. SCPE can enhance the synaptic structure plasticity by promoting the release of brain-derived neurotrophic factor (BDNF) secretion and improving actin polymerization and coordinates cofilin activity. In addition, SCPE also enhances synaptic functional plasticity by increasing the density of postsynaptic densified 95 (PSD95) proteins and the expression of functional receptor AMPA. SCPE is effective for treatment of AD and the mechanism is related to increasing estrogenic effects and improving synaptic plasticity. Our study revealed the synergistic effect of SCPE at the system level and showed that SCPE exhibits anti-AD effects in a multi-component, multi-target and multi-pathway manner. All these provide experimental support for the clinical application and drug development of SCPE in the prevention and treatment of AD.
Collapse
Affiliation(s)
- De-Ping Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xia Lei
- Institute of Traditional Chinese Medicine, Wuxi Traditional Chinese Medicine Hospital, Jiangsu, Wuxi, China
| | - Yue-Ying Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ao Xue
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chen-Yu Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yan-Ming Xu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang, China
| | - Yue Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Guo-Liang Liu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang, China
| | - Fang Geng
- Key Laboratory of Photochemistry Biomaterials and Energy Storage Materials of Heilongjiang Province, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang, China
- *Correspondence: Fang Geng, ; Hong-Dan Xu, ; Ning Zhang,
| | - Hong-Dan Xu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang, China
- College of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi, Jiangsu, China
- *Correspondence: Fang Geng, ; Hong-Dan Xu, ; Ning Zhang,
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang, China
- *Correspondence: Fang Geng, ; Hong-Dan Xu, ; Ning Zhang,
| |
Collapse
|
26
|
Chen X, He JL, Liu XT, Zhao N, Geng F, Zhu MM, Liu GP, Ren QG. DI-3-n-butylphthalide mitigates stress-induced cognitive deficits in mice through inhibition of NLRP3-Mediated neuroinflammation. Neurobiol Stress 2022; 20:100486. [PMID: 36160816 PMCID: PMC9489537 DOI: 10.1016/j.ynstr.2022.100486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022] Open
Abstract
Our previous study has demonstrated that chronic stress could cause cognitive deficits and tau pathology. However, the underlying mechanism and whether/how DI-3-n-Butylphthalide (NBP) ameliorates these effects are still unclear. Here, Wild-type mice were subjected to chronic unpredictable and mild stress (CUMS) for 8 weeks. Following the initial 4 weeks, the stressed animals were separated into susceptible (depressive) and unsusceptible (resilient) groups based on behavioral tests. Then, NBP (30 mg/kg i.g) was administered for 4 weeks. Morris water maze (MWM), Western-blot, Golgi staining, immunofluorescence staining and ELISA were used to examine behavioral, biochemical, and pathological changes. The results showed that both depressive and resilient mice displayed spatial memory deficits and an accumulation of tau in the hippocampus. Activated microglia and NLRP3 inflammasome were found after 8-week chronic stress. We also found a decreased level of postsynaptic density (PSD) related proteins (PSD93 and PSD95) and decreased the number of dendritic spines in the hippocampus. Interestingly, almost all the pathological changes in depressive and resilient mice previously mentioned could be reversed by NBP treatment. To further investigate the role of NLRP3 inflammasome in chronic stress-induced cognitive deficits, NLRP3 KO mice were also exposed to chronic stress. And these changes induced by chronic stress could not be found in NLRP3 KO mice. These results show an important role for the NLRP3/caspase-1/IL-1β axis in chronic stress-induced cognitive deficits and NBP meliorates cognitive impairments and selectively attenuates phosphorylated tau accumulation in stressed mice through regulation of NLRP3 inflammatory signaling pathway.
Collapse
Affiliation(s)
- Xiu Chen
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Juan-Ling He
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xue-Ting Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Na Zhao
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Fan Geng
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Meng-Meng Zhu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Gong-Ping Liu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Co-innovation Center of Neurodegeneration, Nantong University, Nantong, 226019, China
| | - Qing-Guo Ren
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, 210009, China
| |
Collapse
|
27
|
Pelucchi S, Gardoni F, Di Luca M, Marcello E. Synaptic dysfunction in early phases of Alzheimer's Disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:417-438. [PMID: 35034752 DOI: 10.1016/b978-0-12-819410-2.00022-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The synapse is the locus of plasticity where short-term alterations in synaptic strength are converted to long-lasting memories. In addition to the presynaptic terminal and the postsynaptic compartment, a more holistic view of the synapse includes the astrocytes and the extracellular matrix to form a tetrapartite synapse. All these four elements contribute to synapse health and are crucial for synaptic plasticity events and, thereby, for learning and memory processes. Synaptic dysfunction is a common pathogenic trait of several brain disorders. In Alzheimer's Disease, the degeneration of synapses can be detected at the early stages of pathology progression before neuronal degeneration, supporting the hypothesis that synaptic failure is a major determinant of the disease. The synapse is the place where amyloid-β peptides are generated and is the target of the toxic amyloid-β oligomers. All the elements constituting the tetrapartite synapse are altered in Alzheimer's Disease and can synergistically contribute to synaptic dysfunction. Moreover, the two main hallmarks of Alzheimer's Disease, i.e., amyloid-β and tau, act in concert to cause synaptic deficits. Deciphering the mechanisms underlying synaptic dysfunction is relevant for the development of the next-generation therapeutic strategies aimed at modifying the disease progression.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
28
|
Chen C, Lu J, Peng W, Mak MS, Yang Y, Zhu Z, Wang S, Hou J, Zhou X, Xin W, Hu Y, Tsim KWK, Han Y, Liu Q, Pi R. Acrolein, an endogenous aldehyde induces Alzheimer's disease-like pathologies in mice: A new sporadic AD animal model. Pharmacol Res 2022; 175:106003. [PMID: 34838693 DOI: 10.1016/j.phrs.2021.106003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that mainly affects elderly people. However, the translational research of AD is frustrating, suggesting that the development of new AD animal models is crucial. By gavage administration of acrolein, we constructed a simple sporadic AD animal model which showed classic pathologies of AD in 1 month. The AD-like phenotypes and pathological changes were as followed. 1) olfactory dysfunctions, cognitive impairments and psychological symptoms in C57BL/6 mice; 2) increased levels of Aβ1-42 and Tau phosphorylation (S396/T231) in cortex and hippocampus; 3) astrocytes and microglia proliferation; 4) reduced levels of postsynaptic density 95(PSD95) and Synapsin1, as well as the density of dendritic spines in the CA1 and DG neurons of the hippocampus; 5) high-frequency stimulation failed to induce the long-term potentiation (LTP) in the hippocampus after exposure to acrolein for 4 weeks; 6) decreased blood oxygen level-dependent (BOLD) signal in the olfactory bulb and induced high T2 signals in the hippocampus, which matched to the clinical observation in the brain of AD patients, and 7) activated RhoA/ROCK2/ p-cofilin-associated pathway in hippocampus of acrolein-treated mice, which may be the causes of synaptic damage and neuroinflammation in acrolein mice model. Taken together, the acrolein-induced sporadic AD mouse model closely reflects the pathological features of AD, which will be useful for the research on the mechanism of AD onset and the development of anti-AD drugs.
Collapse
Affiliation(s)
- Chen Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Junfeng Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Weijia Peng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Marvin Sh Mak
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yang Yang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zeyu Zhu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shuyi Wang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jiawei Hou
- Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xin Zhou
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenjun Xin
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510014, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| | - Qinyu Liu
- The seventh affiliated hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangzhou, Guangzhou 510006, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
29
|
Super-resolution microscopy: a closer look at synaptic dysfunction in Alzheimer disease. Nat Rev Neurosci 2021; 22:723-740. [PMID: 34725519 DOI: 10.1038/s41583-021-00531-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 11/08/2022]
Abstract
The synapse has emerged as a critical neuronal structure in the degenerative process of Alzheimer disease (AD), in which the pathogenic signals of two key players - amyloid-β (Aβ) and tau - converge, thereby causing synaptic dysfunction and cognitive deficits. The synapse presents a dynamic, confined microenvironment in which to explore how key molecules travel, localize, interact and assume different levels of organizational complexity, thereby affecting neuronal function. However, owing to their small size and the diffraction-limited resolution of conventional light microscopic approaches, investigating synaptic structure and dynamics has been challenging. Super-resolution microscopy (SRM) techniques have overcome the resolution barrier and are revolutionizing our quantitative understanding of biological systems in unprecedented spatio-temporal detail. Here we review critical new insights provided by SRM into the molecular architecture and dynamic organization of the synapse and, in particular, the interactions between Aβ and tau in this compartment. We further highlight how SRM can transform our understanding of the molecular pathological mechanisms that underlie AD. The application of SRM for understanding the roles of synapses in AD pathology will provide a stepping stone towards a broader understanding of dysfunction in other subcellular compartments and at cellular and circuit levels in this disease.
Collapse
|
30
|
Sánchez-Hidalgo AC, Arias-Aragón F, Romero-Barragán MT, Martín-Cuevas C, Delgado-García JM, Martinez-Mir A, Scholl FG. Selective expression of the neurexin substrate for presenilin in the adult forebrain causes deficits in associative memory and presynaptic plasticity. Exp Neurol 2021; 347:113896. [PMID: 34662541 DOI: 10.1016/j.expneurol.2021.113896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/27/2021] [Accepted: 10/10/2021] [Indexed: 01/25/2023]
Abstract
Presenilins (PS) form the active subunit of the gamma-secretase complex, which mediates the proteolytic clearance of a broad variety of type-I plasma membrane proteins. Loss-of-function mutations in PSEN1/2 genes are the leading cause of familial Alzheimer's disease (fAD). However, the PS/gamma-secretase substrates relevant for the neuronal deficits associated with a loss of PS function are not completely known. The members of the neurexin (Nrxn) family of presynaptic plasma membrane proteins are candidates to mediate aspects of the synaptic and memory deficits associated with a loss of PS function. Previous work has shown that fAD-linked PS mutants or inactivation of PS by genetic and pharmacological approaches failed to clear Nrxn C-terminal fragments (NrxnCTF), leading to its abnormal accumulation at presynaptic terminals. Here, we generated transgenic mice that selectively recreate the presynaptic accumulation of NrxnCTF in adult forebrain neurons, leaving unaltered the function of PS/gamma-secretase complex towards other substrates. Behavioral characterization identified selective impairments in NrxnCTF mice, including decreased fear-conditioning memory. Electrophysiological recordings in medial prefrontal cortex-basolateral amygdala (mPFC-BLA) of behaving mice showed normal synaptic transmission and uncovered specific defects in synaptic facilitation. These data functionally link the accumulation of NrxnCTF with defects in associative memory and short-term synaptic plasticity, pointing at impaired clearance of NrxnCTF as a new mediator in AD.
Collapse
Affiliation(s)
- Ana C Sánchez-Hidalgo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | - Francisco Arias-Aragón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | | | - Celia Martín-Cuevas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | | | - Amalia Martinez-Mir
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain
| | - Francisco G Scholl
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain.
| |
Collapse
|
31
|
Kron NS, Fieber LA. Aplysia Neurons as a Model of Alzheimer's Disease: Shared Genes and Differential Expression. J Mol Neurosci 2021; 72:287-302. [PMID: 34664226 PMCID: PMC8840921 DOI: 10.1007/s12031-021-01918-3] [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: 08/05/2021] [Accepted: 09/18/2021] [Indexed: 11/19/2022]
Abstract
Although Alzheimer’s disease (AD) is the most common form of dementia in the United States, development of therapeutics has proven difficult. Invertebrate alternatives to current mammalian AD models have been successfully employed to study the etiology of the molecular hallmarks of AD. The marine snail Aplysia californica offers a unique and underutilized system in which to study the physiological, behavioral, and molecular impacts of AD. Mapping of the Aplysia proteome to humans and cross-referencing with two databases of genes of interest in AD research identified 898 potential orthologs of interest in Aplysia. Included among these orthologs were alpha, beta and gamma secretases, amyloid-beta, and tau. Comparison of age-associated differential expression in Aplysia sensory neurons with that of late-onset AD in the frontal lobe identified 59 ortholog with concordant differential expression across data sets. The 21 concordantly upregulated genes suggested increased cellular stress and protein dyshomeostasis. The 47 concordantly downregulated genes included important components of diverse neuronal processes, including energy metabolism, mitochondrial homeostasis, synaptic signaling, Ca++ regulation, and cellular cargo transport. Compromised functions in these processes are known hallmarks of both human aging and AD, the ramifications of which are suggested to underpin cognitive declines in aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Nicholas S Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA.
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| |
Collapse
|
32
|
Eysert F, Coulon A, Boscher E, Vreulx AC, Flaig A, Mendes T, Hughes S, Grenier-Boley B, Hanoulle X, Demiautte F, Bauer C, Marttinen M, Takalo M, Amouyel P, Desai S, Pike I, Hiltunen M, Chécler F, Farinelli M, Delay C, Malmanche N, Hébert SS, Dumont J, Kilinc D, Lambert JC, Chapuis J. Alzheimer's genetic risk factor FERMT2 (Kindlin-2) controls axonal growth and synaptic plasticity in an APP-dependent manner. Mol Psychiatry 2021; 26:5592-5607. [PMID: 33144711 PMCID: PMC8758496 DOI: 10.1038/s41380-020-00926-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Although APP metabolism is being intensively investigated, a large fraction of its modulators is yet to be characterized. In this context, we combined two genome-wide high-content screenings to assess the functional impact of miRNAs and genes on APP metabolism and the signaling pathways involved. This approach highlighted the involvement of FERMT2 (or Kindlin-2), a genetic risk factor of Alzheimer's disease (AD), as a potential key modulator of axon guidance, a neuronal process that depends on the regulation of APP metabolism. We found that FERMT2 directly interacts with APP to modulate its metabolism, and that FERMT2 underexpression impacts axonal growth, synaptic connectivity, and long-term potentiation in an APP-dependent manner. Last, the rs7143400-T allele, which is associated with an increased AD risk and localized within the 3'UTR of FERMT2, induced a downregulation of FERMT2 expression through binding of miR-4504 among others. This miRNA is mainly expressed in neurons and significantly overexpressed in AD brains compared to controls. Altogether, our data provide strong evidence for a detrimental effect of FERMT2 underexpression in neurons and insight into how this may influence AD pathogenesis.
Collapse
Affiliation(s)
- Fanny Eysert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Audrey Coulon
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Emmanuelle Boscher
- Centre de Recherche du CHU de Québec-Université Laval, CHUL, Axe Neurosciences, Québec City, QC, Canada
- Faculté de Médecine, Département de Psychiatrie et de Neurosciences, Université Laval, Québec City, QC, Canada
| | - Anaїs-Camille Vreulx
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Amandine Flaig
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Tiago Mendes
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Sandrine Hughes
- E-Phy-Science, Bioparc de Sophia Antipolis, 2400 route des Colles, Biot, 06410, France
| | - Benjamin Grenier-Boley
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Xavier Hanoulle
- Université de Lille, CNRS, UMR8576-Labex DISTALZ, Villeneuve d'Ascq, 59655, France
| | - Florie Demiautte
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Charlotte Bauer
- Université Côte d'Azur, Inserm, CNRS, IPMC, DistAlz Laboratory of Excellence, Valbonne, France
| | - Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Philippe Amouyel
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Shruti Desai
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Ian Pike
- Proteome Sciences plc, Hamilton House, London, WC1H 9BB, UK
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Frédéric Chécler
- Université Côte d'Azur, Inserm, CNRS, IPMC, DistAlz Laboratory of Excellence, Valbonne, France
| | - Mélissa Farinelli
- E-Phy-Science, Bioparc de Sophia Antipolis, 2400 route des Colles, Biot, 06410, France
| | - Charlotte Delay
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Nicolas Malmanche
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Sébastien S Hébert
- Centre de Recherche du CHU de Québec-Université Laval, CHUL, Axe Neurosciences, Québec City, QC, Canada
- Faculté de Médecine, Département de Psychiatrie et de Neurosciences, Université Laval, Québec City, QC, Canada
| | - Julie Dumont
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Devrim Kilinc
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Julien Chapuis
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France.
| |
Collapse
|
33
|
Piubelli L, Murtas G, Rabattoni V, Pollegioni L. The Role of D-Amino Acids in Alzheimer's Disease. J Alzheimers Dis 2021; 80:475-492. [PMID: 33554911 DOI: 10.3233/jad-201217] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), the main cause of dementia worldwide, is characterized by a complex and multifactorial etiology. In large part, excitatory neurotransmission in the central nervous system is mediated by glutamate and its receptors are involved in synaptic plasticity. The N-methyl-D-aspartate (NMDA) receptors, which require the agonist glutamate and a coagonist such as glycine or the D-enantiomer of serine for activation, play a main role here. A second D-amino acid, D-aspartate, acts as agonist of NMDA receptors. D-amino acids, present in low amounts in nature and long considered to be of bacterial origin, have distinctive functions in mammals. In recent years, alterations in physiological levels of various D-amino acids have been linked to various pathological states, ranging from chronic kidney disease to neurological disorders. Actually, the level of NMDA receptor signaling must be balanced to promote neuronal survival and prevent neurodegeneration: this signaling in AD is affected mainly by glutamate availability and modulation of the receptor's functions. Here, we report the experimental findings linking D-serine and D-aspartate, through NMDA receptor modulation, to AD and cognitive functions. Interestingly, AD progression has been also associated with the enzymes related to D-amino acid metabolism as well as with glucose and serine metabolism. Furthermore, the D-serine and D-/total serine ratio in serum have been recently proposed as biomarkers of AD progression. A greater understanding of the role of D-amino acids in excitotoxicity related to the pathogenesis of AD will facilitate novel therapeutic treatments to cure the disease and improve life expectancy.
Collapse
Affiliation(s)
- Luciano Piubelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Giulia Murtas
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Valentina Rabattoni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
34
|
Abstract
GABAB receptors are implicated in numerous central nervous system-based behaviours and mechanisms, including cognitive processing in preclinical animal models. Homeostatic changes in the expression and function of these receptors across brain structures have been found to affect cognitive processing. Numerous preclinical studies have focused on the role of GABAB receptors in learning, memory and cognition per se with some interesting, although sometimes contradictory, findings. The majority of the existing clinical literature focuses on alterations in GABAB receptor function in conditions and disorders whose main symptomatology includes deficits in cognitive processing. The aim of this chapter is to delineate the role of GABAB receptors in cognitive processes in health and disease of animal models and human clinical populations. More specifically, this review aims to present literature on the role of GABAB receptors in animal models with cognitive deficits, especially those of learning and memory. Further, it aims to capture the progress and advances of research studies on the effects of GABAB receptor compounds in neurodevelopmental and neurodegenerative conditions with cognitive dysfunctions. The neurodevelopmental conditions covered include autism spectrum disorders, fragile X syndrome and Down's syndrome and the neurodegenerative conditions discussed are Alzheimer's disease, epilepsy and autoimmune anti-GABAB encephalitis. Although some findings are contradictory, results indicate a possible therapeutic role of GABAB receptor compounds for the treatment of cognitive dysfunction and learning/memory impairments for some of these conditions, especially in neurodegeneration. Moreover, future research efforts should aim to develop selective GABAB receptor compounds with minimal, if any, side effects.
Collapse
|
35
|
Cortical Circuitry and Synaptic Dysfunctions in Alzheimer's Disease and Other Dementias. Neural Plast 2021; 2021:9796576. [PMID: 34394342 PMCID: PMC8360713 DOI: 10.1155/2021/9796576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
|
36
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
37
|
Byrne RAJ, Torvell M, Daskoulidou N, Fathalla D, Kokkali E, Carpanini SM, Morgan BP. Novel Monoclonal Antibodies Against Mouse C1q: Characterisation and Development of a Quantitative ELISA for Mouse C1q. Mol Neurobiol 2021; 58:4323-4336. [PMID: 34002346 PMCID: PMC8487419 DOI: 10.1007/s12035-021-02419-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
Recent studies have identified roles for complement in synaptic pruning, both physiological during development and pathological in Alzheimer's disease (AD). These reports suggest that C1q initiates complement activation on synapses and C3 fragments then tag them for removal by microglia. There is an urgent need to characterise these processes in rodent AD models; this requires the development of reagents and methods for detection and quantification of rodent C1q in fluids and pathological tissues. These will enable better evaluation of the role of C1q in disease and its value as disease biomarker. We describe the generation in C1q-deficient mice of novel monoclonal antibodies against mouse and rat C1q that enabled development of a sensitive, specific, and quantitative ELISA for mouse and rat C1q capable of measuring C1q in biological fluids and tissue extracts. Serum C1q levels were measured in wild-type (WT), C1q knockout (KO), C3 KO, C7 KO, Crry KO, and 3xTg and APPNL-G-F AD model mice through ageing. C1q levels significantly decreased in WT, APPNL-G-F, and C7 KO mice with ageing. C1q levels were reduced in APPNL-G-F compared to WT at all ages and in 3xTg at 12 months; C3 KO and C7 KO, but not Crry KO mice, also demonstrated significantly lower C1q levels compared to matched WT. In brain homogenates, C1q levels increased with age in both WT and APPNL-G-F mice. This robust and adaptable assay for quantification of mouse and rat C1q provides a vital tool for investigating the expression of C1q in rodent models of AD and other complement-driven pathologies.
Collapse
Affiliation(s)
- Robert A J Byrne
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Megan Torvell
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Dina Fathalla
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Eirini Kokkali
- School of Optometry and Visual Sciences, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK. .,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK.
| |
Collapse
|
38
|
Haytural H, Jordà-Siquier T, Winblad B, Mulle C, Tjernberg LO, Granholm AC, Frykman S, Barthet G. Distinctive alteration of presynaptic proteins in the outer molecular layer of the dentate gyrus in Alzheimer's disease. Brain Commun 2021; 3:fcab079. [PMID: 34013204 PMCID: PMC8117432 DOI: 10.1093/braincomms/fcab079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Synaptic degeneration has been reported as one of the best pathological correlates of cognitive deficits in Alzheimer's disease. However, the location of these synaptic alterations within hippocampal sub-regions, the vulnerability of the presynaptic versus postsynaptic compartments, and the biological mechanisms for these impairments remain unknown. Here, we performed immunofluorescence labelling of different synaptic proteins in fixed and paraffin-embedded human hippocampal sections and report reduced levels of several presynaptic proteins of the neurotransmitter release machinery (complexin-1, syntaxin-1A, synaptotagmin-1 and synaptogyrin-1) in Alzheimer's disease cases. The deficit was restricted to the outer molecular layer of the dentate gyrus, whereas other hippocampal sub-fields were preserved. Interestingly, standard markers of postsynaptic densities (SH3 and multiple ankyrin repeat domains protein 2) and dendrites (microtubule-associated protein 2) were unaltered, as well as the relative number of granule cells in the dentate gyrus, indicating that the deficit is preferentially presynaptic. Notably, staining for the axonal components, myelin basic protein, SMI-312 and Tau, was unaffected, suggesting that the local presynaptic impairment does not result from axonal loss or alterations of structural proteins of axons. There was no correlation between the reduction in presynaptic proteins in the outer molecular layer and the extent of the amyloid load or of the dystrophic neurites expressing phosphorylated forms of Tau. Altogether, this study highlights the distinctive vulnerability of the outer molecular layer of the dentate gyrus and supports the notion of presynaptic failure in Alzheimer's disease.
Collapse
Affiliation(s)
- Hazal Haytural
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Tomàs Jordà-Siquier
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
- Karolinska University Hospital, Theme Aging, 141 86 Huddinge, Sweden
| | - Christophe Mulle
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Ann-Charlotte Granholm
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
- Knoebel Institute for Healthy Aging, University of Denver, Denver 80208, CO, USA
| | - Susanne Frykman
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Gaël Barthet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| |
Collapse
|
39
|
Xu Y, Song X, Wang D, Wang Y, Li P, Li J. Proteomic insights into synaptic signaling in the brain: the past, present and future. Mol Brain 2021; 14:37. [PMID: 33596935 PMCID: PMC7888154 DOI: 10.1186/s13041-021-00750-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/09/2021] [Indexed: 12/29/2022] Open
Abstract
Chemical synapses in the brain connect neurons to form neural circuits, providing the structural and functional bases for neural communication. Disrupted synaptic signaling is closely related to a variety of neurological and psychiatric disorders. In the past two decades, proteomics has blossomed as a versatile tool in biological and biomedical research, rendering a wealth of information toward decoding the molecular machinery of life. There is enormous interest in employing proteomic approaches for the study of synapses, and substantial progress has been made. Here, we review the findings of proteomic studies of chemical synapses in the brain, with special attention paid to the key players in synaptic signaling, i.e., the synaptic protein complexes and their post-translational modifications. Looking toward the future, we discuss the technological advances in proteomics such as data-independent acquisition mass spectrometry (DIA-MS), cross-linking in combination with mass spectrometry (CXMS), and proximity proteomics, along with their potential to untangle the mystery of how the brain functions at the molecular level. Last but not least, we introduce the newly developed synaptomic methods. These methods and their successful applications marked the beginnings of the synaptomics era.
Collapse
Affiliation(s)
- Yalan Xu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Xiuyue Song
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Dong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Jing Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
40
|
Piubelli L, Pollegioni L, Rabattoni V, Mauri M, Princiotta Cariddi L, Versino M, Sacchi S. Serum D-serine levels are altered in early phases of Alzheimer's disease: towards a precocious biomarker. Transl Psychiatry 2021; 11:77. [PMID: 33500383 PMCID: PMC7838302 DOI: 10.1038/s41398-021-01202-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
D-Serine acts as a co-agonist of N-methyl-D-aspartate receptors (NMDAR) which appear overactivated in AD, while D-aspartate is a modulatory molecule acting on NMDAR as a second agonist. The aim of this work is to clarify whether the levels of these D-amino acids in serum are deregulated in AD, with the final goal to identify novel and precocious biomarkers in AD. Serum levels of L- and D-enantiomers of serine and aspartate were determined by HPLC using a pre-column derivatization procedure and a selective enzymatic degradation. Experimental data obtained from age-matched healthy subjects (HS) and AD patients were statistically evaluated by considering age, gender, and disease progression, and compared. Minor changes were apparent in the serum L- and D-aspartate levels in AD patients compared to HS. A positive correlation for the D-serine level and age was apparent in the AD cohort. Notably, the serum D-serine level and the D-/total serine ratio significantly increased with the progression of the disease. Gender seems to have a minor effect on the levels of all analytes tested. This work proposes that the serum D-serine level and D-/total serine ratio values as novel and valuable biomarkers for the progression of AD: the latter parameter allows to discriminate CDR 2 and CDR 1 patients from healthy (CDR 0) individuals.
Collapse
Affiliation(s)
- Luciano Piubelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Valentina Rabattoni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Marco Mauri
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Neurology Unit, Ospedale di Circolo and Fondazione Macchi, ASST Settelaghi, Varese, Italy
| | - Lucia Princiotta Cariddi
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Maurizio Versino
- Neurology Unit, Ospedale di Circolo and Fondazione Macchi, ASST Settelaghi, Varese, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Silvia Sacchi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
41
|
Kees AL, Marneffe C, Mulle C. Lighting up pre-synaptic potentiation: An Editorial for "SynaptoPAC, an optogenetic tool for induction of presynaptic plasticity" on page 324. J Neurochem 2020; 156:270-272. [PMID: 33274445 DOI: 10.1111/jnc.15236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 10/22/2022]
Abstract
This is an Editorial Highlight of a manuscript by Oldani et al. (2020) (Oldani et al. 2020) in the current issue of the Journal of Neurochemistry, in which the authors describe synaptoPAC, a new optogenetic tool. SynaptoPAC is targeted to pre-synaptic compartments and can be used for light-induced increase of the levels of cAMP. Pre-synaptic plasticity, defined as activity-dependent modulation of neurotransmitter release, occurs over a variety of time scales. At a subset of synapses in the brain, long-term forms of pre-synaptic facilitation depend on an increase in the levels of cAMP. Light-induced modulation of cAMP at synapses expressing cAMP-dependent facilitation, has the great potential to mimic pre-synaptic plasticity at genetically targeted synapses. Therefore, synaptoPAC constitutes a powerful tool to study the role of pre-synaptic potentiation in the activity of selected neuronal circuits in relation to behaving animals with a high temporal and spatial precision.
Collapse
Affiliation(s)
- Ashley L Kees
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, University of Bordeaux, Bordeaux, France
| | - Catherine Marneffe
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, University of Bordeaux, Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, University of Bordeaux, Bordeaux, France
| |
Collapse
|
42
|
Lam AD, Noebels J. Night Watch on the Titanic: Detecting Early Signs of Epileptogenesis in Alzheimer Disease. Epilepsy Curr 2020; 20:369-374. [PMID: 33081517 PMCID: PMC7818196 DOI: 10.1177/1535759720964775] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aberrant cortical network excitability is an inextricable feature of Alzheimer disease (AD) that can negatively impact memory and accelerate cognitive decline. Surface electroencephalogram spikes and intracranial recordings of nocturnal silent seizures in human AD, coupled with the abnormal neural synchrony that precedes development of behavioral seizures in mouse AD models, build the case for epileptogenesis as an early therapeutic target for AD. Since most individuals with AD do not develop overt seizures, leveraging functional biomarkers of epilepsy risk to stratify a heterogeneous AD patient population for treatment is research priority for successful clinical trial design. Who will benefit from antiseizure interventions, which one, and when should it begin?
Collapse
Affiliation(s)
- Alice D. Lam
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jeffrey Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
43
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|