1
|
Chaigneau T, Sha S, Roux CM, Aïd S, Faucher A, Chantran Y, Dorothée G, Krantic S. Subtle Alterations in Hippocampal Neuronal Activity Coincide With Early Sex-Specific Differences in Amyloidosis and Microglia in a Pre-Symptomatic Mouse Model of Alzheimer-Like Pathology. Glia 2025. [PMID: 40304030 DOI: 10.1002/glia.70029] [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: 09/04/2024] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
Growing evidence highlights sex-related differences in the pathogenesis of Alzheimer's disease (AD). Yet, early impact of sex on neuronal activity and microglia in the hippocampus, a main site of memory formation and one of the most vulnerable brain areas in AD, remains poorly understood. We thus assessed these issues by using APPPS1 mouse model of AD-like amyloid pathology at a pre-symptomatic stage (5-6 months). Our electrophysiological data point to opposite alterations in hippocampal CA1 neurons' basal glutamatergic neurotransmission and response to excitatory inputs between male and female APPPS1 mice. These complex changes in neuronal activity are likely to precede plasticity impairments, which do not yet translate into sexual dimorphism of Long-Term Potentiation (LTP) at the studied age. Alteration in synaptic transmission in males coincides with an increased number and coverage of microglia, together with increased plaque coverage, as compared to the female hippocampus. Such increased microgliosis in males is accompanied by complex sex-related differences in the expression of specific transcriptomic markers Disease-Associated Microglia (DAM)/Microglial neurodegenerative phenotype (MGnD), whereas homeostatic (M0) markers were unaffected. Our data show for the first time that subtle alterations in hippocampal neuronal activity coincide with early sex-related differences in amyloidosis and microglia already at the pre-symptomatic stage of AD-like pathology.
Collapse
Affiliation(s)
- Thomas Chaigneau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Sha Sha
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Candice M Roux
- Normandie University, UNICAEN, INSERM, COMETE, CYCERON, CHU de Caen, Caen, France
| | - Saba Aïd
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Alice Faucher
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Yannick Chantran
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Guillaume Dorothée
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Slavica Krantic
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Neuroimmunology, Inflammation and Therapeutics Laboratory, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
2
|
Pereyra G, Mateo MI, Miaja P, Martin-Bermejo MJ, Martinez-Baños M, Klaassen R, Gruart A, Rueda-Carrasco J, Fernández-Rodrigo A, López-Merino E, Esteve P, Esteban JA, Smit AB, Delgado-García JM, Bovolenta P. SFRP1 upregulation causes hippocampal synaptic dysfunction and memory impairment. Cell Rep 2025; 44:115535. [PMID: 40198223 DOI: 10.1016/j.celrep.2025.115535] [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: 10/22/2024] [Revised: 01/30/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
Impaired neuronal and synaptic function are hallmarks of early Alzheimer's disease (AD), preceding other neuropathological traits and cognitive decline. We previously showed that SFRP1, a glial-derived protein elevated in AD brains from preclinical stages, contributes to disease progression, implicating glial factors in early pathogenesis. Here, we generate and analyze transgenic mice overexpressing astrocytic SFRP1. SFRP1 accumulation causes early dendritic and synaptic defects in adult mice, followed by impaired synaptic long-term potentiation and cognitive decline, evident only when the animals age, thereby mimicking AD's structural-functional temporal distinction. This phenotype correlates with proteomic changes, including increased structural synaptic proteins like neurexin, which localizes in close proximity with SFRP1 in cultured hippocampal neurons. We conclude that excessive SFRP1 hinders synaptic protein turnover, reducing synaptic plasticity-a mechanism that may underlie the synaptopathy observed in the brains of prodromal AD patients.
Collapse
Affiliation(s)
- Guadalupe Pereyra
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - María Inés Mateo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Pablo Miaja
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - María Jesús Martin-Bermejo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Marcos Martinez-Baños
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Remco Klaassen
- Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 Amsterdam, the Netherlands
| | - Agnès Gruart
- División de Neurociencias, Universidad Pablo de Olavide, 41013 Seville, Spain
| | - Javier Rueda-Carrasco
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Alba Fernández-Rodrigo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Esperanza López-Merino
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Pilar Esteve
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 Amsterdam, the Netherlands
| | | | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| |
Collapse
|
3
|
Moreira P, Macedo J, Matos P, Bicker J, Fortuna A, Figueirinha A, Salgueiro L, Batista MT, Silva A, Silva S, Resende R, Branco PC, Cruz MT, Pereira CF. Effect of bioactive extracts from Eucalyptus globulus leaves in experimental models of Alzheimer's disease. Biomed Pharmacother 2024; 181:117652. [PMID: 39486370 DOI: 10.1016/j.biopha.2024.117652] [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: 08/01/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Current therapies for Alzheimer's disease (AD) do not delay its progression, therefore, novel disease-modifying strategies are urgently needed. Recently, an increasing number of compounds from natural origin with protective properties against AD have been identified. Mixtures or extracts obtained from natural products containing several bioactive compounds have multifunctional properties and have drawn the attention because multiple AD pathways can be simultaneously modulated. This study evaluated the in vitro and in vivo effect of the essential oil (EO) obtained from the hydrodistillation of Eucalyptus globulus leaves, and an extract obtained from the hydrodistillation residual water (HRW). It was observed that EO and HRW have anti-inflammatory effect in brain immune cells modeling AD, namely lipopolysaccharide (LPS)- and amyloid-beta (Aβ)-stimulated microglia. In cell models that mimic AD-related neuronal dysfunction, HRW attenuated Aβ secretion and Aβ-induced mitochondrial dysfunction. Since the HRW's major components did not cross the blood-brain barrier, both EO and HRW were administered to the APP/PS1 transgenic AD mouse model by an intranasal route, which reduced cortical and hippocampal Aβ levels, and to rescue memory deficits and anxiety-like behaviors. Finally, HRW and EO were found to regulate cholesterol levels in aged mice after intranasal administration, suggesting that these extracts can reduce hypercholesterolemia and avoid risk for AD development. Overall, findings support a protective role of E. globulus extracts against AD‑like pathology and cognitive impairment highlighting the underlying mechanisms. These extracts obtained from underused forest biomass could be useful to develop nutraceutical supplements helpful to avoid AD risk and to prevent its progression.
Collapse
Affiliation(s)
- Patrícia Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Jéssica Macedo
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Patrícia Matos
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Maria Teresa Batista
- CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Silva
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Sónia Silva
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; iCBR-Coimbra Institute for Clinical and Biomedical Research, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Pedro Costa Branco
- RAIZ-Forest and Paper Research Institute, Eixo, Aveiro 3800-783, Portugal
| | - Maria Teresa Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Cláudia Fragão Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| |
Collapse
|
4
|
Grosso Jasutkar H, Wasserlein EM, Ishola A, Litt N, Staniszewski A, Arancio O, Yamamoto A. Adult-onset deactivation of autophagy leads to loss of synapse homeostasis and cognitive impairment, with implications for alzheimer disease. Autophagy 2024; 20:2540-2555. [PMID: 38949671 PMCID: PMC11572145 DOI: 10.1080/15548627.2024.2368335] [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: 03/03/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024] Open
Abstract
A growing number of studies link dysfunction of macroautophagy/autophagy to the pathogenesis of diseases such as Alzheimer disease (AD). Given the global importance of autophagy for homeostasis, how its dysfunction can lead to specific neurological changes is puzzling. To examine this further, we compared the global deactivation of autophagy in the adult mouse using the atg7iKO with the impact of AD-associated pathogenic changes in autophagic processing of synaptic proteins. Isolated forebrain synaptosomes, rather than total homogenates, from atg7iKO mice demonstrated accumulation of synaptic proteins, suggesting that the synapse might be a vulnerable site for protein homeostasis disruption. Moreover, the deactivation of autophagy resulted in impaired cognitive performance over time, whereas gross locomotor skills remained intact. Despite deactivation of autophagy for 6.5 weeks, changes in cognition were in the absence of cell death or synapse loss. In the symptomatic APP PSEN1 double-transgenic mouse model of AD, we found that the impairment in autophagosome maturation coupled with diminished presence of discrete synaptic proteins in autophagosomes isolated from these mice, leading to the accumulation of one of these proteins in the detergent insoluble protein fraction. This protein, SLC17A7/Vglut, also accumulated in atg7iKO mouse synaptosomes. Taken together, we conclude that synaptic autophagy plays a role in maintaining protein homeostasis, and that while decreasing autophagy interrupts normal cognitive function, the preservation of locomotion suggests that not all circuits are affected similarly. Our data suggest that the disruption of autophagic activity in AD may have relevance for the cognitive impairment in this adult-onset neurodegenerative disease. Abbreviations: 2dRAWM: 2-day radial arm water maze; AD: Alzheimer disease; Aβ: amyloid-beta; AIF1/Iba1: allograft inflammatory factor 1; APP: amyloid beta precursor protein; ATG7: autophagy related 7; AV: autophagic vacuole; CCV: cargo capture value; Ctrl: control; DLG4/PSD-95: discs large MAGUK scaffold protein 4; GFAP: glial fibrillary acidic protein; GRIN2B/NMDAR2b: glutamate ionotropic receptor NMDA type subunit 2B; LTD: long-term depression; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; m/o: months-old; PNS: post-nuclear supernatant; PSEN1/PS1: presenilin 1; SHB: sucrose homogenization buffer; SLC32A1/Vgat: solute carrier family 32 member 1; SLC17A7/Vglut1: solute carrier family 17 member 7; SNAP25: synaptosome associated protein 25; SQSTM1/p62: sequestosome 1; SYN1: synapsin I; SYP: synaptophysin ; SYT1: synaptotagmin 1; Tam: tamoxifen; VAMP2: vesicle associated membrane protein 2; VCL: vinculin; wks: weeks.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | | - Azeez Ishola
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Nicole Litt
- Department of Neurology, Columbia University, New York, NY, USA
| | - Agnieszka Staniszewski
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Ai Yamamoto
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
5
|
da Silva Correia A, Schmitz M, Fischer A, da Silva Correia S, Simonetti FL, Saher G, Goya‐Maldonado R, Arora AS, Fischer A, Outeiro TF, Zerr I. Cellular prion protein acts as mediator of amyloid beta uptake by caveolin-1 causing cellular dysfunctions in vitro and in vivo. Alzheimers Dement 2024; 20:6776-6792. [PMID: 39212313 PMCID: PMC11485400 DOI: 10.1002/alz.14120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Cellular prion protein (PrPC) was implicated in amyloid beta (Aβ)-induced toxicity in Alzheimer's disease (AD), but the precise molecular mechanisms involved in this process are unclear. METHODS Double transgenic mice were generated by crossing Prnp knockout (KO) with 5xFAD mice, and light-sheet microscopy was used for whole brain tissue analyses. PrPC-overexpressing cells were developed for in vitro studies, and microscopy was used to assess co-localization of proteins of interest. Surface-plasmon resonance (SPR) was used to investigate protein-binding characteristics. RESULTS In vivo, PrPC levels correlated with reduced lifespan and cognitive and motor function, and its ablation disconnected behavior deficits from Aβ levels. Light-sheet microscopy showed that PrPC influenced Aβ-plaque burden but not the distribution of those plaques. Interestingly, caveolin-1 (Cav-1) KO neurons significantly reduced intracellular Aβ-oligomer (Aβo) uptake when compared to wild-type neurons. DISCUSSION The findings shed new light on the relevance of intracellular Aβo, suggesting that PrPC and Cav-1 modulate intracellular Aβ levels and the Aβ-plaque load. HIGHLIGHTS PrPC expression adversely affects lifespan and behavior in 5xFAD mice. PrPC increases Aβ1-40 and Aβ1-42 levels and Aβ-plaque load in 5xFAD mice. Cav-1 interacts with both PrPC and Aβ peptides. Knocking out Cav-1 leads to a significant reduction in intracellular Aβ levels.
Collapse
Affiliation(s)
- Angela da Silva Correia
- Department of NeurologyUniversity Medical Center and the German Center for Neurodegenerative Diseases (DZNE)Georg‐August UniversityGoettingenGermany
| | - Matthias Schmitz
- Department of NeurologyUniversity Medical Center and the German Center for Neurodegenerative Diseases (DZNE)Georg‐August UniversityGoettingenGermany
| | - Anna‐Lisa Fischer
- Department of NeurologyUniversity Medical Center and the German Center for Neurodegenerative Diseases (DZNE)Georg‐August UniversityGoettingenGermany
| | - Susana da Silva Correia
- Department of NeurologyUniversity Medical Center and the German Center for Neurodegenerative Diseases (DZNE)Georg‐August UniversityGoettingenGermany
| | | | - Gesine Saher
- Department of NeurogeneticsMax Planck Institute for Multidisciplinary SciencesGoettingenGermany
| | - Roberto Goya‐Maldonado
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP‐Lab)Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMG)GoettingenGermany
| | - Amandeep Singh Arora
- Texas Therapeutics InstituteBrown Foundation Institute of Molecular MedicineUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Andre Fischer
- Department of Psychiatry and PsychotherapyUniversity Medical Center GoettingenGoettingenGermany
- Department for Systems Medicine and Epigenetics in Neurodegenerative DiseasesGerman Center for Neurodegenerative Diseases (DZNE) GoettingenGoettingenGermany
| | - Tiago F. Outeiro
- Department of Experimental NeurodegenerationCenter for Nanoscale Microscopy and Molecular Physiology of the BrainCenter for Biostructural Imaging of NeurodegenerationUniversity Medical Center GoettingenGoettingenGermany
- Max Planck Institute for Multidisciplinary SciencesGoettingenGermany
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastleUK
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)GoettingenGermany
| | - Inga Zerr
- Department of NeurologyUniversity Medical Center and the German Center for Neurodegenerative Diseases (DZNE)Georg‐August UniversityGoettingenGermany
| |
Collapse
|
6
|
Yu L, Li Y, Lv Y, Gu B, Cai J, Liu QS, Zhao L. Treadmill Exercise Facilitates Synaptic Plasticity in APP/PS1 Mice by Regulating Hippocampal AMPAR Activity. Cells 2024; 13:1608. [PMID: 39404372 PMCID: PMC11475322 DOI: 10.3390/cells13191608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Accumulating evidence underscores exercise as a straightforward and cost-effective lifestyle intervention capable of mitigating the risk and slowing the emergence and progression of Alzheimer's disease (AD). However, the intricate cellular and molecular mechanisms mediating these exercise-induced benefits in AD remain elusive. The present study delved into the impact of treadmill exercise on memory retrieval performance, hippocampal synaptic plasticity, synaptic morphology, and the expression and activity of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPARs) in 6-month-old APP/PS1 mice. APP/PS1 mice (4-month-old males) were randomly assigned to either a treadmill exercise group or a sedentary group, with C57BL/6J mice (4-month-old males) as the control group (both exercise and sedentary). The exercise regimen spanned 8 weeks. Our findings revealed that 8-week treadmill exercise reversed memory retrieval impairment in step-down fear conditioning in 6-month-old APP/PS1 mice. Additionally, treadmill exercise enhanced basic synaptic strength, short-term potentiation (STP), and long-term potentiation (LTP) of the hippocampus in these mice. Moreover, treadmill exercise correlated with an augmentation in synapse numbers, refinement of synaptic structures, and heightened expression and activity of AMPARs. Our findings suggest that treadmill exercise improves behavioral performance and facilitates synaptic transmission by increasing structural synaptic plasticity and the activity of AMPARs in the hippocampus of 6-month-old APP/PS1 mice, which is involved in pre- and postsynaptic processes.
Collapse
Affiliation(s)
- Laikang Yu
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing 100084, China;
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yan Li
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yuanyuan Lv
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Boya Gu
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Jiajia Cai
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| |
Collapse
|
7
|
Sánchez-Fernández N, Gómez-Acero L, Castañé A, Adell A, Campa L, Bonaventura J, Brito V, Ginés S, Queiróz F, Silva H, Lopes JP, Lopes CR, Radošević M, Gasull X, Cunha RA, Köfalvi A, Ferreira SG, Ciruela F, Aso E. A combination of Δ 9-tetrahydrocannabinol and cannabidiol modulates glutamate dynamics in the hippocampus of an animal model of Alzheimer's disease. Neurotherapeutics 2024; 21:e00439. [PMID: 39232876 PMCID: PMC11581878 DOI: 10.1016/j.neurot.2024.e00439] [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: 04/25/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
A combination of Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) at non-psychoactive doses was previously demonstrated to reduce cognitive decline in APP/PS1 mice, an animal model of Alzheimer's disease (AD). However, the neurobiological substrates underlying these therapeutic properties of Δ9-THC and CBD are not fully understood. Considering that dysregulation of glutamatergic activity contributes to cognitive impairment in AD, the present study evaluates the hypothesis that the combination of these two natural cannabinoids might reverse the alterations in glutamate dynamics within the hippocampus of this animal model of AD. Interestingly, our findings reveal that chronic treatment with Δ9-THC and CBD, but not with any of them alone, reduces extracellular glutamate levels and the basal excitability of the hippocampus in APP/PS1 mice. These effects are not related to significant changes in the function and structure of glutamate synapses, as no relevant changes in synaptic plasticity, glutamate signaling or in the levels of key components of these synapses were observed in cannabinoid-treated mice. Our data instead indicate that these cannabinoid effects are associated with the control of glutamate uptake and/or to the regulation of the hippocampal network. Taken together, these results support the potential therapeutic properties of combining these natural cannabinoids against the excitotoxicity that occurs in AD brains.
Collapse
Affiliation(s)
- Nuria Sánchez-Fernández
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Laura Gómez-Acero
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Anna Castañé
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08036 Barcelona, Spain; Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500 Vic, Spain
| | - Albert Adell
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria, 39011 Santander, Spain
| | - Leticia Campa
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Bonaventura
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Verónica Brito
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Silvia Ginés
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Francisco Queiróz
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Henrique Silva
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Cátia R Lopes
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Marija Radošević
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Xavier Gasull
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; FMUC-Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Attila Köfalvi
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Samira G Ferreira
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
8
|
Abdulhameed N, Babin A, Hansen K, Weaver R, Banks WA, Talbot K, Rhea EM. Comparing regional brain uptake of incretin receptor agonists after intranasal delivery in CD-1 mice and the APP/PS1 mouse model of Alzheimer's disease. Alzheimers Res Ther 2024; 16:173. [PMID: 39085976 PMCID: PMC11293113 DOI: 10.1186/s13195-024-01537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
Targeting brain insulin resistance (BIR) has become an attractive alternative to traditional therapeutic treatments for Alzheimer's disease (AD). Incretin receptor agonists (IRAs), targeting either or both of the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors, have proven to reverse BIR and improve cognition in mouse models of AD. We previously showed that many, but not all, IRAs can cross the blood-brain barrier (BBB) after intravenous (IV) delivery. Here we determined if widespread brain uptake of IRAs could be achieved by circumventing the BBB using intranasal (IN) delivery, which has the added advantage of minimizing adverse gastrointestinal effects of systemically delivered IRAs. Of the 5 radiolabeled IRAs tested (exenatide, dulaglutide, semaglutide, DA4-JC, and DA5-CH) in CD-1 mice, exenatide, dulaglutide, and DA4-JC were successfully distributed throughout the brain following IN delivery. We observed significant sex differences in uptake for DA4-JC. Dulaglutide and DA4-JC exhibited high uptake by the hippocampus and multiple neocortical areas. We further tested and found the presence of AD-associated Aβ pathology minimally affected uptake of dulaglutide and DA4-JC. Of the 5 tested IRAs, dulaglutide and DA4-JC are best capable of accessing brain regions most vulnerable in AD (neocortex and hippocampus) after IN administration. Future studies will need to be performed to determine if IN IRA delivery can reduce BIR in AD or animal models of that disorder.
Collapse
Affiliation(s)
- Noor Abdulhameed
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Alice Babin
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Kim Hansen
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Riley Weaver
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA.
| |
Collapse
|
9
|
Song C, Li S, Mai Y, Li L, Dai G, Zhou Y, Liang X, Zou OM, Wang Y, Zhou L, Liu J, Zou Y. Dysregulated expression of miR-140 and miR-122 compromised microglial chemotaxis and led to reduced restriction of AD pathology. J Neuroinflammation 2024; 21:167. [PMID: 38956605 PMCID: PMC11218311 DOI: 10.1186/s12974-024-03162-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Deposition of amyloid β, which is produced by amyloidogenic cleavage of APP by β- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology. RESULTS In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aβ were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aβ phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets. CONCLUSIONS Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.
Collapse
Affiliation(s)
- Chao Song
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Shufang Li
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yingren Mai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Linpeng Li
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Guoku Dai
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yuan Zhou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaosheng Liang
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Olivia Meilan Zou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Ya Wang
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Jun Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| | - Yi Zou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
10
|
Wang M, Wei T, Yu C, Li R, Yin Y, Yang H, Di R, Xia X, Qin Q, Tang Y. Integrative Metabolomics and Whole Transcriptome Sequencing Reveal Role for TREM2 in Metabolism Homeostasis in Alzheimer's Disease. Mol Neurobiol 2024; 61:4188-4202. [PMID: 38066402 DOI: 10.1007/s12035-023-03840-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/27/2023] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia worldwide. Dysregulation of various metabolism pathways may mediate the development of AD pathology and cognitive dysfunction. Variants of triggering receptor expressed on myeloid cells-2 (TREM2) are known to increase the risk of developing AD. TREM2 plays a role in AD development by maintaining cellular energy and biosynthesis, but the precise mechanism through which it accomplishes this is unknown. Metabolomic analysis of hippocampal tissue from APP/PS1 and APP/PS1-TREM2 knockout (KO) mice found that TREM2 KO was associated with abnormalities in several metabolism pathways, and the effect was particularly pronounced in lipid metabolism and glucose metabolism pathways. Consistently, transcriptomic analysis of these mice determined that most differentially expressed genes were involved in energy metabolism pathways. We screened seven differentially expressed genes in APP/PS1-TREM2 KO mice that may influence AD development by altering energy metabolism. Integrative analysis of the metabolomic and transcriptomic profiles showed that TREM2 may regulate lipid metabolism and sphingolipid metabolism by affecting lipoprotein lipase (LPL) expression, thereby influencing AD progression. Our results prompt further studies of the interactions among TREM2, LPL, glucolipid metabolism, and sphingolipid metabolism in AD to identify new diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Meng Wang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Tao Wei
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Chaoji Yu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Ruiyang Li
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yunsi Yin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Hanchen Yang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Run Di
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Xinyi Xia
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qi Qin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- National Center for Neurological Disorders, 45 Changchun Street, Beijing, 100053, China.
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- National Center for Neurological Disorders, 45 Changchun Street, Beijing, 100053, China.
| |
Collapse
|
11
|
Zhang J, Chen Y, Zhao Y, Wang P, Ding H, Liu C, Lyu J, Le W. Terahertz Irradiation Improves Cognitive Impairments and Attenuates Alzheimer's Neuropathology in the APP SWE/PS1 DE9 Mouse: A Novel Therapeutic Intervention for Alzheimer's Disease. Neurosci Bull 2024; 40:857-871. [PMID: 37971654 PMCID: PMC11250709 DOI: 10.1007/s12264-023-01145-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the deposition of amyloid-β (Aβ), neurofibrillary tangles, neuroinflammation, and neurodegeneration in the brain. In recent years, considering the unsatisfied benefits of pharmacological therapies, non-pharmacological therapy has become a research hotspot for AD intervention. Terahertz (THz) waves with a range between microwave and infrared regions in the electromagnetic spectrum and high permeability to a wide range of materials have great potential in the bioengineering field. However, its biological impacts on the central nervous system, under either physiological or pathological conditions, are poorly investigated. In this study, we first measured the 0.14 THz waves penetration across the skull of a C57BL/6 mouse and found the percentage of THz penetration to be ~70%, guaranteeing that THz waves can reach the relevant brain regions. We then exposed the APPSWE/PS1DE9 mouse model of AD to repeated low-frequency THz waves on the head. We demonstrated that THz waves treatment significantly improved the cognitive impairment and alleviated AD neuropathology including Aβ deposition and tau hyperphosphorylation in the AD mice. Moreover, THz waves treatment effectively attenuated mitochondrial impairment, neuroinflammation, and neuronal loss in the AD mouse brain. Our findings reveal previously unappreciated beneficial effects of THz waves treatment in AD and suggest that THz waves may have the potential to be used as a novel therapeutic intervention for this devastating disease.
Collapse
Affiliation(s)
- Jun Zhang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yarui Zhao
- School of Physics, Dalian University of Technology, Dalian, 116024, China
| | - Panpan Wang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Hongbin Ding
- School of Physics, Dalian University of Technology, Dalian, 116024, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Junhong Lyu
- Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
- Department of Neurology and Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Medical School, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Cortese GP, Bartosch AMW, Xiao H, Gribkova Y, Lam TG, Argyrousi EK, Sivakumar S, Cardona C, Teich AF. ZCCHC17 knockdown phenocopies Alzheimer's disease-related loss of synaptic proteins and hyperexcitability. J Neuropathol Exp Neurol 2024; 83:626-635. [PMID: 38630575 PMCID: PMC11187431 DOI: 10.1093/jnen/nlae033] [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] [Indexed: 04/19/2024] Open
Abstract
ZCCHC17 is a master regulator of synaptic gene expression and has recently been shown to play a role in splicing of neuronal mRNA. We previously showed that ZCCHC17 protein declines in Alzheimer's disease (AD) brain tissue before there is significant gliosis and neuronal loss, that ZCCHC17 loss partially replicates observed splicing abnormalities in AD brain tissue, and that maintenance of ZCCHC17 levels is predicted to support cognitive resilience in AD. Here, we assessed the functional consequences of reduced ZCCHC17 expression in primary cortical neuronal cultures using siRNA knockdown. Consistent with its previously identified role in synaptic gene expression, loss of ZCCHC17 led to loss of synaptic protein expression. Patch recording of neurons shows that ZCCHC17 loss significantly disrupted the excitation/inhibition balance of neurotransmission, and favored excitatory-dominant synaptic activity as measured by an increase in spontaneous excitatory post synaptic currents and action potential firing rate, and a decrease in spontaneous inhibitory post synaptic currents. These findings are consistent with the hyperexcitable phenotype seen in AD animal models and in patients. We are the first to assess the functional consequences of ZCCHC17 knockdown in neurons and conclude that ZCCHC17 loss partially phenocopies AD-related loss of synaptic proteins and hyperexcitability.
Collapse
Affiliation(s)
- Giuseppe P Cortese
- College of Arts, Sciences, and Education, Program in Biology, Montana State University Northern, Havre, Montana, USA
| | - Anne Marie W Bartosch
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Yelizaveta Gribkova
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Tiffany G Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Elentina K Argyrousi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Sharanya Sivakumar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Christopher Cardona
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Andrew F Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
13
|
Gunawan C, Fleming C, Irga PJ, Jien Wong R, Amal R, Torpy FR, Mojtaba Golzan S, McGrath KC. Neurodegenerative effects of air pollutant Particles: Biological mechanisms implicated for Early-Onset Alzheimer's disease. ENVIRONMENT INTERNATIONAL 2024; 185:108512. [PMID: 38412566 DOI: 10.1016/j.envint.2024.108512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Sporadic Alzheimer's disease (AD) occurs in 99% of all cases and can be influenced by air pollution such as diesel emissions and more recently, an iron oxide particle, magnetite, detected in the brains of AD patients. However, a mechanistic link between air pollutants and AD development remains elusive. AIM To study the development of AD-relevant pathological effects induced by air pollutant particle exposures and their mechanistic links, in wild-type and AD-predisposed models. METHODS C57BL/6 (n = 37) and APP/PS1 transgenic (n = 38) mice (age 13 weeks) were exposed to model pollutant iron-based particle (Fe0-Fe3O4, dTEM = 493 ± 133 nm), hydrocarbon-based diesel combustion particle (43 ± 9 nm) and magnetite (Fe3O4, 153 ± 43 nm) particles (66 µg/20 µL/third day) for 4 months, and were assessed for behavioural changes, neuronal cell loss, amyloid-beta (Aβ) plaque, immune response and oxidative stress-biomarkers. Neuroblastoma SHSY5Y (differentiated) cells were exposed to the particles (100 μg/ml) for 24 h, with assessments on immune response biomarkers and reactive oxygen species generation. RESULTS Pollutant particle-exposure led to increased anxiety and stress levels in wild-type mice and short-term memory impairment in AD-prone mice. Neuronal cell loss was shown in the hippocampal and somatosensory cortex, with increased detection of Aβ plaque, the latter only in the AD-predisposed mice, with the wild-type not genetically disposed to form the plaque. The particle exposures however, increased AD-relevant immune system responses, including inflammation, in both strains of mice. Exposures also stimulated oxidative stress, although only observed in wild-type mice. The in vitro studies complemented the immune response and oxidative stress observations. CONCLUSIONS This study provides insights into the mechanistic links between inflammation and oxidative stress to pollutant particle-induced AD pathologies, with magnetite apparently inducing the most pathological effects. No exacerbation of the effects was observed in the AD-predisposed model when compared to the wild-type, indicating a particle-induced neurodegeneration that is independent of disease state.
Collapse
Affiliation(s)
- Cindy Gunawan
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, Australia.
| | - Charlotte Fleming
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Peter J Irga
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Roong Jien Wong
- School of Chemical Engineering, University of New South Wales, Australia; Institute of Sustainability for Chemicals, Energy and Environment, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Rose Amal
- School of Chemical Engineering, University of New South Wales, Australia
| | - Fraser R Torpy
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - S Mojtaba Golzan
- Vision Science Group, Graduate School of Health, University of Technology Sydney, Sydney, Australia
| | - Kristine C McGrath
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
| |
Collapse
|
14
|
Tzavellas NP, Tsamis KI, Katsenos AP, Davri AS, Simos YV, Nikas IP, Bellos S, Lekkas P, Kanellos FS, Konitsiotis S, Labrakakis C, Vezyraki P, Peschos D. Firing Alterations of Neurons in Alzheimer's Disease: Are They Merely a Consequence of Pathogenesis or a Pivotal Component of Disease Progression? Cells 2024; 13:434. [PMID: 38474398 PMCID: PMC10930991 DOI: 10.3390/cells13050434] [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: 01/14/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, yet its underlying causes remain elusive. The conventional perspective on disease pathogenesis attributes alterations in neuronal excitability to molecular changes resulting in synaptic dysfunction. Early hyperexcitability is succeeded by a progressive cessation of electrical activity in neurons, with amyloid beta (Aβ) oligomers and tau protein hyperphosphorylation identified as the initial events leading to hyperactivity. In addition to these key proteins, voltage-gated sodium and potassium channels play a decisive role in the altered electrical properties of neurons in AD. Impaired synaptic function and reduced neuronal plasticity contribute to a vicious cycle, resulting in a reduction in the number of synapses and synaptic proteins, impacting their transportation inside the neuron. An understanding of these neurophysiological alterations, combined with abnormalities in the morphology of brain cells, emerges as a crucial avenue for new treatment investigations. This review aims to delve into the detailed exploration of electrical neuronal alterations observed in different AD models affecting single neurons and neuronal networks.
Collapse
Affiliation(s)
- Nikolaos P. Tzavellas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Konstantinos I. Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Andreas P. Katsenos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Athena S. Davri
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Yannis V. Simos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Ilias P. Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Stefanos Bellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Panagiotis Lekkas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Foivos S. Kanellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Spyridon Konitsiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Charalampos Labrakakis
- Department of Biological Applications and Technology, University of Ioannina, 451 10 Ioannina, Greece
| | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| |
Collapse
|
15
|
Martins IM, Lima A, de Graaff W, Cristóvão JS, Brosens N, Aronica E, Kluskens LD, Gomes CM, Azeredo J, Kessels HW. M13 phage grafted with peptide motifs as a tool to detect amyloid-β oligomers in brain tissue. Commun Biol 2024; 7:134. [PMID: 38280942 PMCID: PMC10821927 DOI: 10.1038/s42003-024-05806-5] [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: 03/27/2023] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
Oligomeric clusters of amyloid-β (Aβ) are one of the major biomarkers for Alzheimer's disease (AD). However, proficient methods to detect Aβ-oligomers in brain tissue are lacking. Here we show that synthetic M13 bacteriophages displaying Aβ-derived peptides on their surface preferentially interact with Aβ-oligomers. When exposed to brain tissue isolated from APP/PS1-transgenic mice, these bacteriophages detect small-sized Aβ-aggregates in hippocampus at an early age, prior to the occurrence of Aβ-plaques. Similarly, the bacteriophages reveal the presence of such small Aβ-aggregates in post-mortem hippocampus tissue of AD-patients. These results advocate bacteriophages displaying Aβ-peptides as a convenient and low-cost tool to identify Aβ-oligomers in post-mortem brain tissue of AD-model mice and AD-patients.
Collapse
Affiliation(s)
- Ivone M Martins
- CEB- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal.
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal.
- Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Alexandre Lima
- CEB- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Wim de Graaff
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Joana S Cristóvão
- Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Niek Brosens
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Leon D Kluskens
- CEB- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Cláudio M Gomes
- Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Azeredo
- CEB- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Helmut W Kessels
- Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Bartosch AMW, Youth EHH, Hansen S, Wu Y, Buchanan HM, Kaufman ME, Xiao H, Koo SY, Ashok A, Sivakumar S, Soni RK, Dumitrescu LC, Lam TG, Ropri AS, Lee AJ, Klein HU, Vardarajan BN, Bennett DA, Young-Pearse TL, De Jager PL, Hohman TJ, Sproul AA, Teich AF. ZCCHC17 Modulates Neuronal RNA Splicing and Supports Cognitive Resilience in Alzheimer's Disease. J Neurosci 2024; 44:e2324222023. [PMID: 38050142 PMCID: PMC10860597 DOI: 10.1523/jneurosci.2324-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 09/22/2023] [Accepted: 11/07/2023] [Indexed: 12/06/2023] Open
Abstract
ZCCHC17 is a putative master regulator of synaptic gene dysfunction in Alzheimer's disease (AD), and ZCCHC17 protein declines early in AD brain tissue, before significant gliosis or neuronal loss. Here, we investigate the function of ZCCHC17 and its role in AD pathogenesis using data from human autopsy tissue (consisting of males and females) and female human cell lines. Co-immunoprecipitation (co-IP) of ZCCHC17 followed by mass spectrometry analysis in human iPSC-derived neurons reveals that ZCCHC17's binding partners are enriched for RNA-splicing proteins. ZCCHC17 knockdown results in widespread RNA-splicing changes that significantly overlap with splicing changes found in AD brain tissue, with synaptic genes commonly affected. ZCCHC17 expression correlates with cognitive resilience in AD patients, and we uncover an APOE4-dependent negative correlation of ZCCHC17 expression with tangle burden. Furthermore, a majority of ZCCHC17 interactors also co-IP with known tau interactors, and we find a significant overlap between alternatively spliced genes in ZCCHC17 knockdown and tau overexpression neurons. These results demonstrate ZCCHC17's role in neuronal RNA processing and its interaction with pathology and cognitive resilience in AD, and suggest that the maintenance of ZCCHC17 function may be a therapeutic strategy for preserving cognitive function in the setting of AD pathology.
Collapse
Affiliation(s)
- Anne Marie W Bartosch
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Elliot H H Youth
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Shania Hansen
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Yiyang Wu
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Heather M Buchanan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Maria E Kaufman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - So Yeon Koo
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Archana Ashok
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Sharanya Sivakumar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, New York 10032
| | - Logan C Dumitrescu
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Tiffany G Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Ali S Ropri
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Annie J Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
- Department of Neurology, Center for Translational & Computational Neuroimmunology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, New York 10032
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
- Department of Neurology, Center for Translational & Computational Neuroimmunology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, New York 10032
| | - Badri N Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
- Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, New York 10032
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Tracy L Young-Pearse
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138
| | - Philip L De Jager
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
- Department of Neurology, Center for Translational & Computational Neuroimmunology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, New York 10032
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Andrew A Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
| | - Andrew F Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York 10032
- Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, New York 10032
| |
Collapse
|
17
|
Tracy GC, Huang KY, Hong YT, Ding S, Noblet HA, Lim KH, Kim EC, Chung HJ, Kong H. Intracerebral Nanoparticle Transport Facilitated by Alzheimer Pathology and Age. NANO LETTERS 2023; 23:10971-10982. [PMID: 37991895 PMCID: PMC11404402 DOI: 10.1021/acs.nanolett.3c03222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Nanoparticles have emerged as potential transporters of drugs targeting Alzheimer's disease (AD), but their design should consider the blood-brain barrier (BBB) integrity and neuroinflammation of the AD brain. This study presents that aging is a significant factor for the brain localization and retention of nanoparticles, which we engineered to bind with reactive astrocytes and activated microglia. We assembled 200 nm-diameter particles using a block copolymer of poly(lactic-co-glycolic acid) (PLGA) and CD44-binding hyaluronic acid (HA). The resulting PLGA-b-HA nanoparticles displayed increased binding to CD44-expressing reactive astrocytes and activated microglia. Upon intravascular injection, nanoparticles were localized to the hippocampi of both APP/PS1 AD model mice and their control littermates at 13-16 months of age due to enhanced transvascular transport through the leaky BBB. No particles were found in the hippocampi of young adult mice. These findings demonstrate the brain localization of nanoparticles due to aging-induced BBB breakdown regardless of AD pathology.
Collapse
Affiliation(s)
- Gregory C. Tracy
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kai-Yu Huang
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yu-Tong Hong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shengzhe Ding
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hayden A. Noblet
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ki H. Lim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Eung Chang Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul 02841, South Korea
| |
Collapse
|
18
|
Wang X, Xie J, Tan L, Lu Y, Shen N, Li J, Hu H, Li H, Li X, Cheng L. N6-methyladenosine-modified circRIMS2 mediates synaptic and memory impairments by activating GluN2B ubiquitination in Alzheimer's disease. Transl Neurodegener 2023; 12:53. [PMID: 38012808 PMCID: PMC10683276 DOI: 10.1186/s40035-023-00386-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Synaptic degeneration occurs in the early stage of Alzheimer's disease (AD) before devastating symptoms, strongly correlated with cognitive decline. Circular RNAs (circRNAs) are abundantly enriched in neural tissues, and aberrant expression of circRNAs precedes AD symptoms, significantly correlated with clinical dementia severity. However, the direct relationship between circRNA dysregulation and synaptic impairment in the early stage of AD remains poorly understood. METHODS Hippocampal whole-transcriptome sequencing was performed to identify dysregulated circRNAs and miRNAs in 4-month-old wild-type and APP/PS1 mice. RNA antisense purification and mass spectrometry were utilized to unveil interactions between circRIMS2 and methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit (METTL3). The roles of circRIMS2/miR-3968 in synaptic targeting of UBE2K-mediated ubiquitination of GluN2B subunit of NMDA receptor were evaluated via numerous lentiviruses followed by morphological staining, co-immunoprecipitation and behavioral testing. Further, a membrane-permeable peptide was used to block the ubiquitination of K1082 on GluN2B in AD mice. RESULTS circRIMS2 was significantly upregulated in 4-month-old APP/PS1 mice, which was mediated by METTL3-dependent N6-methyladenosine (m6A) modification. Overexpression of circRIMS2 led to synaptic and memory impairments in 4-month-old C57BL/6 mice. MiR-3968/UBE2K was validated as the downstream of circRIMS2. Elevated UBE2K induced synaptic dysfunction of AD through ubiquitinating K1082 on GluN2B. Silencing METTL3 or blocking the ubiquitination of K1082 on GluN2B with a short membrane-permeable peptide remarkably rescued synaptic dysfunction in AD mice. CONCLUSIONS In conclusion, our study demonstrated that m6A-modified circRIMS2 mediates the synaptic and memory impairments in AD by activating the UBE2K-dependent ubiquitination and degradation of GluN2B via sponging miR-3968, providing novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jiazhao Xie
- Departments of Pathophysiology, Guangxi Medical University, Nanning, 530021, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu Tan
- Hepatic Biliary Pancreatic Surgery Department, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Na Shen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiaoyuan Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui Hu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huijun Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
19
|
Aurelian S, Ciobanu A, Cărare R, Stoica SI, Anghelescu A, Ciobanu V, Onose G, Munteanu C, Popescu C, Andone I, Spînu A, Firan C, Cazacu IS, Trandafir AI, Băilă M, Postoiu RL, Zamfirescu A. Topical Cellular/Tissue and Molecular Aspects Regarding Nonpharmacological Interventions in Alzheimer's Disease-A Systematic Review. Int J Mol Sci 2023; 24:16533. [PMID: 38003723 PMCID: PMC10671501 DOI: 10.3390/ijms242216533] [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: 09/22/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
One of the most complex and challenging developments at the beginning of the third millennium is the alarming increase in demographic aging, mainly-but not exclusively-affecting developed countries. This reality results in one of the harsh medical, social, and economic consequences: the continuously increasing number of people with dementia, including Alzheimer's disease (AD), which accounts for up to 80% of all such types of pathology. Its large and progressive disabling potential, which eventually leads to death, therefore represents an important public health matter, especially because there is no known cure for this disease. Consequently, periodic reappraisals of different therapeutic possibilities are necessary. For this purpose, we conducted this systematic literature review investigating nonpharmacological interventions for AD, including their currently known cellular and molecular action bases. This endeavor was based on the PRISMA method, by which we selected 116 eligible articles published during the last year. Because of the unfortunate lack of effective treatments for AD, it is necessary to enhance efforts toward identifying and improving various therapeutic and rehabilitative approaches, as well as related prophylactic measures.
Collapse
Affiliation(s)
- Sorina Aurelian
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| | - Adela Ciobanu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Roxana Cărare
- Faculty of Medicine, University of Southampton, Southampton SO16 7NS, UK;
| | - Simona-Isabelle Stoica
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Aurelian Anghelescu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Constantin Munteanu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Cristina Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ioana Andone
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Aura Spînu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Carmen Firan
- NeuroRehabilitation Compartment, The Physical and Rehabilitation Medicine & Balneology Clinic Division, Teaching Emergency Hospital of the Ilfov County, 022104 Bucharest, Romania;
| | - Ioana Simona Cazacu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea-Iulia Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Mihai Băilă
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ruxandra-Luciana Postoiu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea Zamfirescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| |
Collapse
|
20
|
Zhang XL, Hollander CM, Khan MY, D'silva M, Ma H, Yang X, Bai R, Keeter CK, Galkina EV, Nadler JL, Stanton PK. Myeloid cell deficiency of the inflammatory transcription factor Stat4 protects long-term synaptic plasticity from the effects of a high-fat, high-cholesterol diet. Commun Biol 2023; 6:967. [PMID: 37783748 PMCID: PMC10545833 DOI: 10.1038/s42003-023-05304-0] [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: 07/04/2022] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer's and Parkinson's. The cytokine interleukin-12 activates signal transducer and activator of transcription 4 (Stat4), and consumption of a high-fat, high-cholesterol diet (HFD-C) and Stat4 activity are associated with inflammation, atherosclerosis, and a diabetic metabolic phenotype. In studies of in vitro hippocampal slices from control Stat4fl/flLdlr-/- mice fed a HFD-C diabetogenic diet, we show that Schaffer collateral-CA1 synapses exhibited larger reductions in activity-dependent, long-term potentiation (LTP) of synaptic transmission, compared to mice fed a standard diet. Glucose tolerance and insulin sensitivity shifts produced by HFD-C diet were reduced in Stat4ΔLysMLdlr-/- mice compared to Stat4fl/flLdlr-/- controls. Stat4ΔLysMLdlr-/- mice, which lack Stat4 under control of the LysMCre promoter, were resistant to HFD-C induced impairments in LTP. In contrast, Schaffer collateral-CA1 synapses in Stat4ΔLysMLdlr-/- mice fed the HFD-C diet showed larger LTP than control Stat4fl/flLdlr-/- mice. Expression of a number of neuroinflammatory and synaptic plasticity genes was reduced by HFD-C diet in control mice, and less affected by HFD-C diet in Stat4ΔLysMLdlr-/- mice. These data suggest that suppression of Stat4 activation may protect against effects of Western diet on cognition, type 2 diabetes, and reduce risk of Alzheimer's disease and other neurodegenerative disorders associated with neuroinflammation.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Callie M Hollander
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Mohammad Yasir Khan
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Melinee D'silva
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Haoqin Ma
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Xinyuan Yang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Robin Bai
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Coles K Keeter
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Jerry L Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
- ACOS-Research VA Northern California Health Care System, Sacramento, CA, 95655, USA
| | - Patric K Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
21
|
Abdelmoaty MM, Yeapuri P, Machhi J, Lu Y, Namminga KL, Kadry R, Lu E, Bhattarai S, Mosley RL, Gendelman HE. Immune senescence in aged APP/PS1 mice. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:317-330. [PMID: 38023614 PMCID: PMC10659760 DOI: 10.1515/nipt-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023]
Abstract
Objectives To evaluate the linkage between age and deficits in innate and adaptive immunity which heralds both Alzheimer's disease (AD) onset and progression. The pathobiological events which underlie and tie these outcomes remain not fully understood. Methods To investigate age-dependent immunity in AD, we evaluated innate and adaptive immunity in coordinate studies of regulatory T cell (Treg) function, T cell frequencies, and microglial integrity. These were assessed in blood, peripheral lymphoid tissues, and the hippocampus of transgenic (Tg) amyloid precursor protein/presenilin 1 (APP/PS1) against non-Tg mice. Additionally, immune arrays of hippocampal tissue were performed at 4, 6, 12, and 20 months of age. Results APP/PS1 mice showed progressive impairment of Treg immunosuppressive function with age. There was partial restoration of Treg function in 20-month-old mice. Ingenuity pathway analyses of hippocampal tissues were enriched in inflammatory, oxidative, and cellular activation pathways that paralleled advancing age and AD-pathobiology. Operative genes in those pathways included, but were not limited to triggering receptor on myeloid cells 1 (TREM1), T helper type 1 (Th1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Interleukin-17 (IL-17), nitric oxide, acute phase, and T cell receptor signaling pathways were also perturbed. Significant inflammation was observed at 6- and 12-months. However, at 20-months, age associated partial restoration of Treg function reduced inflammatory phenotype. Conclusions Impaired Treg function, inflammation and oxidative stress were associated with AD pathology. Age associated partial restoration of Treg function in old mice reduced the hippocampal inflammatory phenotype. Restoring Treg suppressive function can be a therapeutic modality for AD.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
22
|
Deng LJ, Wu D, Yang XF, Li T. miR-146a-5p Modulates Adult Hippocampal Neurogenesis Deficits Through Klf4/p-Stat3 Signaling in APP/PS1 Mice. Neuroscience 2023; 526:314-325. [PMID: 37321367 DOI: 10.1016/j.neuroscience.2023.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and currently, no effective treatment strategies exist for this condition. MicroRNAs (miRNAs) have emerged as promising therapeutic targets of AD. Previous studies have highlighted the significant role of miR-146a-5p in regulating adult hippocampal neurogenesis (AHN). Here, we aimed to investigate whether miR-146a-5p plays a role in the mechanisms of AD. We employed quantitative real-time PCR (qRT-PCR) to assess the expression of miR-146a-5p. Additionally, we examined the expression of Krüppel-like factor 4 (Klf4), Signal transducer and activator of transcription 3 (Stat3), and phosphorylated Stat3 (p-Stat3) using western blot analysis. Furthermore, we validated the interaction between miR-146a-5p and Klf4 using a dual-luciferase reporter assay. Immunofluorescence staining was employed to evaluate AHN. And Contextual fear conditioning discrimination learning (CFC-DL) experiment was used to detect pattern separation. Our findings in the hippocampus of APP/PS1 mice revealed upregulated levels of miR-146a-5p and p-Stat3, while Klf4 levels were downregulated. Interestingly, both miR-146a-5p antagomir and p-Stat3 inhibitor obviously rescued neurogenesis and pattern separation in APP/PS1 mice. Moreover, application of miR-146a-5p agomir reversed the protective effects of Klf4 upregulation. These findings open new avenues for protection against AD through the modulation of neurogenesis and cognitive decline via the miR-146a-5p/Klf4/p-Stat3 pathway.
Collapse
Affiliation(s)
- Li-Jun Deng
- Department of Neurosurgery, The Affiliated Hospital of Jianghan University, Wuhan 430022, PR China
| | - Dan Wu
- Department of Neurosurgery, The Affiliated Hospital of Jianghan University, Wuhan 430022, PR China
| | - Xiao-Fan Yang
- Department of Hand Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Tao Li
- Department of Hand Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| |
Collapse
|
23
|
Huang YR, Xie XX, Yang J, Sun XY, Niu XY, Yang CG, Li LJ, Zhang L, Wang D, Liu CY, Hou SJ, Jiang CY, Xu YM, Liu RT. ArhGAP11A mediates amyloid-β generation and neuropathology in an Alzheimer's disease-like mouse model. Cell Rep 2023; 42:112624. [PMID: 37302068 DOI: 10.1016/j.celrep.2023.112624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Amyloid-β (Aβ) plays an important role in the neuropathology of Alzheimer's disease (AD), but some factors promoting Aβ generation and Aβ oligomer (Aβo) neurotoxicity remain unclear. We here find that the levels of ArhGAP11A, a Ras homology GTPase-activating protein, significantly increase in patients with AD and amyloid precursor protein (APP)/presenilin-1 (PS1) mice. Reducing the ArhGAP11A level in neurons not only inhibits Aβ generation by decreasing the expression of APP, PS1, and β-secretase (BACE1) through the RhoA/ROCK/Erk signaling pathway but also reduces Aβo neurotoxicity by decreasing the expressions of apoptosis-related p53 target genes. In APP/PS1 mice, specific reduction of the ArhGAP11A level in neurons significantly reduces Aβ production and plaque deposition and ameliorates neuronal damage, neuroinflammation, and cognitive deficits. Moreover, Aβos enhance ArhGAP11A expression in neurons by activating E2F1, which thus forms a deleterious cycle. Our results demonstrate that ArhGAP11A may be involved in AD pathogenesis and that decreasing ArhGAP11A expression may be a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Ya-Ru Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi-Xiu Xie
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Yang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Henan Medical Key Laboratory of Neurogenetic and Neurodegenerative Disease, Zhengzhou 450052, Henan, China
| | - Xiao-Ying Sun
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Yun Niu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Ningxia University, Yinchuan 750021, Ningxia, China
| | - Cheng-Gang Yang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China; Department of Research and Development, Gu'an Bojian Bio-Technology Co., Ltd., Langfang 065000, Hebei, China
| | - Ling-Jie Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lun Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Dan Wang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China
| | - Chun-Yu Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Shandong Agricultural University, Tai'an 271000, Shandong, China
| | - Sheng-Jie Hou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen-Yang Jiang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Key Laboratory of Cerebrovascular Disease of Henan Province, Zhengzhou 450052, Henan, China.
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
24
|
Sha S, Chaigneau T, Krantic S. Pre-symptomatic synaptic dysfunction and longitudinal decay of hippocampal synaptic function in APPPS1 mouse model of Alzheimer's disease is sex-independent. Brain Res Bull 2023; 198:36-49. [PMID: 37080395 DOI: 10.1016/j.brainresbull.2023.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/19/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Alzheimer's disease (AD) is an incurable, age-related and progressive neurodegenerative disease characterized by cognitive impairments. Deficits in synaptic plasticity were reported in various models of AD-like pathology and are considered as an early contributing factor of cognitive impairment. However, the majority of previous studies were focused on overt, symptomatic stages of pathology and assessed long-term potentiation (LTP), whereas long-term depression (LTD) was much less investigated and the precise nature of its involvement remains poorly defined. To better understand the earliest synaptic dysfunctions along the pre-symptomatic stage of AD-like pathology, we performed a detailed analysis of underlying mechanisms and quantified basal synaptic activity, presynaptic release probability, and synaptic plasticity such as post-tetanic potentiation (PTP), as well as LTP and LTD. These parameters were studied in APPPS1 mouse model at two time points (early- and mid-) along the pre-symptomatic stage, which were compared with alterations monitored at two later time-points, i.e. the onset of cognitive deficits and the overt stage of full-blown pathology. Because sex is known to be an instrumental biological parameter in AD pathophysiology, all alterations were assessed in both males and females. Our data show that, as compared to wild-type (WT) littermates, initial neuronal hyperexcitability, seen at early pre-symptomatic stage shifts subsequently towards hypoexcitability at mid-pre-symptomatic stage and remains impaired at advanced stages. The pre-symptomatic changes also involve increased synaptic plasticity as assessed by paired-pulse facilitation (PPF), which returns to basal level at the onset of pathology and remains stable afterwards. Synaptic plasticity is impaired by mid-pre-symptomatic stage and manifests as lowered LTP and absence of LTD induction, the latter being reported here for the first time. Observed LTP and LTD impairments both persist in older APPPS1 mice. Remarkably, none of the observed differences was gender-dependent. Altogether, our data evidence that major impairments in basal synaptic efficacy and plasticity are detectable already during mid-pre-symptomatic stage of AD-like pathogenesis and likely involve hyperexcitability as the underlying mechanism. Our study also uncovers synaptic alterations that may become critical read-outs for testing the efficiency of novel, pre-symptomatic stage-targeted therapies for AD.
Collapse
Affiliation(s)
- Sha Sha
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France; Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Thomas Chaigneau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Slavica Krantic
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, F-75012 Paris, France.
| |
Collapse
|
25
|
Lopes CR, Silva JS, Santos J, Rodrigues MS, Madeira D, Oliveira A, Moreira-de-Sá A, Lourenço VS, Gonçalves FQ, Silva HB, Simões AP, Rolo AP, Canas PM, Tomé ÂR, Palmeira CM, Lopes JP, Cunha RA, Agostinho P, Ferreira SG. Downregulation of Sirtuin 1 Does Not Account for the Impaired Long-Term Potentiation in the Prefrontal Cortex of Female APPswe/PS1dE9 Mice Modelling Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24086968. [PMID: 37108131 PMCID: PMC10139121 DOI: 10.3390/ijms24086968] [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: 03/17/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD), which predominantly affects women, involves at its onset a metabolic deregulation associated with a synaptic failure. Here, we performed a behavioral, neurophysiological and neurochemical characterization of 9-month-old female APPswe/PS1dE9 (APP/PS1) mice as a model of early AD. These animals showed learning and memory deficits in the Morris water maze, increased thigmotaxis and anxiety-like behavior and showed signs of fear generalization. Long-term potentiation (LTP) was decreased in the prefrontal cortex (PFC), but not in the CA1 hippocampus or amygdala. This was associated with a decreased density of sirtuin-1 in cerebrocortical synaptosomes and a decreased density of sirtuin-1 and sestrin-2 in total cerebrocortical extracts, without alterations of sirtuin-3 levels or of synaptic markers (syntaxin, synaptophysin, SNAP25, PSD95). However, activation of sirtuin-1 did not affect or recover PFC-LTP deficit in APP/PS1 female mice; instead, inhibition of sirtuin-1 increased PFC-LTP magnitude. It is concluded that mood and memory dysfunction in 9-month-old female APP/PS1 mice is associated with a parallel decrease in synaptic plasticity and in synaptic sirtuin-1 levels in the prefrontal cortex, although sirtiun1 activation failed to restore abnormal cortical plasticity.
Collapse
Affiliation(s)
- Cátia R Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Joana S Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Joana Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Matilde S Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniela Madeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Andreia Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Moreira-de-Sá
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Vanessa S Lourenço
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Q Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Henrique B Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Patrícia Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Anabela P Rolo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Paula M Canas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ângelo R Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3004-531 Coimbra, Portugal
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula Agostinho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Samira G Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
26
|
Lanza M, Cuzzocrea S, Oddo S, Esposito E, Casili G. The Role of miR-128 in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:6024. [PMID: 37046996 PMCID: PMC10093830 DOI: 10.3390/ijms24076024] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Several neurodegenerative disorders are characterized by the accumulation of misfolded proteins and are collectively known as proteinopathies. Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD) represent some of the most common neurodegenerative disorders whose steady increase in prevalence is having a major socio-economic impact on our society. Multiple laboratories have reported hundreds of changes in gene expression in selective brain regions of AD, PD, and HD brains. While the mechanisms underlying these changes remain an active area of investigation, alterations in the expression of noncoding RNAs, which are common in AD, PD, and HD, may account for some of the changes in gene expression in proteinopathies. In this review, we discuss the role of miR-128, which is highly expressed in mammalian brains, in AD, PD, and HD. We highlight how alterations in miR-128 may account, at least in part, for the gene expression changes associated with proteinopathies. Indeed, miR-128 is involved, among other things, in the regulation of neuronal plasticity, cytoskeletal organization, and neuronal death, events linked to various proteinopathies. For example, reducing the expression of miR-128 in a mouse model of AD ameliorates cognitive deficits and reduces neuropathology. Overall, the data in the literature suggest that targeting miR-128 might be beneficial to mitigate the behavioral phenotype associated with these diseases.
Collapse
Affiliation(s)
| | | | - Salvatore Oddo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | | |
Collapse
|
27
|
Bartosch AMW, Youth EHH, Hansen S, Kaufman ME, Xiao H, Koo SY, Ashok A, Sivakumar S, Soni RK, Dumitrescu LC, Lam TG, Ropri AS, Lee AJ, Klein HU, Vardarajan BN, Bennett DA, Young-Pearse TL, De Jager PL, Hohman TJ, Sproul AA, Teich AF. ZCCHC17 modulates neuronal RNA splicing and supports cognitive resilience in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533654. [PMID: 36993746 PMCID: PMC10055234 DOI: 10.1101/2023.03.21.533654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
ZCCHC17 is a putative master regulator of synaptic gene dysfunction in Alzheimer's Disease (AD), and ZCCHC17 protein declines early in AD brain tissue, before significant gliosis or neuronal loss. Here, we investigate the function of ZCCHC17 and its role in AD pathogenesis. Co-immunoprecipitation of ZCCHC17 followed by mass spectrometry analysis in human iPSC-derived neurons reveals that ZCCHC17's binding partners are enriched for RNA splicing proteins. ZCCHC17 knockdown results in widespread RNA splicing changes that significantly overlap with splicing changes found in AD brain tissue, with synaptic genes commonly affected. ZCCHC17 expression correlates with cognitive resilience in AD patients, and we uncover an APOE4 dependent negative correlation of ZCCHC17 expression with tangle burden. Furthermore, a majority of ZCCHC17 interactors also co-IP with known tau interactors, and we find significant overlap between alternatively spliced genes in ZCCHC17 knockdown and tau overexpression neurons. These results demonstrate ZCCHC17's role in neuronal RNA processing and its interaction with pathology and cognitive resilience in AD, and suggest that maintenance of ZCCHC17 function may be a therapeutic strategy for preserving cognitive function in the setting of AD pathology.
Collapse
Affiliation(s)
- Anne Marie W. Bartosch
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Elliot H. H. Youth
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Shania Hansen
- Vanderbilt Memory & Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Maria E. Kaufman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - So Yeon Koo
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Archana Ashok
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Sharanya Sivakumar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Rajesh K. Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, NY 10032
| | - Logan C. Dumitrescu
- Vanderbilt Memory & Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Tiffany G. Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Ali S. Ropri
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY 10032
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY 10032
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY 10032
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612
| | - Tracy L. Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY 10032
| | - Timothy J. Hohman
- Vanderbilt Memory & Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Andrew A. Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Andrew F. Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY 10032
| |
Collapse
|
28
|
The Neuroprotective Activities of the Novel Multi-Target Iron-Chelators in Models of Alzheimer's Disease, Amyotrophic Lateral Sclerosis and Aging. Cells 2023; 12:cells12050763. [PMID: 36899898 PMCID: PMC10001413 DOI: 10.3390/cells12050763] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/03/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
The concept of chelation therapy as a valuable therapeutic approach in neurological disorders led us to develop multi-target, non-toxic, lipophilic, brain-permeable compounds with iron chelation and anti-apoptotic properties for neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), age-related dementia and amyotrophic lateral sclerosis (ALS). Herein, we reviewed our two most effective such compounds, M30 and HLA20, based on a multimodal drug design paradigm. The compounds have been tested for their mechanisms of action using animal and cellular models such as APP/PS1 AD transgenic (Tg) mice, G93A-SOD1 mutant ALS Tg mice, C57BL/6 mice, Neuroblastoma × Spinal Cord-34 (NSC-34) hybrid cells, a battery of behavior tests, and various immunohistochemical and biochemical techniques. These novel iron chelators exhibit neuroprotective activities by attenuating relevant neurodegenerative pathology, promoting positive behavior changes, and up-regulating neuroprotective signaling pathways. Taken together, these results suggest that our multifunctional iron-chelating compounds can upregulate several neuroprotective-adaptive mechanisms and pro-survival signaling pathways in the brain and might function as ideal drugs for neurodegenerative disorders, such as PD, AD, ALS, and aging-related cognitive decline, in which oxidative stress and iron-mediated toxicity and dysregulation of iron homeostasis have been implicated.
Collapse
|
29
|
Demuth H, Hosseini S, Düsedeau HP, Dunay IR, Korte M, Zagrebelsky M. Deletion of p75 NTR rescues the synaptic but not the inflammatory status in the brain of a mouse model for Alzheimer's disease. Front Mol Neurosci 2023; 16:1163087. [PMID: 37213691 PMCID: PMC10198655 DOI: 10.3389/fnmol.2023.1163087] [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: 02/10/2023] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Alzheimer's disease (AD), is characterized by a gradual cognitive decline associated with the accumulation of Amyloid beta (Aβ)-oligomers, progressive neuronal degeneration and chronic neuroinflammation. Among the receptors shown to bind and possibly transduce the toxic effects of Aβ-oligomers is the p75 neurotrophin receptor (p75NTR). Interestingly, p75NTR mediates several crucial processes in the nervous system, including neuronal survival and apoptosis, maintenance of the neuronal architecture, and plasticity. Furthermore, p75NTR is also expressed in microglia, the resident immune cells of the brain, where it is markedly increased under pathological conditions. These observations indicate p75NTR as a potential candidate for mediating Aβ-induced toxic effects at the interface between the nervous and the immune system, thereby potentially participating in the crosstalk between these two systems. Methods Here we used APP/PS1 transgenic mice (APP/PS1tg) and compared the Aβ-induced alterations in neuronal function, chronic inflammation as well as their cognitive consequences between 10 months old APP/PS1tg and APP/PS1tg x p75NTRexonIV knockout mice. Results Electrophysiological recordings show that a loss of p75NTR rescues the impairment in long-term potentiation at the Schaffer collaterals in the hippocampus of APP/PS1tg mice. Interestingly, however loss of p75NTR does not influence the severity of neuroinflammation, microglia activation or the decline in spatial learning and memory processes observed in APP/PS1tg mice. Conclusion Together these results indicate that while a deletion of p75NTR rescues the synaptic defect and the impairment in synaptic plasticity, it does not affect the progression of the neuroinflammation and the cognitive decline in a mouse model for AD.
Collapse
Affiliation(s)
- Hendrik Demuth
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Shirin Hosseini
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Henning Peter Düsedeau
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von- Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von- Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, Braunschweig, Germany
- *Correspondence: Marta Zagrebelsky,
| |
Collapse
|
30
|
Brosens N, Samouil D, Stolker S, Katsika EV, Weggen S, Lucassen PJ, Krugers HJ. Early Life Stress Enhances Cognitive Decline and Alters Synapse Function and Interneuron Numbers in Young Male APP/PS1 Mice. J Alzheimers Dis 2023; 96:1097-1113. [PMID: 37980670 PMCID: PMC10741326 DOI: 10.3233/jad-230727] [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] [Accepted: 09/12/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Exposure to stress early in life increases the susceptibility to Alzheimer's disease (AD) pathology in aged AD mouse models. So far, the underlying mechanisms have remained elusive. OBJECTIVE To investigate 1) effects of early life stress (ELS) on early functional signs that precede the advanced neuropathological changes, and 2) correlate synaptosomal protein content with cognition to identify neural correlates of AD. METHODS APPswe/PS1dE9 mice and littermates were subjected to ELS by housing dams and pups with limited bedding and nesting material from postnatal days 2-9. At 3 months of age, an age where no cognitive loss or amyloid-β (Aβ) pathology is typically reported in this model, we assessed hippocampal Aβ pathology, synaptic strength and synapse composition and interneuron populations. Moreover, cognitive flexibility was assessed and correlated with synaptosomal protein content. RESULTS While ELS did not affect Aβ pathology, it increased synaptic strength and decreased the number of calretinin+ interneurons in the hippocampal dentate gyrus. Both genotype and condition further affected the level of postsynaptic glutamatergic protein content. Finally, APP/PS1 mice were significantly impaired in cognitive flexibility at 3 months of age, and ELS exacerbated this impairment, but only at relatively high learning criteria. CONCLUSIONS ELS reduced cognitive flexibility in young APP/PS1 mice and altered markers for synapse and network function. These findings at an early disease stage provide novel insights in AD etiology and in how ELS could increase AD susceptibility.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Dimitris Samouil
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Sabine Stolker
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Liu YS, Zhao HF, Li Q, Cui HW, Huang GD. Research Progress on the Etiology and Pathogenesis of Alzheimer's Disease from the Perspective of Chronic Stress. Aging Dis 2022:AD.2022.1211. [PMID: 37163426 PMCID: PMC10389837 DOI: 10.14336/ad.2022.1211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/12/2022] [Indexed: 05/12/2023] Open
Abstract
Due to its extremely complex pathogenesis, no effective drugs to prevent, delay progression, or cure Alzheimer's disease (AD) exist at present. The main pathological features of AD are senile plaques composed of β-amyloid, neurofibrillary tangles formed by hyperphosphorylation of the tau protein, and degeneration or loss of neurons in the brain. Many risk factors associated with the onset of AD, including gene mutations, aging, traumatic brain injury, endocrine and cardiovascular diseases, education level, and obesity. Growing evidence points to chronic stress as one of the major risk factors for AD, as it can promote the onset and development of AD-related pathologies via a mechanism that is not well known. The use of murine stress models, including restraint, social isolation, noise, and unpredictable stress, has contributed to improving our understanding of the relationship between chronic stress and AD. This review summarizes the evidence derived from murine models on the pathological features associated with AD and the related molecular mechanisms induced by chronic stress. These results not only provide a retrospective interpretation for understanding the pathogenesis of AD, but also provide a window of opportunity for more effective preventive and identifying therapeutic strategies for stress-induced AD.
Collapse
Affiliation(s)
- Yun-Sheng Liu
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Hua-Fu Zhao
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Qian Li
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Han-Wei Cui
- The Central Laboratory, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Central Laboratory, Shenzhen Samii Medical Center, Shenzhen, China
| | - Guo-Dong Huang
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
32
|
Promoted CD4 + T cell-derived IFN-γ/IL-10 by photobiomodulation therapy modulates neurogenesis to ameliorate cognitive deficits in APP/PS1 and 3xTg-AD mice. J Neuroinflammation 2022; 19:253. [PMID: 36217178 PMCID: PMC9549637 DOI: 10.1186/s12974-022-02617-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background The immune system has been implicated in synaptic plasticity, inflammation, and the progression of Alzheimer's disease (AD). However, there were few studies on improving the niche microenvironment of neural stem cells (NSCs) in the brain of AD to promote adult hippocampal neurogenesis (AHN) by regulating the function of non-parenchymal immune cells. Methods The lymph nodes of amyloid precursor protein/presenilin 1 (APP/PS1) and 3xTg (APP/PS1/tau) mouse models of AD were treated with photobiomodulation therapy (PBMT) for 10 J/cm2 per day for 1 month (10 min for each day), T lymphocytes isolated from these two AD models were treated with PBMT for 2 J/cm2 (5 min for each time). The NSCs isolated from hippocampus of these two AD models at E14, and the cells were co-cultivated with PBMT-treated T lymphocyte conditioned medium for NSCs differentiation. Results Our results showed that PBMT treatment could promote AHN and reverse cognitive deficits in AD mouse model. The expression of interferon-γ (IFN-γ) and interleukin-10 (IL-10) was upregulated in the brain of these two AD models after PBMT treated, which was induced by the activation of Janus kinase 2 (JAK2)-mediated signal transducer and activator of transcription 4 (STAT4)/STAT5 signaling pathway in CD4+ T cells. In addition, elevated CD4+ T cell levels and upregulated transforming growth factor-β1 (TGFβ1)/insulin-like growth factors-1 (IGF-1)/brain-derived neurotrophic factor (BDNF) protein expression levels were also detected in the brain. More importantly, co-cultivated the PBMT-treated T lymphocyte conditioned medium with NSCs derived from these two AD models was shown to promote NSCs differentiation, which was reflected in the upregulation of both neuronal class-III β-tubulin (Tuj1) and postsynaptic density protein 95 (PSD95), but the effects of PBMT was blocked by reactive oxygen species (ROS) scavenger or JAK2 inhibitor. Conclusion Our research suggests that PBMT exerts a beneficial neurogenesis modulatory effect through activating the JAK2/STAT4/STAT5 signaling pathway to promote the expression of IFN-γ/IL-10 in non-parenchymal CD4+ T cells, induction of improvement of brain microenvironmental conditions and alleviation of cognitive deficits in APP/PS1 and 3xTg-AD mouse models. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02617-5.
Collapse
|
33
|
Lopes van den Broek S, Sehlin D, Andersen JV, Aldana BI, Beschörner N, Nedergaard M, Knudsen GM, Syvänen S, Herth MM. The Alzheimer's disease 5xFAD mouse model is best suited to investigate pretargeted imaging approaches beyond the blood-brain barrier. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:1001722. [PMID: 39390994 PMCID: PMC11466232 DOI: 10.3389/fnume.2022.1001722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/08/2022] [Indexed: 10/12/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with an increasing prevalence. Currently, there is no ideal diagnostic molecular imaging agent for diagnosing AD. Antibodies (Abs) have been proposed to close this gap as they can bind selectively and with high affinity to amyloid β (Aβ)-one of the molecular hallmarks of AD. Abs can even be designed to selectively bind Aβ oligomers or isoforms, which are difficult to target with small imaging agents. Conventionally, Abs must be labeled with long-lived radionuclides which typically results in in high radiation burden to healthy tissue. Pretargeted imaging could solve this challenge as it allows for the use of short-lived radionuclides. To develop pretargeted imaging tools that can enter the brain, AD mouse models are useful as they allow testing of the imaging approach in a relevant animal model that could predict its clinical applicability. Several mouse models for AD have been developed with different characteristics. Commonly used models are: 5xFAD, APP/PS1 and tg-ArcSwe transgenic mice. In this study, we aimed to identify which of these models were best suited to investigate pretargeted imaging approaches beyond the blood brain barrier. We evaluated this by pretargeted autoradiography using the Aβ-targeting antibody 3D6 and an 111In-labeled Tz. Evaluation criteria were target-to-background ratios and accessibility. APP/PS1 mice showed Aβ accumulation in high and low binding brain regions and is as such less suitable for pretargeted purposes. 5xFAD and tg-ArcSwe mice showed similar uptake in high binding regions whereas low uptake in low binding regions and are better suited to evaluate pretargeted imaging approaches. 5xFAD mice are advantaged over tg-ArcSwe mice as pathology can be traced early (6 months compared to 18 months of age) and as 5xFAD mice are commercially available.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dag Sehlin
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Uppsala, Sweden
| | - Jens V. Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca I. Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschörner
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M. Knudsen
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Stina Syvänen
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Uppsala, Sweden
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine / PET, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
34
|
Huffels CFM, van Dijk RE, Karst H, Meye FJ, Hol EM, Middeldorp J. Systemic Injection of Aged Blood Plasma in Adult C57BL/6 Mice Induces Neurophysiological Impairments in the Hippocampal CA1. J Alzheimers Dis 2022; 89:283-297. [PMID: 35871343 DOI: 10.3233/jad-220337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Aging is characterized by systemic alterations and forms an important risk factor for Alzheimer's disease. Recently, it has been indicated that blood-borne factors present in the systemic milieu contribute to the aging process. Exposing young mice to aged blood plasma results in impaired neurogenesis and synaptic plasticity in the dentate gyrus, as well as impaired cognition. Vice versa, treating aged mice with young blood plasma rescues impairments associated with aging. OBJECTIVE Whether blood-borne factors are sufficient to drive impairments outside the dentate gyrus, how they impact neurophysiology, and how the functional outcome compares to impairments found in mouse models for AD is still unclear. METHODS Here, we treated adult mice with blood plasma from aged mice and assessed neurophysiological parameters in the hippocampal CA1. RESULTS Mice treated with aged blood plasma show significantly impaired levels of long-term potentiation (LTP), similar to those present in APP/PS1 mice. These impaired levels of LTP in plasma-treated mice are associated with alterations in basic properties of glutamatergic transmission and the enhanced activity of voltage-gated Ca2 + channels. CONCLUSION Together, the data presented in this study show that blood-borne factors are sufficient to drive neurophysiological impairments in the hippocampal CA1.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Roland E van Dijk
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
35
|
Wang Y, Yang J, Chen Q, Su J, Shi WJ, Zhang L, Xia C, Yan J. Rotor-Tuning Boron Dipyrromethenes for Dual-Functional Imaging of Aβ Oligomers and Viscosity. ACS APPLIED BIO MATERIALS 2022; 5:3049-3056. [PMID: 35671477 DOI: 10.1021/acsabm.2c00335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD), known as a common incurable and elderly neurodegenerative disease, has been widely explored for accurate detection of its biomarker (Aβ oligomers) for early diagnosis. Although great efforts have been made, it is still of great importance to develop fluorescence probes for Aβ oligomers with good selectivity and low background. Herein, starting from BODIPY493/503 (a commercial dye for neutral lipid droplets), which exhibited a small Stokes shift and no response toward Aβ peptides, two fluorescence probes 5MB-SZ and B-SZ with a benzothiazole rotor at the 2-position of the BODIPY core and a methyl or benzyl group at the meso position have been designed and synthesized, which exhibited excellent optical properties/stability and could successfully image β-amyloid fibrils and viscosity. Upon exposure to Aβ oligomers, the fluorescence intensity of 5MB-SZ was enhanced by 43.64-fold with the corresponding fluorescence quantum yields changing from 0.85% to 27.43%. Meanwhile, probe 5MB-SZ showed a highly sensitive viscosity response in both solutions and living cells. In vitro and in vivo experiments confirmed that probe 5MB-SZ exhibited an excellent capacity for imaging β-amyloid fibrils. Therefore, 5MB-SZ, as a rotor-tuning BODIPY analogue, could possibly serve as a highly potential and powerful fluorescence probe for early diagnosis of AD.
Collapse
Affiliation(s)
- Yuxuan Wang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Jinrong Yang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Qingxiu Chen
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai 519041, PR China
| | - Junyi Su
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai 519041, PR China
| | - Wen-Jing Shi
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, PR China
| | - Lei Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Chunli Xia
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai 519041, PR China
| | - Jinwu Yan
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| |
Collapse
|
36
|
Wu L, Jiang W, Zhao N, Wang F. Heparan sulfate from porcine mucosa promotes amyloid-beta clearance in APP/PS1 mice and alleviates Alzheimer's pathology. Carbohydr Polym 2022; 285:119205. [DOI: 10.1016/j.carbpol.2022.119205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/28/2022] [Indexed: 12/11/2022]
|
37
|
Ni X, Mori H. Complex Processes Underlying the Dynamic Changes of D-serine Levels in AD Brains. Curr Alzheimer Res 2022; 19:485-493. [PMID: 35346007 DOI: 10.2174/1567205019666220328123048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/02/2022] [Accepted: 02/10/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular β-amyloid (Aβ) plaques and cognitive impairments. D-Serine, produced by the enzyme serine racemase (SR) in the brain, functions as an endogenous co-agonist at the glycine-binding site of N-methyl-D-aspartate receptor (NMDAR), has been implicated in the pathophysiological progression of AD. OBJECTIVES Evidence regarding the understanding of the role and dynamic modulation of D-serine during AD progression remains controversial. This literature review aims to offer novel research directions for studying the functions and metabolisms of D-serine in AD brains. METHODS We searched PubMed, using D-serine/SR and AD as keywords. Studies related to NMDAR dysfunction, neuronal excitotoxicity, D-serine dynamic changes and inflammatory response were included. RESULTS This review primarily discusses: (i) Aβ oligomers' role in NMDAR dysregulation, and the subsequent synaptic dysfunction and neuronal damage in AD, (ii) D-serine's role in NMDAR-elicited excitotoxicity, and (iii) the involvement of D-serine and SR in AD-related inflammatory pathological progression. CONCLUSION We also presented supposed metabolism and dynamic changes of D-serine during AD progression and hypothesized that: (i) the possible modulation of D-serine levels or SR expression as an effective method of alleviating neurotoxicity during AD pathophysiological progression, and (ii) the dynamic changes of D-serine levels in AD brains possibly resulting from complex processes.
Collapse
Affiliation(s)
- Xiance Ni
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan
| | - Hisashi Mori
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan.,Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
38
|
Transferrin Receptor Binding BBB-Shuttle Facilitates Brain Delivery of Anti-Aβ-Affibodies. Pharm Res 2022; 39:1509-1521. [PMID: 35538266 PMCID: PMC9246779 DOI: 10.1007/s11095-022-03282-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/29/2022] [Indexed: 12/13/2022]
Abstract
Affibodies targeting amyloid-beta (Aβ) could potentially be used as therapeutic and diagnostic agents in Alzheimer's disease (AD). Affibodies display suitable characteristics for imaging applications such as high stability and a short biological half-life. The aim of this study was to explore brain delivery and retention of Aβ protofibril-targeted affibodies in wild-type (WT) and AD transgenic mice and to evaluate their potential as imaging agents. Two affibodies, Z5 and Z1, were fused with the blood-brain barrier (BBB) shuttle single-chain variable fragment scFv8D3. In vitro binding of 125I-labeled affibodies with and without scFv8D3 was evaluated by ELISA and autoradiography. Brain uptake and retention of the affibodies at 2 h and 24 h post injection was studied ex vivo in WT and transgenic (tg-Swe and tg-ArcSwe) mice. At 2 h post injection, [125I]I-Z5 and [125I]I-Z1 displayed brain concentrations of 0.37 ± 0.09% and 0.46 ± 0.08% ID/g brain, respectively. [125I]I-scFv8D3-Z5 and [125I]I-scFv8D3-Z1 showed increased brain concentrations of 0.53 ± 0.16% and 1.20 ± 0.35%ID/g brain. At 24 h post injection, brain retention of [125I]I-Z1 and [125I]I-Z5 was low, while [125I]I-scFv8D3-Z1 and [125I]I-scFv8D3-Z5 showed moderate brain retention, with a tendency towards higher retention of [125I]I-scFv8D3-Z5 in AD transgenic mice. Nuclear track emulsion autoradiography showed greater parenchymal distribution of [125I]I-scFv8D3-Z5 and [125I]I-scFv8D3-Z1 compared with the affibodies without scFv8D3, but could not confirm specific affibody accumulation around Aβ deposits. Affibody-scFv8D3 fusions displayed increased brain and parenchymal delivery compared with the non-fused affibodies. However, fast brain washout and a suboptimal balance between Aβ and mTfR1 affinity resulted in low intrabrain retention around Aβ deposits.
Collapse
|
39
|
Wang Y, Yang Y, Liu Y, Guo A, Zhang Y. Cognitive impairments in type 1 diabetes mellitus model mice are associated with synaptic protein disorders. Neurosci Lett 2022; 777:136587. [PMID: 35337951 DOI: 10.1016/j.neulet.2022.136587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
An association between type 1 diabetes mellitus (T1DM) and cognitive impairment was recently reported. However, the mechanisms by which T1DM induces cognitive impairment are still unknown. Here, we confirmed that T1DM mice induced by streptozotocin (STZ) injection had impaired working memory and spatial memory. We observed long-term potentiation (LTP) induction defects and synaptic loss in mice 20 weeks after STZ injection. We also found decreased levels of synaptic proteins, including the N-methyl-D-aspartic acid receptor (NMDAR) subunit NR2A, synaptophysin (SYP), and postsynaptic density 95 (PSD95), in the hippocampus and prefrontal cortex, revealing similarities in the alteration patterns of these synaptic proteins in aged Alzheimer's disease (AD) APP/PS1 mice and T1DM mice. Taken together, these findings expand our understanding of the mechanisms underlying T1DM-induced cognitive impairment.
Collapse
Affiliation(s)
- Yiming Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yueqi Yang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yiqiong Liu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Angyang Guo
- Duke Kunshan University, Kunshan, Jiangsu 215316, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
40
|
Xiong Y, Ruan YT, Zhao J, Yang YW, Chen LP, Mai YR, Yu Q, Cao ZY, Liu FF, Liao W, Liu J. Magnesium-L-threonate exhibited a neuroprotective effect against oxidative stress damage in HT22 cells and Alzheimer’s disease mouse model. World J Psychiatry 2022; 12:410-424. [PMID: 35433327 PMCID: PMC8968501 DOI: 10.5498/wjp.v12.i3.410] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Oxidative stress results in the production of excess reactive oxygen species (ROS) and triggers hippocampal neuronal damage as well as occupies a key role in the pathological mechanisms of neurodegenerative disorders such as Alzheimer’s disease (AD). A recent study confirmed that magnesium had an inhibitory effect against oxidative stress-related malondialdehyde in vitro. However, whether Magnesium-L-threonate (MgT) is capable of suppressing oxidative stress damage in amyloid β (Aβ)25-35-treated HT22 cells and the AD mouse model still remains to be investigated.
AIM To explore the neuroprotective effect of MgT against oxidative stress injury in vitro and in vivo, and investigate the mechanism.
METHODS Aβ25-35-induced HT22 cells were preconditioned with MgT for 12 h. APPswe/PS1dE9 (APP/PS1) mice were orally administered with MgT daily for 3 mo. After MgT treatment, the viability of Aβ25-35-treated HT22 cells was determined via conducting cell counting kit-8 test and the cognition of APP/PS1 mice was measured through the Morris Water Maze. Flow cytometry experiments were applied to assess the ROS levels of HT22 cells and measure the apoptosis rate of HT22 cells or hippocampal neurons. Expression of B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X (Bax), hypoxia-inducible factor (HIF)-1α, NADPH oxidase (NOX) 4, Aβ1-42 and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway proteins was quantified by Western blot.
RESULTS In vitro data confirmed that Aβ25–35-induced HT22 cells had a significantly lower cell viability, higher ROS level and higher apoptosis rates compared with those of control cells (all P < 0.001). MgT prevented the Aβ25-35-triggered oxidative stress damage by elevating viability and decreasing ROS formation and apoptosis of HT22 cells (all P < 0.001). APP/PS1 mice exhibited worse cognitive performance and higher apoptosis rate of hippocampal neurons than wild-type (WT) mice (all P < 0.01). Meanwhile, significant higher expression of Aβ1-42 and NOX4 proteins was detected in APP/PS1 mice than those of WT mice (both P < 0.01). MgT also ameliorated the cognitive deficit, suppressed the apoptosis of hippocampal neuron and downregulated the expression of Aβ1-42 and NOX4 proteins in APP/PS1 mouse (all P < 0.05). Moreover, MgT intervention significantly downregulated HIF-1α and Bax, upregulated Bcl-2 and activated the PI3K/Akt pathway both in vitro and in vivo (all P < 0.05).
CONCLUSION MgT exhibits neuroprotective effects against oxidative stress and hippocampal neuronal apoptosis in Aβ25-35-treated HT22 cells and APP/PS1 mice.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Yu-Ting Ruan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, Guangdong Province, China
| | - Jing Zhao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Yu-Wen Yang
- Department of Medical Ultrasound, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
| | - Li-Ping Chen
- Department of Medical Ultrasound, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
| | - Ying-Ren Mai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Qun Yu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Zhi-Yu Cao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Fei-Fei Liu
- Department of Medical Ultrasound, Xiang’an Hospital of Xiamen University, Xiamen 361000, Fujian Province, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, Guangdong Province, China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| |
Collapse
|
41
|
Liu Y, Hu PP, Zhai S, Feng WX, Zhang R, Li Q, Marshall C, Xiao M, Wu T. Aquaporin 4 deficiency eliminates the beneficial effects of voluntary exercise in a mouse model of Alzheimer's disease. Neural Regen Res 2022; 17:2079-2088. [PMID: 35142700 PMCID: PMC8848602 DOI: 10.4103/1673-5374.335169] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Regular exercise has been shown to reduce the risk of Alzheimer's disease (AD). Our previous study showed that the protein aquaporin 4 (AQP4), which is specifically expressed on the paravascular processes of astrocytes, is necessary for glymphatic clearance of extracellular amyloid beta (Aβ) from the brain, which can delay the progression of Alzheimer's disease. However, it is not known whether AQP4-regulated glymphatic clearance of extracellular Aβ is involved in beneficial effects of exercise in AD patients. Our results showed that after 2 months of voluntary wheel exercise, APP/PS1 mice that were 3 months old at the start of the intervention exhibited a decrease in Aβ burden, glial activation, perivascular AQP4 mislocalization, impaired glymphatic transport, synapse protein loss, and learning and memory defects compared with mice not subjected to the exercise intervention. In contrast, APP/PS1 mice that were 7 months old at the start of the intervention exhibited impaired AQP4 polarity and reduced glymphatic clearance of extracellular Aβ, and the above-mentioned impairments were not alleviated after the 2-month exercise intervention. Compared with age-matched APP/PS1 mice, AQP4 knockout APP/PS1 mice had more serious defects in glymphatic function, Aβ plaque deposition, and cognitive impairment, which could not be alleviated after the exercise intervention. These findings suggest that AQP4-dependent glymphatic transport is the neurobiological basis for the beneficial effects of voluntary exercises that protect against the onset of AD.
Collapse
Affiliation(s)
- Yun Liu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Pan-Pan Hu
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University; Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shuang Zhai
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wei-Xi Feng
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University; Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qian Li
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Charles Marshall
- College of Health Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University; Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ting Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
42
|
Bao J, Liang Z, Gong X, Zhao Y, Wu M, Liu W, Tu C, Wang X, Shu X. Tangeretin Inhibits BACE1 Activity and Attenuates Cognitive Impairments in AD Model Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1536-1546. [PMID: 35084179 DOI: 10.1021/acs.jafc.1c07241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tangeretin (TAN) exhibits many bioactivities, including neuroprotective effects. However, the efficacy of TAN in Alzheimer's disease (AD) has not been sufficiently investigated. In the present study, we integrated behavioral tests, pathology assessment, and biochemical analyses to elucidate the antidementia activity of TAN in APPswe/PSEN1dE9 transgenic (Tg) mice. At supplementation levels of 100 mg/kg body weight per day, TAN significantly attenuated the cognitive impairment of Tg mice in behavioral tests. These effects were associated with less synaptic impairments and fewer β-amyloid accumulations after TAN administration. Furthermore, our study revealed that TAN possessed powerful inhibitory activity against β-secretase both in vitro and in vivo, which played a crucial role in the process of Aβ generation. These findings indicate that TAN is a potential drug for preventing AD pathology. The key mechanism underlying the antidementia effect of TAN may include its inhibitory activity against β-secretase.
Collapse
Affiliation(s)
- Jian Bao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Zheng Liang
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaokang Gong
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yanna Zhao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Mengjuan Wu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Wei Liu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Chenyu Tu
- School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiji Shu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| |
Collapse
|
43
|
Zhu MH, Jogdand AH, Jang J, Nagella SC, Das B, Milosevic MM, Yan R, Antic SD. Evoked Cortical Depolarizations Before and After the Amyloid Plaque Accumulation: Voltage Imaging Study. J Alzheimers Dis 2022; 88:1443-1458. [PMID: 35811528 PMCID: PMC10493004 DOI: 10.3233/jad-220249] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND In Alzheimer's disease (AD), synaptic dysfunction is thought to occur many years before the onset of cognitive decline. OBJECTIVE Detecting synaptic dysfunctions at the earliest stage of AD would be desirable in both clinic and research settings. METHODS Population voltage imaging allows monitoring of synaptic depolarizations, to which calcium imaging is relatively blind. We developed an AD mouse model (APPswe/PS1dE9 background) expressing a genetically-encoded voltage indicator (GEVI) in the neocortex. GEVI was restricted to the excitatory pyramidal neurons (unlike the voltage-sensitive dyes). RESULTS Expression of GEVI did not disrupt AD model formation of amyloid plaques. GEVI expression was stable in both AD model mice and Control (healthy) littermates (CTRL) over 247 days postnatal. Brain slices were stimulated in layer 2/3. From the evoked voltage waveforms, we extracted several parameters for comparison AD versus CTRL. Some parameters (e.g., temporal summation, refractoriness, and peak latency) were weak predictors, while other parameters (e.g., signal amplitude, attenuation with distance, and duration (half-width) of the evoked transients) were stronger predictors of the AD condition. Around postnatal age 150 days (P150) and especially at P200, synaptically-evoked voltage signals in brain slices were weaker in the AD groups versus the age- and sex-matched CTRL groups, suggesting an AD-mediated synaptic weakening that coincides with the accumulation of plaques. However, at the youngest ages examined, P40 and P80, the AD groups showed differentially stronger signals, suggesting "hyperexcitability" prior to the formation of plaques. CONCLUSION Our results indicate bidirectional alterations in cortical physiology in AD model mice; occurring both prior (P40-80), and after (P150-200) the amyloid deposition.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Aditi H Jogdand
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Jinyoung Jang
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Sai C Nagella
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Brati Das
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Milena M Milosevic
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| | - Srdjan D Antic
- Department of Neuroscience, UConn Health, School of Medicine, Farmington, CT, USA
| |
Collapse
|
44
|
Al-Onaizi MA, Thériault P, Lecordier S, Prefontaine P, Rivest S, ElAli A. Early monocyte modulation by the non-erythropoietic peptide ARA 290 decelerates AD-like pathology progression. Brain Behav Immun 2022; 99:363-382. [PMID: 34343617 DOI: 10.1016/j.bbi.2021.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/06/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) pathology is characterized by amyloid-β (Aβ) deposition and tau hyper-phosphorylation, accompanied by a progressive cognitive decline. Monocytes have been recently shown to play a major role in modulating Aβ pathology, and thereby have been pointed as potential therapeutic targets. However, the main challenge remains in identifying clinically relevant interventions that could modulate monocyte immune functions in absence of undesired off-target effects. Erythropoietin (EPO), a key regulator of erythrocyte production, has been shown to possess immunomodulatory potential and to provide beneficial effects in preclinical models of AD. However, the transition to use recombinant human EPO in clinical trials was hindered by unwanted erythropoietic effects that could lead to thrombosis. Here, we used a recently identified non-erythropoietic analogue of EPO, ARA 290, to evaluate its therapeutic potential in AD therapy. We first evaluated the effects of early systemic ARA 290 administration on AD-like pathology in an early-onset model, represented by young APP/PS1 mice. Our findings indicate that ARA 290 early treatment decelerated Aβ pathology progression in APP/PS1 mice while improving cognitive functions. ARA 290 potently increased the levels of total monocytes by specifically stimulating the generation of Ly6CLow patrolling subset, which are implicated in clearing Aβ from the cerebral vasculature, and subsequently reducing overall Aβ burden in the brain. Moreover, ARA 290 increased the levels of monocyte progenitors in the bone marrow. Using chimeric APP/PS1 mice in which Ly6CLow patrolling subset are selectively depleted, ARA 290 was inefficient in attenuating Aβ pathology and ameliorating cognitive functions in young animals. Interestingly, ARA 290 effects were compromised when delivered in a late-onset model, represented by aged APP1/PS1. In aged APP/PS1 mice in which AD-like pathology is at advanced stages, ARA 290 failed to reverse Aβ pathology and to increase the levels of circulating monocytes. Our study suggests that ARA 290 early systemic treatment could prevent AD-like progression via modulation of monocyte functions by specifically increasing the ratio of patrolling monocytes.
Collapse
Affiliation(s)
- Mohammed A Al-Onaizi
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Thériault
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Paul Prefontaine
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Serge Rivest
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
45
|
Zhao YB, Hou XF, Li X, Zhu LS, Zhu J, Ma GR, Liu YX, Miao YC, Zhou QY, Xu L, Zhou QX. Early Memory Impairment is Accompanied by Changes in GluA1/ p-GluA1 in APP/PS1 Mice. Curr Alzheimer Res 2022; 19:667-673. [PMID: 36278470 DOI: 10.2174/1567205020666221019124543] [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: 07/17/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
Abstract
AIMS Exploring the neurobiological mechanisms of early AD damage. BACKGROUND The early diagnosis of Alzheimer's disease (AD) has a very important impact on the prognosis of AD. However, the early symptoms of AD are not obvious and difficult to diagnose. Existing studies have rarely explored the mechanism of early AD. AMPARs are early important learning memory-related receptors. However, it is not clear how the expression levels of AMPARs change in early AD. OBJECTIVE We explored learning memory abilities and AMPAR expression changes in APP/PS1 mice at 4 months, 8 months, and 12 months. METHODS We used the classic Morris water maze to explore the learning and memory impairment of APP/PS1 mice and used western blotting to explore the changes in AMPARs in APP/PS1 mice. RESULTS We found that memory impairment occurred in APP/PS1 mice as early as 4 months of age, and the impairment of learning and memory gradually became serious with age. The changes in GluA1 and p-GluA1 were most pronounced in the early stages of AD in APP/PS1 mice. CONCLUSION Our study found that memory impairment in APP/PS1 mice could be detected as early as 4 months of age, and this early injury may be related to GluA1.
Collapse
Affiliation(s)
- Ya-Bo Zhao
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Xue-Fei Hou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xin Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Li-Su Zhu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jing Zhu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Guo-Rui Ma
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yu-Xuan Liu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yu-Can Miao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qian-Yu Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Lin Xu
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Qi-Xin Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan 650500, China
| |
Collapse
|
46
|
Yang Q, Song D, Xie Z, He G, Zhao J, Wang Z, Dong Z, Zhang H, Yang L, Jiang M, Wu Y, Shi Q, Li J, Yang J, Bai Z, Quan Z, Qing H. Optogenetic stimulation of CA3 pyramidal neurons restores synaptic deficits to improve spatial short-term memory in APP/PS1 mice. Prog Neurobiol 2021; 209:102209. [PMID: 34953962 DOI: 10.1016/j.pneurobio.2021.102209] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 12/11/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022]
Abstract
The hippocampal CA3 region, that is involved in the encoding and retrieval of spatial memory, is found to be synaptically impaired in the early-onset of Alzheimer's disease (AD). It is reported optogenetic manipulation of DG or CA1 can rescue the memory impairment of APP/PS1 mice, however, how CA3 region contributes to AD-related deficits in cognitive function is still unknown. Our work shows optogenetic stimulation of CA3 pyramidal neurons (PNs) significantly restores the impaired spatial short-term memory of APP/PS1 mice. This enhances the anatomical synaptic density/strength and synaptic plasticity as well as activates astrocytes. Chemogenetic inhibiting the activity of CA3 astrocytes reverses the effect of optogenetic stimulation of CA3 PNs that leads to reduced anatomical synaptic density/strength, decreased synaptic protein and AMPA receptors GluA3/4, thus disrupting the cognitive restoration of APP/PS1 mice. These results reveal the molecular mechanism of optogenetic activation of CA3 PNs on restoration of the spatial short-term memory of APP/PS1 mice and unveil a potential strategy of manipulating CA3 for AD treatment.
Collapse
Affiliation(s)
- Qinghu Yang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China; College of Life Sciences & Research Center for Resource Peptide Drugs, Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, YananUniversity, Yanan, 716000, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China
| | - Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China
| | - Guiqiong He
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China; Department of Anatomy, Chongqing Medical University, Chongqing, 400016, China
| | - Juan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China
| | - Zhe Wang
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China; The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Heao Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China
| | - Liang Yang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China; College of Life Sciences & Research Center for Resource Peptide Drugs, Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, YananUniversity, Yanan, 716000, China
| | - Ming Jiang
- College of Life Sciences & Research Center for Resource Peptide Drugs, Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, YananUniversity, Yanan, 716000, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, 272067, China; Shandong Key Laboratory of Behavioral Medicine, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, 272067, China
| | - Qing Shi
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing, 100081, China
| | - Junjie Li
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jun Yang
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Zhantao Bai
- College of Life Sciences & Research Center for Resource Peptide Drugs, Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, YananUniversity, Yanan, 716000, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 10008, China.
| |
Collapse
|
47
|
Zhao Y, Bao J, Liu W, Gong X, Liang Z, Li W, Wu M, Xiao Y, Sun B, Wang X, Wang JZ, Wang J, Shu X. Spatial Training Attenuates Long-Term Alzheimer’s Disease-Related Pathogenic Processes in APP/PS1 Mice. J Alzheimers Dis 2021; 85:1453-1466. [DOI: 10.3233/jad-215016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background: Alzheimer’s disease (AD), with cognitive impairment as the main clinical manifestation, is a progressive neurodegenerative disease. The assembly of amyloid-β (Aβ) as senile plaques is one of the most well-known histopathological alterations in AD. Several studies reported that cognitive training reduced Aβ deposition and delayed memory loss. However, the long-term benefits of spatial training and the underlying neurobiological mechanisms have not yet been elucidated. Objective: To explore the long-term effects of spatial training on AD-related pathogenic processes in APP/PS1 mice. Methods: We used Morris water maze (MWM), Open Field, Barnes Maze, western blotting, qPCR, and immunofluorescence. Results: One-month MWM training in APP/PS1 mice at 2.5 months of age could attenuate Aβ deposition and decrease the expression of β-secretase (BACE1) and amyloid-β protein precursor (AβPP) with long-term effects. Simultaneously, regular spatial training increased the expression of synapse-related proteins in the hippocampus. Moreover, MWM training increased adult hippocampal neurogenesis in AD model mice. Nonetheless, cognitive deficits in APP/PS1 transgenic mice at 7 months of age were not attenuated by MWM training at an early stage. Conclusion: Our study demonstrates that MWM training alleviates amyloid plaque burden and adult hippocampal neurogenesis deficits with long-term effects in AD model mice.
Collapse
Affiliation(s)
- Yang Zhao
- International Joint Research Center for General Health, Precision Medicine & Nutrition, Hubei University of Technology, Wuhan, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
| | - Jian Bao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Wei Liu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Xiaokang Gong
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
| | - Zheng Liang
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
| | - Wenshuang Li
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
| | - Mengjuan Wu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Yifan Xiao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Binlian Sun
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Wang
- International Joint Research Center for General Health, Precision Medicine & Nutrition, Hubei University of Technology, Wuhan, China
| | - Xiji Shu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| |
Collapse
|
48
|
Park J, Kim CH. Regulation of common neurological disorders by gut microbial metabolites. Exp Mol Med 2021; 53:1821-1833. [PMID: 34857900 PMCID: PMC8741890 DOI: 10.1038/s12276-021-00703-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/06/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The gut is connected to the CNS by immunological mediators, lymphocytes, neurotransmitters, microbes and microbial metabolites. A mounting body of evidence indicates that the microbiome exerts significant effects on immune cells and CNS cells. These effects frequently result in the suppression or exacerbation of inflammatory responses, the latter of which can lead to severe tissue damage, altered synapse formation and disrupted maintenance of the CNS. Herein, we review recent progress in research on the microbial regulation of CNS diseases with a focus on major gut microbial metabolites, such as short-chain fatty acids, tryptophan metabolites, and secondary bile acids. Pathological changes in the CNS are associated with dysbiosis and altered levels of microbial metabolites, which can further exacerbate various neurological disorders. The cellular and molecular mechanisms by which these gut microbial metabolites regulate inflammatory diseases in the CNS are discussed. We highlight the similarities and differences in the impact on four major CNS diseases, i.e., multiple sclerosis, Parkinson's disease, Alzheimer's disease, and autism spectrum disorder, to identify common cellular and molecular networks governing the regulation of cellular constituents and pathogenesis in the CNS by microbial metabolites.
Collapse
Affiliation(s)
- Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
- Mary H. Weiser Food Allergy Center, Center for Gastrointestinal Research, and Rogel Center for Cancer Research, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
49
|
Navakkode S, Gaunt JR, Pavon MV, Bansal VA, Abraham RP, Chong YS, Ch'ng TH, Sajikumar S. Sex-specific accelerated decay in time/activity-dependent plasticity and associative memory in an animal model of Alzheimer's disease. Aging Cell 2021; 20:e13502. [PMID: 34796608 PMCID: PMC8672784 DOI: 10.1111/acel.13502] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/02/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical studies have shown that female brains are more predisposed to neurodegenerative diseases such as Alzheimer's disease (AD), but the cellular and molecular mechanisms behind this disparity remain unknown. In several mouse models of AD, synaptic plasticity dysfunction is an early event and appears before significant accumulation of amyloid plaques and neuronal degeneration. However, it is unclear whether sexual dimorphism at the synaptic level contributes to the higher risk and prevalence of AD in females. Our studies on APP/PS1 (APPSwe/PS1dE9) mouse model show that AD impacts hippocampal long‐term plasticity in a sex‐specific manner. Long‐term potentiation (LTP) induced by strong tetanic stimulation (STET), theta burst stimulation (TBS) and population spike timing‐dependent plasticity (pSTDP) show a faster decay in AD females compared with age‐matched AD males. In addition, behavioural tagging (BT), a model of associative memory, is specifically impaired in AD females with a faster decay in memory compared with males. Together with the plasticity and behavioural data, we also observed an upregulation of neuroinflammatory markers, along with downregulation of transcripts that regulate cellular processes associated with synaptic plasticity and memory in females. Immunohistochemistry of AD brains confirms that female APP/PS1 mice carry a higher amyloid plaque burden and have enhanced microglial activation compared with male APP/PS1 mice. Their presence in the diseased mice also suggests a link between the impairment of LTP and the upregulation of the inflammatory response. Overall, our data show that synaptic plasticity and associative memory impairments are more prominent in females and this might account for the faster progression of AD in females.
Collapse
Affiliation(s)
- Sheeja Navakkode
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Department of Physiology National University of Singapore Singapore Singapore
| | - Jessica Ruth Gaunt
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Maria Vazquez Pavon
- Department of Physiology National University of Singapore Singapore Singapore
| | | | - Riya Prasad Abraham
- Department of Physiology National University of Singapore Singapore Singapore
| | - Yee Song Chong
- Department of Physiology National University of Singapore Singapore Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Science Nanyang Technological University Singapore Singapore
| | - Sreedharan Sajikumar
- Department of Physiology National University of Singapore Singapore Singapore
- Healthy Longevity Translational Research Programme Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Life Sciences Institute Neurobiology Programme National University of Singapore Singapore Singapore
| |
Collapse
|
50
|
Yue X, Zhou Y, Qiao M, Zhao X, Huang X, Zhao T, Cheng X, Fan M, Zhao Y, Chen R, Zhu L. Intermittent hypoxia treatment alleviates memory impairment in the 6-month-old APPswe/PS1dE9 mice and reduces amyloid beta accumulation and inflammation in the brain. Alzheimers Res Ther 2021; 13:194. [PMID: 34844651 PMCID: PMC8630860 DOI: 10.1186/s13195-021-00935-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Background Alzheimer’s disease (AD) is a progressive, degenerative, and terminal disease without cure. There is an urgent need for a new strategy to treat AD. The aim of this study was to investigate the effects of intermittent hypoxic treatment (IHT) on cognitive functions in a mouse model of AD and unravel the mechanism of action of IHT. Methods Six-month-old APPswe/PS1dE9 (APP/PS1) male mice were exposed to hypoxic environment (14.3% O2) 4 h/day for 14 days or 28 days. Cognitive functions were measured by Morris water maze test after either 14 days or 42 days of interval. Thereafter the distribution of amyloid plaque and microglial activation were determined by mouse brain immunohistochemistry, while the amyloid beta (Aβ) and inflammatory cytokines were measured by ELISA and Western Blot. Microarray was used for studying gene expressions in the hippocampus. Results IHT for 14 days or 28 days significantly improved the spatial memory ability of the 6-month-old APP/PS1 mice. The memory improvement by 14 days IHT lasted to 14 days, but not to 42 days. The level of Aβ plaques and neurofilament accumulations was reduced markedly after the IHT exposure. IHT reduced the pro-inflammatory cytokines IL-1β, IL-6 levels, and β-secretase cleavage of APP processing which implies reduced Aβ production. Microarray analysis revealed a large number of genes in the hippocampus were significantly altered which are known to be metabolism-regulated genes. Conclusions This study provides evidence of the beneficial effect of IHT on the progression of AD by alleviating memory impairment, reducing Aβ accumulation and inflammation in the brain. IHT can be developed as a novel measure to relieve the progression of AD by targeting multiple pathways in the AD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00935-z.
Collapse
Affiliation(s)
- Xiangpei Yue
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yanzhao Zhou
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Meng Qiao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xingnan Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Tong Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xiang Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ming Fan
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yongqi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ruoli Chen
- Institute for Science and Technology in Medicine, School of Pharmacy, Keele University, Kelle, UK.
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China. .,Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|