101
|
Portalés A, Sánchez-Aguilera A, Royo M, Jurado S. Assessment of social behavior and chemosensory cue detection in an animal model of neurodegeneration. Methods Cell Biol 2024; 185:137-150. [PMID: 38556445 DOI: 10.1016/bs.mcb.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Numerous studies have shown that aging in humans leads to a decline in olfactory function, resulting in deficits in acuity, detection threshold, discrimination, and olfactory-associated memories. Furthermore, impaired olfaction has been identified as a potential indicator for the onset of age-related neurodegenerative diseases, including Alzheimer's disease (AD). Studies conducted on mouse models of AD have largely mirrored the findings in humans, thus providing a valuable system to investigate the cellular and circuit adaptations of the olfactory system during natural and pathological aging. However, the majority of previous research has focused on assessing the detection of neutral or synthetic odors, with little attention given to the impact of aging and neurodegeneration on the recognition of social cues-a critical feature for the survival of mammalian species. Therefore, in this study, we present a battery of olfactory tests that use conspecific urine samples to examine the changes in social odor recognition in a mouse model of neurodegeneration.
Collapse
Affiliation(s)
- Adrián Portalés
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Alicante, Spain.
| | - Alberto Sánchez-Aguilera
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, IdISSC, Madrid, Spain
| | - Maria Royo
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Alicante, Spain
| | - Sandra Jurado
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
102
|
Kotah JM, Kater MSJ, Brosens N, Lesuis SL, Tandari R, Blok TM, Marchetto L, Yusaf E, Koopmans FTW, Smit AB, Lucassen PJ, Krugers HJ, Verheijen MHG, Korosi A. Early-life stress and amyloidosis in mice share pathogenic pathways involving synaptic mitochondria and lipid metabolism. Alzheimers Dement 2024; 20:1637-1655. [PMID: 38055782 PMCID: PMC10984508 DOI: 10.1002/alz.13569] [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: 05/30/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Early-life stress (ES) increases the risk for Alzheimer's disease (AD). We and others have shown that ES aggravates amyloid-beta (Aβ) pathology and promotes cognitive dysfunction in APP/PS1 mice, but underlying mechanisms remain unclear. METHODS We studied how ES affects the hippocampal synaptic proteome in wild-type (WT) and APP/PS1 mice at early and late pathological stages, and validated hits using electron microscopy and immunofluorescence. RESULTS The hippocampal synaptosomes of both ES-exposed WT and early-stage APP/PS1 mice showed a relative decrease in actin dynamics-related proteins and a relative increase in mitochondrial proteins. ES had minimal effects on older WT mice, while strongly affecting the synaptic proteome of advanced stage APP/PS1 mice, particularly the expression of astrocytic and mitochondrial proteins. DISCUSSION Our data show that ES and amyloidosis share pathogenic pathways involving synaptic mitochondrial dysfunction and lipid metabolism, which may underlie the observed impact of ES on the trajectory of AD.
Collapse
Affiliation(s)
- Janssen M. Kotah
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mandy S. J. Kater
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Niek Brosens
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Roberta Tandari
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Thomas M. Blok
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Luca Marchetto
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ella Yusaf
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Frank T. W. Koopmans
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - August B. Smit
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Paul J. Lucassen
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Harm J. Krugers
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Aniko Korosi
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
103
|
Kourti M, Metaxas A. A systematic review and meta-analysis of tau phosphorylation in mouse models of familial Alzheimer's disease. Neurobiol Dis 2024; 192:106427. [PMID: 38307366 DOI: 10.1016/j.nbd.2024.106427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Transgenic models of familial Alzheimer's disease (AD) serve as valuable tools for probing the molecular mechanisms associated with amyloid-beta (Aβ)-induced pathology. In this meta-analysis, we sought to evaluate levels of phosphorylated tau (p-tau) and explore potential age-related variations in tau hyperphosphorylation, within mouse models of AD. The PubMed and Scopus databases were searched for studies measuring soluble p-tau in 5xFAD, APPswe/PSEN1de9, J20 and APP23 mice. Data were extracted and analyzed using standardized procedures. For the 5xFAD model, the search yielded 36 studies eligible for meta-analysis. Levels of p-tau were higher in 5xFAD mice relative to control, a difference that was evident in both the carboxy-terminal (CT) and proline-rich (PR) domains of tau. Age negatively moderated the relationship between genotype and CT phosphorylated tau in studies using hybrid mice, female mice, and preparations from the neocortex. For the APPswe/PSEN1de9 model, the search yielded 27 studies. Analysis showed tau hyperphosphorylation in transgenic vs. control animals, evident in both the CT and PR regions of tau. Age positively moderated the relationship between genotype and PR domain phosphorylated tau in the neocortex of APPswe/PSEN1de9 mice. A meta-analysis was not performed for the J20 and APP23 models, due to the limited number of studies measuring p-tau levels in these mice (<10 studies). Although tau is hyperphosphorylated in both 5xFAD and APPswe/PSEN1de9 mice, the effects of ageing on p-tau are contingent upon the model being examined. These observations emphasize the importance of tailoring model selection to the appropriate disease stage when considering the relationship between Aβ and tau, and suggest that there are optimal intervention points for the administration of both anti-amyloid and anti-tau therapies.
Collapse
Affiliation(s)
- Malamati Kourti
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Centre, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus.
| | - Athanasios Metaxas
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
104
|
Pádua MS, Guil-Guerrero JL, Prates JAM, Lopes PA. Insights on the Use of Transgenic Mice Models in Alzheimer's Disease Research. Int J Mol Sci 2024; 25:2805. [PMID: 38474051 DOI: 10.3390/ijms25052805] [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: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, presents a significant global health challenge with no known cure to date. Central to our understanding of AD pathogenesis is the β-amyloid cascade hypothesis, which underlies drug research and discovery efforts. Despite extensive studies, no animal models of AD have completely validated this hypothesis. Effective AD models are essential for accurately replicating key pathological features of the disease, notably the formation of β-amyloid plaques and neurofibrillary tangles. These pathological markers are primarily driven by mutations in the amyloid precursor protein (APP) and presenilin 1 (PS1) genes in familial AD (FAD) and by tau protein mutations for the tangle pathology. Transgenic mice models have been instrumental in AD research, heavily relying on the overexpression of mutated APP genes to simulate disease conditions. However, these models do not entirely replicate the human condition of AD. This review aims to provide a comprehensive evaluation of the historical and ongoing research efforts in AD, particularly through the use of transgenic mice models. It is focused on the benefits gathered from these transgenic mice models in understanding β-amyloid toxicity and the broader biological underpinnings of AD. Additionally, the review critically assesses the application of these models in the preclinical testing of new therapeutic interventions, highlighting the gap between animal models and human clinical realities. This analysis underscores the need for refinement in AD research methodologies to bridge this gap and enhance the translational value of preclinical studies.
Collapse
Affiliation(s)
- Mafalda Soares Pádua
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| | - José L Guil-Guerrero
- Departamento de Tecnología de Alimentos, Universidad de Almería, 04120 Almería, Spain
| | - José A M Prates
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| | - Paula Alexandra Lopes
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| |
Collapse
|
105
|
Papazoglou A, Henseler C, Weickhardt S, Teipelke J, Papazoglou P, Daubner J, Schiffer T, Krings D, Broich K, Hescheler J, Sachinidis A, Ehninger D, Scholl C, Haenisch B, Weiergräber M. Sex- and region-specific cortical and hippocampal whole genome transcriptome profiles from control and APP/PS1 Alzheimer's disease mice. PLoS One 2024; 19:e0296959. [PMID: 38324617 PMCID: PMC10849391 DOI: 10.1371/journal.pone.0296959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024] Open
Abstract
A variety of Alzheimer's disease (AD) mouse models has been established and characterized within the last decades. To get an integrative view of the sophisticated etiopathogenesis of AD, whole genome transcriptome studies turned out to be indispensable. Here we carried out microarray data collection based on RNA extracted from the retrosplenial cortex and hippocampus of age-matched, eight months old male and female APP/PS1 AD mice and control animals to perform sex- and brain region specific analysis of transcriptome profiles. The results of our studies reveal novel, detailed insight into differentially expressed signature genes and related fold changes in the individual APP/PS1 subgroups. Gene ontology and Venn analysis unmasked that intersectional, upregulated genes were predominantly involved in, e.g., activation of microglial, astrocytic and neutrophilic cells, innate immune response/immune effector response, neuroinflammation, phagosome/proteasome activation, and synaptic transmission. The number of (intersectional) downregulated genes was substantially less in the different subgroups and related GO categories included, e.g., the synaptic vesicle docking/fusion machinery, synaptic transmission, rRNA processing, ubiquitination, proteasome degradation, histone modification and cellular senescence. Importantly, this is the first study to systematically unravel sex- and brain region-specific transcriptome fingerprints/signature genes in APP/PS1 mice. The latter will be of central relevance in future preclinical and clinical AD related studies, biomarker characterization and personalized medicinal approaches.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Sandra Weickhardt
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Jenni Teipelke
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Panagiota Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Teresa Schiffer
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Damian Krings
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Jürgen Hescheler
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Dan Ehninger
- Translational Biogerontology, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
| | - Catharina Scholl
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
- Center for Translational Medicine, Medical Faculty, University of Bonn, Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
106
|
Wong D, Bellyou M, Li A, Prado MAM, Beauchet O, Annweiler C, Montero-Odasso M, Bartha R. Magnetic resonance spectroscopy in the hippocampus of adult APP/PS1 mice following chronic vitamin D deficiency. Behav Brain Res 2024; 457:114713. [PMID: 37838248 DOI: 10.1016/j.bbr.2023.114713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Vitamin D (VitD) deficiency can exacerbate AD progression and may cause changes in brain metabolite levels that can be detected by magnetic resonance spectroscopy (MRS). The purpose of this study was to determine whether chronic VitD deficiency in an AD mouse model caused persistent metabolite levels changes in the hippocampus associated with memory performance. Six-month-old APPSwe/PS1ΔE9 (APP/PS1) mice (N = 14 mice/group) were fed either a VitD deficient (VitD-) diet or a control diet. Metabolite level changes in the hippocampus were evaluated by 1H MRS using a 9.4 T MRI. Ventricle volume was assessed by imaging and spatial memory was evaluated using the Barnes maze. All measurements were made at 6, 9, 12, and 15 months of age. At 15 months of age, amyloid plaque load and astrocyte number were evaluated histologically (N = 4 mice/group). Levels of N-acetyl aspartate and creatine were lower in VitD- mice compared to control diet mice at 12 months of age. VitD deficiency did not change ventricle volume. Lactate levels increased over time in VitD- mice and increases from 12 to 15 months were negatively correlated with changes in primary latency to the target hole in the Barns Maze. VitD- mice showed improved spatial memory performance compared to control diet mice. VitD- mice also had more astrocytes in the cortex and hippocampus at 15 months than control diet mice. This study suggests that severe VitD deficiency in APP/PS1 mice may lead to compensatory changes in metabolite and astrocyte levels that contribute to improved performance on spatial memory tasks.
Collapse
Affiliation(s)
- Dickson Wong
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Miranda Bellyou
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alex Li
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Marco A M Prado
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada; Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | | | - Cédric Annweiler
- Department of Geriatric Medicine and Memory Clinic, Research Center on Autonomy and Longevity, University Hospital, Angers, France
| | - Manuel Montero-Odasso
- Department of Medicine, Division of Geriatric Medicine, Parkwood Hospital, Western University, London, ON, Canada; Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Lawson Health Research Institute, London, ON, Canada
| | - Robert Bartha
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| |
Collapse
|
107
|
Lee JD, Won W, Kimball K, Wang Y, Yeboah F, Evitts KM, Neiswanger C, Schattauer S, Rappleye M, Bremner SB, Chun C, Smith N, Mack DL, Young JE, Lee CJ, Chavkin C, Berndt A. Structure-guided engineering of a fast genetically encoded sensor for real-time H 2O 2 monitoring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578117. [PMID: 38352381 PMCID: PMC10862829 DOI: 10.1101/2024.01.31.578117] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Hydrogen Peroxide (H2O2) is a central oxidant in redox biology due to its pleiotropic role in physiology and pathology. However, real-time monitoring of H2O2 in living cells and tissues remains a challenge. We address this gap with the development of an optogenetic hydRogen perOxide Sensor (oROS), leveraging the bacterial peroxide binding domain OxyR. Previously engineered OxyR-based fluorescent peroxide sensors lack the necessary sensitivity or response speed for effective real-time monitoring. By structurally redesigning the fusion of Escherichia coli (E. coli) ecOxyR with a circularly permutated green fluorescent protein (cpGFP), we created a novel, green-fluorescent peroxide sensor oROS-G. oROS-G exhibits high sensitivity and fast on-and-off kinetics, ideal for monitoring intracellular H2O2 dynamics. We successfully tracked real-time transient and steady-state H2O2 levels in diverse biological systems, including human stem cell-derived neurons and cardiomyocytes, primary neurons and astrocytes, and mouse neurons and astrocytes in ex vivo brain slices. These applications demonstrate oROS's capabilities to monitor H2O2 as a secondary response to pharmacologically induced oxidative stress, G-protein coupled receptor (GPCR)-induced cell signaling, and when adapting to varying metabolic stress. We showcased the increased oxidative stress in astrocytes via Aβ-putriscine-MAOB axis, highlighting the sensor's relevance in validating neurodegenerative disease models. oROS is a versatile tool, offering a window into the dynamic landscape of H2O2 signaling. This advancement paves the way for a deeper understanding of redox physiology, with significant implications for diseases associated with oxidative stress, such as cancer, neurodegenerative disorders, and cardiovascular diseases.
Collapse
Affiliation(s)
- Justin Daho Lee
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Kandace Kimball
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Yihan Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Fred Yeboah
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kira M Evitts
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Carlie Neiswanger
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Selena Schattauer
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Michael Rappleye
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Samantha B Bremner
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Changho Chun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Netta Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - David L Mack
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Jessica E Young
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Charles Chavkin
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Andre Berndt
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| |
Collapse
|
108
|
Kommaddi RP, Gowaikar R, P A H, Diwakar L, Singh K, Mondal A. Akt activation ameliorates deficits in hippocampal-dependent memory and activity-dependent synaptic protein synthesis in an Alzheimer's disease mouse model. J Biol Chem 2024; 300:105619. [PMID: 38182004 PMCID: PMC10839450 DOI: 10.1016/j.jbc.2023.105619] [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: 08/24/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/07/2024] Open
Abstract
Protein kinase-B (Akt) and the mechanistic target of rapamycin (mTOR) signaling pathways are implicated in Alzheimer's disease (AD) pathology. Akt/mTOR signaling pathways, activated by external inputs, enable new protein synthesis at the synapse and synaptic plasticity. The molecular mechanisms impeding new protein synthesis at the synapse in AD pathogenesis remain elusive. Here, we aimed to understand the molecular mechanisms prior to the manifestation of histopathological hallmarks by characterizing Akt1/mTOR signaling cascades and new protein synthesis in the hippocampus of WT and amyloid precursor protein/presenilin-1 (APP/PS1) male mice. Intriguingly, compared to those in WT mice, we found significant decreases in pAkt1, pGSK3β, pmTOR, pS6 ribosomal protein, and p4E-BP1 levels in both post nuclear supernatant and synaptosomes isolated from the hippocampus of one-month-old (presymptomatic) APP/PS1 mice. In synaptoneurosomes prepared from the hippocampus of presymptomatic APP/PS1 mice, activity-dependent protein synthesis at the synapse was impaired and this deficit was sustained in young adults. In hippocampal neurons from C57BL/6 mice, downregulation of Akt1 precluded synaptic activity-dependent protein synthesis at the dendrites but not in the soma. In three-month-old APP/PS1 mice, Akt activator (SC79) administration restored deficits in memory recall and activity-dependent synaptic protein synthesis. C57BL/6 mice administered with an Akt inhibitor (MK2206) resulted in memory recall deficits compared to those treated with vehicle. We conclude that dysregulation of Akt1/mTOR and its downstream signaling molecules in the hippocampus contribute to memory recall deficits and loss of activity-dependent synaptic protein synthesis. In AD mice, however, Akt activation ameliorates deficits in memory recall and activity-dependent synaptic protein synthesis.
Collapse
Affiliation(s)
| | - Ruturaj Gowaikar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India; Manipal Academy of Higher Education, Manipal, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Kunal Singh
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Amrita Mondal
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
109
|
Harris BN, Yavari M, Ramalingam L, Mounce PL, Alers Maldonado K, Chavira AC, Thomas S, Scoggin S, Biltz C, Moustaid-Moussa N. Impact of Long-Term Dietary High Fat and Eicosapentaenoic Acid on Behavior and Hypothalamic-Pituitary-Adrenal Axis Activity in Amyloidogenic APPswe/PSEN1dE9 Mice. Neuroendocrinology 2024; 114:553-576. [PMID: 38301617 PMCID: PMC11153005 DOI: 10.1159/000536586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) alters neurocognitive and emotional function and causes dysregulation of multiple homeostatic processes. The leading AD framework pins amyloid beta plaques and tau tangles as primary drivers of dysfunction. However, many additional variables, including diet, stress, sex, age, and pain tolerance, interact in ways that are not fully understood to impact the onset and progression of AD pathophysiology. We asked: (1) does high-fat diet, compared to low-fat diet, exacerbate AD pathophysiology and behavioral decline? And, (2) can supplementation with eicosapentaenoic (EPA)-enriched fish oil prevent high-fat-diet-induced changes? METHODS Male and female APPswePSdE9 mice, and their non-transgenic littermates, were randomly assigned to a diet condition (low-fat, high-fat, high-fat with EPA) and followed from 2 to 10 months of age. We assessed baseline corticosterone concentration during aging, pain tolerance, cognitive function, stress coping, and corticosterone response to a stressor. RESULTS Transgenic mice were consistently more active than non-transgenic mice but did not perform worse on either cognitive task, even though we recently reported that these same transgenic mice exhibited metabolic changes and had increased amyloid beta. Mice fed high-fat diet had higher baseline and post-stressor corticosterone, but diet did not impact cognition or pain tolerance. Sex had the biggest influence, as female mice were consistently more active and had higher corticosterone than males. CONCLUSION Overall, diet, genotype, and sex did not have consistent impacts on outcomes. We found little support for predicted interactions and correlations, suggesting diet impacts metabolic function and amyloid beta levels, but these outcomes do not translate to changes in behaviors measured here.
Collapse
Affiliation(s)
- Breanna N. Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| | - Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Molecular Metabolism, School of Public Health, Harvard University, Boston, MA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Nutritional and Food Studies Syracuse University, Syracuse, NY
| | - P. Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | | | - Angela C. Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Sarah Thomas
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| |
Collapse
|
110
|
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, Kawoos U, Zhu CW, Janssen WGM, Stone JR, Hof PR, Cook DG, Ahlers ST. The Neurovascular Unit as a Locus of Injury in Low-Level Blast-Induced Neurotrauma. Int J Mol Sci 2024; 25:1150. [PMID: 38256223 PMCID: PMC10816929 DOI: 10.3390/ijms25021150] [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: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Blast-induced neurotrauma has received much attention over the past decade. Vascular injury occurs early following blast exposure. Indeed, in animal models that approximate human mild traumatic brain injury or subclinical blast exposure, vascular pathology can occur in the presence of a normal neuropil, suggesting that the vasculature is particularly vulnerable. Brain endothelial cells and their supporting glial and neuronal elements constitute a neurovascular unit (NVU). Blast injury disrupts gliovascular and neurovascular connections in addition to damaging endothelial cells, basal laminae, smooth muscle cells, and pericytes as well as causing extracellular matrix reorganization. Perivascular pathology becomes associated with phospho-tau accumulation and chronic perivascular inflammation. Disruption of the NVU should impact activity-dependent regulation of cerebral blood flow, blood-brain barrier permeability, and glymphatic flow. Here, we review work in an animal model of low-level blast injury that we have been studying for over a decade. We review work supporting the NVU as a locus of low-level blast injury. We integrate our findings with those from other laboratories studying similar models that collectively suggest that damage to astrocytes and other perivascular cells as well as chronic immune activation play a role in the persistent neurobehavioral changes that follow blast injury.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Carolyn W. Zhu
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William G. M. Janssen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James R. Stone
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22903, USA;
| | - Patrick R. Hof
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA 98108, USA;
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
| |
Collapse
|
111
|
Ferré-González L, Balaguer Á, Roca M, Ftara A, Lloret A, Cháfer-Pericás C. Brain areas lipidomics in female transgenic mouse model of Alzheimer's disease. Sci Rep 2024; 14:870. [PMID: 38195731 PMCID: PMC10776612 DOI: 10.1038/s41598-024-51463-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/05/2024] [Indexed: 01/11/2024] Open
Abstract
Lipids are the major component of the brain with important structural and functional properties. Lipid disruption could play a relevant role in Alzheimer's disease (AD). Some brain lipidomic studies showed significant differences compared to controls, but few studies have focused on different brain areas related to AD. Furthermore, AD is more prevalent in females, but there is a lack of studies focusing on this sex. This work aims to perform a lipidomic study in selected brain areas (cerebellum, amygdala, hippocampus, entire cortex) from wild-type (WT, n = 10) and APPswe/PS1dE9 transgenic (TG, n = 10) female mice of 5 months of age, as a model of early AD, to identify alterations in lipid composition. A lipidomic mass spectrometry-based method was optimized and applied to brain tissue. As result, some lipids showed statistically significant differences between mice groups in cerebellum (n = 68), amygdala (n = 49), hippocampus (n = 48), and the cortex (n = 22). In addition, some lipids (n = 15) from the glycerolipid, phospholipid, and sphingolipid families were statistically significant in several brain areas simultaneously between WT and TG. A selection of lipid variables was made to develop a multivariate approach to assess their discriminant potential, showing high diagnostic indexes, especially in cerebellum and amygdala (sensitivity 70-100%, sensibility 80-100%).
Collapse
Affiliation(s)
- Laura Ferré-González
- Alzheimer's Disease Research Group, Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ángel Balaguer
- Faculty of Mathematics, University of Valencia, Valencia, Spain
| | - Marta Roca
- Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | | | - Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer's Disease Research Group, Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| |
Collapse
|
112
|
Koller EJ, Wood CA, Lai Z, Borgenheimer E, Hoffman KL, Jankowsky JL. Doxycycline for transgene control disrupts gut microbiome diversity without compromising acute neuroinflammatory response. J Neuroinflammation 2024; 21:11. [PMID: 38178148 PMCID: PMC10765643 DOI: 10.1186/s12974-023-03004-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
The tetracycline transactivator (tTA) system provides controllable transgene expression through oral administration of the broad-spectrum antibiotic doxycycline. Antibiotic treatment for transgene control in mouse models of disease might have undesirable systemic effects resulting from changes in the gut microbiome. Here we assessed the impact of doxycycline on gut microbiome diversity in a tTA-controlled model of Alzheimer's disease and then examined neuroimmune effects of these microbiome alterations following acute LPS challenge. We show that doxycycline decreased microbiome diversity in both transgenic and wild-type mice and that these changes persisted long after drug withdrawal. Despite the change in microbiome composition, doxycycline treatment had minimal effect on basal transcriptional signatures of inflammation the brain or on the neuroimmune response to LPS challenge. Our findings suggest that central neuroimmune responses may be less affected by doxycycline at doses needed for transgene control than by antibiotic cocktails at doses used for experimental microbiome disruption.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM295, Houston, TX, 77030, USA
| | - Caleb A Wood
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM295, Houston, TX, 77030, USA
| | - Zoe Lai
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM295, Houston, TX, 77030, USA
| | - Ella Borgenheimer
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM295, Houston, TX, 77030, USA
| | - Kristi L Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Joanna L Jankowsky
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Mail Stop BCM295, Houston, TX, 77030, USA.
- Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Huffington Center On Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
113
|
Taylor LW, Simzer EM, Pimblett C, Lacey-Solymar OTT, McGeachan RI, Meftah S, Rose JL, Spires-Jones MP, Holt K, Catterson JH, Koch H, Liaquat I, Clarke JH, Skidmore J, Smith C, Booker SA, Brennan PM, Spires-Jones TL, Durrant CS. p-tau Ser356 is associated with Alzheimer's disease pathology and is lowered in brain slice cultures using the NUAK inhibitor WZ4003. Acta Neuropathol 2024; 147:7. [PMID: 38175261 PMCID: PMC10766794 DOI: 10.1007/s00401-023-02667-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Tau hyperphosphorylation and aggregation is a common feature of many dementia-causing neurodegenerative diseases. Tau can be phosphorylated at up to 85 different sites, and there is increasing interest in whether tau phosphorylation at specific epitopes, by specific kinases, plays an important role in disease progression. The AMP-activated protein kinase (AMPK)-related enzyme NUAK1 has been identified as a potential mediator of tau pathology, whereby NUAK1-mediated phosphorylation of tau at Ser356 prevents the degradation of tau by the proteasome, further exacerbating tau hyperphosphorylation and accumulation. This study provides a detailed characterisation of the association of p-tau Ser356 with progression of Alzheimer's disease pathology, identifying a Braak stage-dependent increase in p-tau Ser356 protein levels and an almost ubiquitous presence in neurofibrillary tangles. We also demonstrate, using sub-diffraction-limit resolution array tomography imaging, that p-tau Ser356 co-localises with synapses in AD postmortem brain tissue, increasing evidence that this form of tau may play important roles in AD progression. To assess the potential impacts of pharmacological NUAK inhibition in an ex vivo system that retains multiple cell types and brain-relevant neuronal architecture, we treated postnatal mouse organotypic brain slice cultures from wildtype or APP/PS1 littermates with the commercially available NUAK1/2 inhibitor WZ4003. Whilst there were no genotype-specific effects, we found that WZ4003 results in a culture-phase-dependent loss of total tau and p-tau Ser356, which corresponds with a reduction in neuronal and synaptic proteins. By contrast, application of WZ4003 to live human brain slice cultures results in a specific lowering of p-tau Ser356, alongside increased neuronal tubulin protein. This work identifies differential responses of postnatal mouse organotypic brain slice cultures and adult human brain slice cultures to NUAK1 inhibition that will be important to consider in future work developing tau-targeting therapeutics for human disease.
Collapse
Affiliation(s)
- Lewis W Taylor
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Elizabeth M Simzer
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire Pimblett
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Robert I McGeachan
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
- The Hospital for Small Animals, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Soraya Meftah
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jamie L Rose
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | | | - Kristján Holt
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - James H Catterson
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Henner Koch
- Department of Neurology, Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Imran Liaquat
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Jonathan H Clarke
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - John Skidmore
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - Colin Smith
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, UK
| | - Paul M Brennan
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Brain Tumour Centre of Excellence, CRUK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire S Durrant
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
114
|
Khan R, Di Gesù CM, Lee J, McCullough LD. The contribution of age-related changes in the gut-brain axis to neurological disorders. Gut Microbes 2024; 16:2302801. [PMID: 38237031 PMCID: PMC10798364 DOI: 10.1080/19490976.2024.2302801] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Trillions of microbes live symbiotically in the host, specifically in mucosal tissues such as the gut. Recent advances in metagenomics and metabolomics have revealed that the gut microbiota plays a critical role in the regulation of host immunity and metabolism, communicating through bidirectional interactions in the microbiota-gut-brain axis (MGBA). The gut microbiota regulates both gut and systemic immunity and contributes to the neurodevelopment and behaviors of the host. With aging, the composition of the microbiota changes, and emerging studies have linked these shifts in microbial populations to age-related neurological diseases (NDs). Preclinical studies have demonstrated that gut microbiota-targeted therapies can improve behavioral outcomes in the host by modulating microbial, metabolomic, and immunological profiles. In this review, we discuss the pathways of brain-to-gut or gut-to-brain signaling and summarize the role of gut microbiota and microbial metabolites across the lifespan and in disease. We highlight recent studies investigating 1) microbial changes with aging; 2) how aging of the maternal microbiome can affect offspring health; and 3) the contribution of the microbiome to both chronic age-related diseases (e.g., Parkinson's disease, Alzheimer's disease and cerebral amyloidosis), and acute brain injury, including ischemic stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Romeesa Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Claudia M. Di Gesù
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
115
|
Chen Y, Li Y, Fan Y, Chen S, Chen L, Chen Y, Chen Y. Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice. Gut Microbes 2024; 16:2302310. [PMID: 38261437 PMCID: PMC10807476 DOI: 10.1080/19490976.2024.2302310] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024] Open
Abstract
The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the "microbiota - metabolite - brain axis" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched Dubosiella affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that Dubosiella enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria Erysipelatoclostridium and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.
Collapse
Affiliation(s)
- Yijing Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yinhu Li
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yingying Fan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Li Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
116
|
Pait MC, Kaye SD, Su Y, Kumar A, Singh S, Gironda SC, Vincent S, Anwar M, Carroll CM, Snipes JA, Lee J, Furdui CM, Deep G, Macauley SL. Novel method for collecting hippocampal interstitial fluid extracellular vesicles (EV ISF ) reveals sex-dependent changes in microglial EV proteome in response to Aβ pathology. J Extracell Vesicles 2024; 13:e12398. [PMID: 38191961 PMCID: PMC10774707 DOI: 10.1002/jev2.12398] [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: 03/10/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Brain-derived extracellular vesicles (EVs) play an active role in Alzheimer's disease (AD), relaying important physiological information about their host tissues. The internal cargo of EVs is protected from degradation, making EVs attractive AD biomarkers. However, it is unclear how circulating EVs relate to EVs isolated from disease-vulnerable brain regions. We developed a novel method for collecting EVs from the hippocampal interstitial fluid (ISF) of live mice. EVs (EVISF ) were isolated via ultracentrifugation and characterized by nanoparticle tracking analysis, immunogold labelling, and flow cytometry. Mass spectrometry and proteomic analyses were performed on EVISF cargo. EVISF were 40-150 nm in size and expressed CD63, CD9, and CD81. Using a model of cerebral amyloidosis (e.g., APPswe, PSEN1dE9 mice), we found protein concentration increased but protein diversity decreased with Aβ deposition. Genotype, age, and Aβ deposition modulated proteostasis- and immunometabolic-related pathways. Changes in the microglial EVISF proteome were sexually dimorphic and associated with a differential response of plaque associated microglia. We found that female APP/PS1 mice have more amyloid plaques, less plaque associated microglia, and a less robust- and diverse- EVISF microglial proteome. Thus, in vivo microdialysis is a novel technique for collecting EVISF and offers a unique opportunity to explore the role of EVs in AD.
Collapse
Affiliation(s)
- Morgan C. Pait
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Sarah D. Kaye
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Yixin Su
- Department of Cancer BiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Ashish Kumar
- Department of Cancer BiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Sangeeta Singh
- Department of Cancer BiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Stephen C. Gironda
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samantha Vincent
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Maria Anwar
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Caitlin M. Carroll
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - James Andy Snipes
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jingyun Lee
- Department of Internal MedicineSection on Molecular MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Proteomics and Metabolomics Shared ResourceWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Cristina M. Furdui
- Department of Internal MedicineSection on Molecular MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Proteomics and Metabolomics Shared ResourceWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Atrium Health Wake Forest Baptist Comprehensive Cancer CenterWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Gagan Deep
- Department of Cancer BiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Atrium Health Wake Forest Baptist Comprehensive Cancer CenterWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Center for Research on Substance Use and AddictionWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- J Paul Sticht Center for Healthy Aging and Alzheimer's PreventionWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Shannon L. Macauley
- Department of Physiology & PharmacologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- J Paul Sticht Center for Healthy Aging and Alzheimer's PreventionWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Internal MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Alzheimer's Disease Research CenterWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Center for Diabetes and MetabolismWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Cardiovascular Sciences CenterWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
117
|
André S, Verteneuil S, Ris L, Kahvecioglu ZC, Nonclercq D, De Winter J, Vander Elst L, Laurent S, Muller RN, Burtea C. Modulation of Cytosolic Phospholipase A2 as a Potential Therapeutic Strategy for Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1395-1426. [PMID: 38225969 PMCID: PMC10789292 DOI: 10.3233/adr-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 01/17/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder lacking any curative treatment up to now. Indeed, actual medication given to the patients alleviates only symptoms. The cytosolic phospholipase A2 (cPLA2-IVA) appears as a pivotal player situated at the center of pathological pathways leading to AD and its inhibition could be a promising therapeutic approach. Objective A cPLA2-IVA inhibiting peptide was identified in the present work, aiming to develop an original therapeutic strategy. Methods We targeted the cPLA2-IVA using the phage display technology. The hit peptide PLP25 was first validated in vitro (arachidonic acid dosage [AA], cPLA2-IVA cellular translocation) before being tested in vivo. We evaluated spatial memory using the Barnes maze, amyloid deposits by MRI and immunohistochemistry (IHC), and other important biomarkers such as the cPLA2-IVA itself, the NMDA receptor, AβPP and tau by IHC after i.v. injection in APP/PS1 mice. Results Showing a high affinity for the C2 domain of this enzyme, the peptide PLP25 exhibited an inhibitory effect on cPLA2-IVA activity by blocking its binding to its substrate, resulting in a decreased release of AA. Coupled to a vector peptide (LRPep2) in order to optimize brain access, we showed an improvement of cognitive abilities of APP/PS1 mice, which also exhibited a decreased number of amyloid plaques, a restored expression of cPLA2-IVA, and a favorable effect on NMDA receptor expression and tau protein phosphorylation. Conclusions cPLA2-IVA inhibition through PLP25 peptide could be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Séverine André
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sébastien Verteneuil
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Research Institute for Health Science and Technologies, Mons, Belgium
| | - Zehra-Cagla Kahvecioglu
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory (SMOs), University of Mons-UMONS, Mons, Belgium
| | - Luce Vander Elst
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Carmen Burtea
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| |
Collapse
|
118
|
Katsuki F, Spratt TJ, Brown RE, Basheer R, Uygun DS. Sleep-Deep-Net learns sleep wake scoring from the end-user and completes each record in their style. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573151. [PMID: 38187568 PMCID: PMC10769368 DOI: 10.1101/2023.12.22.573151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Sleep-wake scoring is a time-consuming, tedious but essential component of clinical and pre-clinical sleep research. Sleep scoring is even more laborious and challenging in rodents due to the smaller EEG amplitude differences between states and the rapid state transitions which necessitate scoring in shorter epochs. Although many automated rodent sleep scoring methods exist, they do not perform as well when scoring new data sets, especially those which involve changes in the EEG/EMG profile. Thus, manual scoring by expert scorers remains the gold-standard. Here we take a different approach to this problem by using a neural network to accelerate the scoring of expert scorers. Sleep-Deep-Net (SDN) creates a bespoke deep convolution neural network model for individual electroencephalographic or local-field-potential records via transfer learning of GoogleNet, by learning from a small subset of manual scores of each EEG/LFP record as provided by the end-user. SDN then automates scoring of the remainder of the EEG/LFP record. A novel REM scoring correction procedure further enhanced accuracy. SDN reliably scores EEG and LFP data and retains sleep-wake architecture in wild-type mice, in sleep induced by the hypnotic zolpidem, in a mouse model of Alzheimer's disease and in a genetic knock-down study, when compared to manual scoring. SDN reduced manual scoring time to 1/12. Since SDN uses transfer learning on each independent recording, it is not biased by previously scored existing data sets. Thus, we find SDN performs well when used on signals altered by a drug, disease model or genetic modification.
Collapse
Affiliation(s)
- Fumi Katsuki
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - Tristan J Spratt
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - Ritchie E Brown
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - Radhika Basheer
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - David S Uygun
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| |
Collapse
|
119
|
Ramirez S, Koerich S, Astudillo N, De Gregorio N, Al-Lahham R, Allison T, Rocha NP, Wang F, Soto C. Plasma Exchange Reduces Aβ Levels in Plasma and Decreases Amyloid Plaques in the Brain in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:17087. [PMID: 38069410 PMCID: PMC10706894 DOI: 10.3390/ijms242317087] [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: 10/28/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, characterized by the abnormal accumulation of protein aggregates in the brain, known as neurofibrillary tangles and amyloid-β (Aβ) plaques. It is believed that an imbalance between cerebral and peripheral pools of Aβ may play a relevant role in the deposition of Aβ aggregates. Therefore, in this study, we aimed to evaluate the effect of the removal of Aβ from blood plasma on the accumulation of amyloid plaques in the brain. We performed monthly plasma exchange with a 5% mouse albumin solution in the APP/PS1 mouse model from 3 to 7 months old. At the endpoint, total Aβ levels were measured in the plasma, and soluble and insoluble brain fractions were analyzed using ELISA. Brains were also analyzed histologically for amyloid plaque burden, plaque size distributions, and gliosis. Our results showed a reduction in the levels of Aβ in the plasma and insoluble brain fractions. Interestingly, histological analysis showed a reduction in thioflavin-S (ThS) and amyloid immunoreactivity in the cortex and hippocampus, accompanied by a change in the size distribution of amyloid plaques, and a reduction in Iba1-positive cells. Our results provide preclinical evidence supporting the relevance of targeting Aβ in the periphery and reinforcing the potential use of plasma exchange as an alternative non-pharmacological strategy for slowing down AD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Claudio Soto
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA; (S.R.); (S.K.); (N.A.); (N.D.G.); (R.A.-L.); (T.A.); (N.P.R.); (F.W.)
| |
Collapse
|
120
|
Le L, Feidler AM, Li H, Kara-Pabani K, Lamantia C, O'Banion MK, Majewska KA. Noradrenergic signaling controls Alzheimer's disease pathology via activation of microglial β2 adrenergic receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569564. [PMID: 38106167 PMCID: PMC10723313 DOI: 10.1101/2023.12.01.569564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
In Alzheimer's disease (AD) pathophysiology, plaque and tangle accumulation trigger an inflammatory response that mounts positive feed-back loops between inflammation and protein aggregation, aggravating neurite damage and neuronal death. One of the earliest brain regions to undergo neurodegeneration is the locus coeruleus (LC), the predominant site of norepinephrine (NE) production in the central nervous system (CNS). In animal models of AD, dampening the impact of noradrenergic signaling pathways, either through administration of beta blockers or pharmacological ablation of the LC, heightened neuroinflammation through increased levels of pro-inflammatory mediators. Since microglia are the resident immune cells of the CNS, it is reasonable to postulate that they are responsible for translating the loss of NE tone into exacerbated disease pathology. Recent findings from our lab demonstrated that noradrenergic signaling inhibits microglia dynamics via β2 adrenergic receptors (β2ARs), suggesting a potential anti-inflammatory role for microglial β2AR signaling. Thus, we hypothesize that microglial β2 adrenergic signaling is progressively impaired during AD progression, which leads to the chronic immune vigilant state of microglia that worsens disease pathology. First, we characterized changes in microglial β2AR signaling as a function of amyloid pathology. We found that LC neurons and their projections degenerate early and progressively in the 5xFAD mouse model of AD; accompanied by mild decrease in the levels of norepinephrine and its metabolite normetanephrine. Interestingly, while 5xFAD microglia, especially plaque-associated microglia, significant downregulated β2AR gene expression early in amyloid pathology, they did not lose their responsiveness to β2AR stimulation. Most importantly, we demonstrated that specific microglial β2AR deletion worsened disease pathology while chronic β2AR stimulation resulted in attenuation of amyloid pathology and associated neuritic damage, suggesting microglial β2AR might be used as potential therapeutic target to modify AD pathology.
Collapse
Affiliation(s)
- L Le
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
| | - A M Feidler
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
| | - H Li
- Medical Scientist Training Program, University of Rochester, Rochester NY
| | - K Kara-Pabani
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
| | - C Lamantia
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
| | - M K O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
| | - K A Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY
- Center for Visual Science, University of Rochester, Rochester NY
| |
Collapse
|
121
|
Zielinski M, Peralta Reyes FS, Gremer L, Schemmert S, Frieg B, Schäfer LU, Willuweit A, Donner L, Elvers M, Nilsson LNG, Syvänen S, Sehlin D, Ingelsson M, Willbold D, Schröder GF. Cryo-EM of Aβ fibrils from mouse models find tg-APP ArcSwe fibrils resemble those found in patients with sporadic Alzheimer's disease. Nat Neurosci 2023; 26:2073-2080. [PMID: 37973869 PMCID: PMC10689242 DOI: 10.1038/s41593-023-01484-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/06/2023] [Indexed: 11/19/2023]
Abstract
The use of transgenic mice displaying amyloid-β (Aβ) brain pathology has been essential for the preclinical assessment of new treatment strategies for Alzheimer's disease. However, the properties of Aβ in such mice have not been systematically compared to Aβ in the brains of patients with Alzheimer's disease. Here, we determined the structures of nine ex vivo Aβ fibrils from six different mouse models by cryogenic-electron microscopy. We found novel Aβ fibril structures in the APP/PS1, ARTE10 and tg-SwDI models, whereas the human type II filament fold was found in the ARTE10, tg-APPSwe and APP23 models. The tg-APPArcSwe mice showed an Aβ fibril whose structure resembles the human type I filament found in patients with sporadic Alzheimer's disease. A detailed assessment of the Aβ fibril structure is key to the selection of adequate mouse models for the preclinical development of novel plaque-targeting therapeutics and positron emission tomography imaging tracers in Alzheimer's disease.
Collapse
Affiliation(s)
- Mara Zielinski
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany
| | | | - Lothar Gremer
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany.
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Sarah Schemmert
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany
| | - Benedikt Frieg
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany
| | - Luisa U Schäfer
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich, Jülich, Germany
| | - Lili Donner
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Lars N G Nilsson
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine and Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dieter Willbold
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany.
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Gunnar F Schröder
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, Jülich, Germany.
- JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, Jülich, Germany.
- Physics Department, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
122
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [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: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
123
|
Bodart-Santos V, Pinheiro LS, da Silva-Junior AJ, Froza RL, Ahrens R, Gonçalves RA, Andrade MM, Chen Y, Alcantara CDL, Grinberg LT, Leite REP, Ferreira ST, Fraser PE, De Felice FG. Alzheimer's disease brain-derived extracellular vesicles reveal altered synapse-related proteome and induce cognitive impairment in mice. Alzheimers Dement 2023; 19:5418-5436. [PMID: 37204850 DOI: 10.1002/alz.13134] [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: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) have been implicated in the spread of neuropathology in Alzheimer's disease (AD), but their involvement in behavioral outcomes linked to AD remains to be determined. METHODS EVs isolated from post mortem brain tissue from control, AD, or frontotemporal dementia (FTD) donors, as well as from APP/PS1 mice, were injected into the hippocampi of wild-type (WT) or a humanized Tau mouse model (hTau/mTauKO). Memory tests were carried out. Differentially expressed proteins in EVs were assessed by proteomics. RESULTS Both AD-EVs and APP/PS1-EVs trigger memory impairment in WT mice. We further demonstrate that AD-EVs and FTD-EVs carry Tau protein, present altered protein composition associated with synapse regulation and transmission, and trigger memory impairment in hTau/mTauKO mice. DISCUSSION Results demonstrate that AD-EVs and FTD-EVs have negative impacts on memory in mice and suggest that, in addition to spreading pathology, EVs may contribute to memory impairment in AD and FTD. HIGHLIGHTS Aβ was detected in EVs from post mortem AD brain tissue and APP/PS1 mice. Tau was enriched in EVs from post mortem AD, PSP and FTD brain tissue. AD-derived EVs and APP/PS1-EVs induce cognitive impairment in wild-type (WT) mice. AD- and FTD-derived EVs induce cognitive impairment in humanized Tau mice. Proteomics findings associate EVs with synapse dysregulation in tauopathies.
Collapse
Affiliation(s)
- Victor Bodart-Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Lisandra S Pinheiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J da Silva-Junior
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rudimar L Froza
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Rosemary Ahrens
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Rafaella A Gonçalves
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
| | - Mayara M Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yan Chen
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Carolina de Lima Alcantara
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lea T Grinberg
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
- Memory and Aging Center, Department of Neurology and Pathology, University of California San Francisco, San Francisco, California, USA
| | - Renata E P Leite
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
- D'OR Institute for Research and Education, Rio de Janeiro, Brazil
| |
Collapse
|
124
|
Zhao Q, Maci M, Miller MR, Zhou H, Zhang F, Algamal M, Lee YF, Hou SS, Perle SJ, Le H, Russ AN, Lo EH, Gerashchenko D, Gomperts SN, Bacskai BJ, Kastanenka KV. Sleep restoration by optogenetic targeting of GABAergic neurons reprograms microglia and ameliorates pathological phenotypes in an Alzheimer's disease model. Mol Neurodegener 2023; 18:93. [PMID: 38041158 PMCID: PMC10693059 DOI: 10.1186/s13024-023-00682-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/17/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) patients exhibit memory disruptions and profound sleep disturbances, including disruption of deep non-rapid eye movement (NREM) sleep. Slow-wave activity (SWA) is a major restorative feature of NREM sleep and is important for memory consolidation. METHODS We generated a mouse model where GABAergic interneurons could be targeted in the presence of APPswe/PS1dE9 (APP) amyloidosis, APP-GAD-Cre mice. An electroencephalography (EEG) / electromyography (EMG) telemetry system was used to monitor sleep disruptions in these animals. Optogenetic stimulation of GABAergic interneurons in the anterior cortex targeted with channelrhodopsin-2 (ChR2) allowed us to examine the role GABAergic interneurons play in sleep deficits. We also examined the effect of optogenetic stimulation on amyloid plaques, neuronal calcium as well as sleep-dependent memory consolidation. In addition, microglial morphological features and functions were assessed using confocal microscopy and flow cytometry. Finally, we performed sleep deprivation during optogenetic stimulation to investigate whether sleep restoration was necessary to slow AD progression. RESULTS APP-GAD-Cre mice exhibited impairments in sleep architecture including decreased time spent in NREM sleep, decreased delta power, and increased sleep fragmentation compared to nontransgenic (NTG) NTG-GAD-Cre mice. Optogenetic stimulation of cortical GABAergic interneurons increased SWA and rescued sleep impairments in APP-GAD-Cre animals. Furthermore, it slowed AD progression by reducing amyloid deposition, normalizing neuronal calcium homeostasis, and improving memory function. These changes were accompanied by increased numbers and a morphological transformation of microglia, elevated phagocytic marker expression, and enhanced amyloid β (Aβ) phagocytic activity of microglia. Sleep was necessary for amelioration of pathophysiological phenotypes in APP-GAD-Cre mice. CONCLUSIONS In summary, our study shows that optogenetic targeting of GABAergic interneurons rescues sleep, which then ameliorates neuropathological as well as behavioral deficits by increasing clearance of Aβ by microglia in an AD mouse model.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Megi Maci
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Morgan R Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Heng Zhou
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Fang Zhang
- Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Moustafa Algamal
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Steven S Hou
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Stephen J Perle
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Hoang Le
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Alyssa N Russ
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Eng H Lo
- Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Dmitry Gerashchenko
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA, 02132, USA
| | - Stephen N Gomperts
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brian J Bacskai
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ksenia V Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
125
|
Yuan L, Xie L, Zhang H, Zhang Y, Wei Y, Feng J, Cui L, Tian R, Feng J, Yu D, Lv C. Low-dose IL-2 Treatment Rescues Cognitive Deficits by Repairing the Imbalance Between Treg and Th17 Cells at the Middle Alzheimer's Disease Stage. J Neuroimmune Pharmacol 2023; 18:674-689. [PMID: 37962810 DOI: 10.1007/s11481-023-10090-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2023] [Indexed: 11/15/2023]
Abstract
Multiple studies highlight the role of effector and regulatory CD4+T cells in the pathophysiology of Alzheimer's disease, and foster low-dose IL-2 treatment which induces regulatory CD4+T (Treg) cells expansion and activation as a promising strategy for its treatment. However, studies demonstrating discrepant Treg functions in AD have been reported. In addition, a compromised immune system associated with aging may substantially impact on these processes. Here, we report that there is an altered balance of activity between Treg cells and IL-17-producing helper T (Th17) cells in periphery and brain of APP/PS1 mice along the disease progression. A dramatic loss of the healthy balance of activity between Treg and Th17 cells was found at the middle disease stage. While peripheral low-dose recombinant human IL-2 administration could selectively modulate the abundance of Treg cells and repair the imbalance between Treg and Th17 subsets at the middle disease stage. We further show that modulation of peripheral immune balance through low-dose IL-2 treatment reduces the neuro-inflammation and increases numbers of plaque-associated microglia, accompanied by marked reduction of Aβ plaque deposition and slower cognitive declines in APP/PS1 mice at the middle disease stage. Our study highlights the therapeutic potential of repurposed IL-2 for innovative immunotherapy based on modulation of the homeostasis of CD4+T cell subsets in Alzheimer's disease at the middle disease stage.
Collapse
Affiliation(s)
- Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Enshi, Hubei, China
| | - Lei Xie
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hao Zhang
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yu Zhang
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yunbo Wei
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
| | - Jinhong Feng
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
| | - Li Cui
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China
| | - Rui Tian
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Enshi, Hubei, China
| | - Jia Feng
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Enshi, Hubei, China
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Cui Lv
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), #19 Keyuan Road, Lixia District, Jinan, 250014, Shandong, China.
| |
Collapse
|
126
|
Purro SA, Farmer M, Noble E, Sarell CJ, Powell M, Yip D, Giggins L, Zakka L, Thomas DX, Farrow M, Nicoll AJ, Walsh D, Collinge J. Two mouse models of Alzheimer's disease accumulate amyloid at different rates and have distinct Aβ oligomer profiles unaltered by ablation of cellular prion protein. PLoS One 2023; 18:e0294465. [PMID: 37976283 PMCID: PMC10655998 DOI: 10.1371/journal.pone.0294465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Oligomers formed from monomers of the amyloid β-protein (Aβ) are thought to be central to the pathogenesis of Alzheimer's disease (AD). Unsurprisingly for a complex disease, current mouse models of AD fail to fully mimic the clinical disease in humans. Moreover, results obtained in a given mouse model are not always reproduced in a different model. Cellular prion protein (PrPC) is now an established receptor for Aβ oligomers. However, studies of the Aβ-PrPC interaction in different mouse models have yielded contradictory results. Here we performed a longitudinal study assessing a range of biochemical and histological features in the commonly used J20 and APP-PS1 mouse models. Our analysis demonstrated that PrPC ablation had no effect on amyloid accumulation or oligomer production. However, we found that APP-PS1 mice had higher levels of oligomers, that these could bind to recombinant PrPC, and were recognised by the OC antibody which distinguishes parallel, in register fibrils. On the other hand, J20 mice had a lower level of Aβ oligomers, which did not interact with PrPC when tested in vitro and were OC-negative. These results suggest the two mouse models produce diverse Aβ assemblies that could interact with different targets, highlighting the necessity to characterise the conformation of the Aβ oligomers concomitantly with the toxic cascade elicited by them. Our results provide an explanation for the apparent contradictory results found in APP-PS1 mice and the J20 mouse line in regards to Aβ toxicity mediated by PrPC.
Collapse
Affiliation(s)
- Silvia A. Purro
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Michael Farmer
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Elizabeth Noble
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Claire J. Sarell
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Megan Powell
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Daniel Yip
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Lauren Giggins
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Leila Zakka
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - David X. Thomas
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Mark Farrow
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Andrew J. Nicoll
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| | - Dominic Walsh
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - John Collinge
- MRC Prion Unit at UCL, Institute of Prion diseases, University College London, London, United Kingdom
| |
Collapse
|
127
|
Lee H, Kim Y, Aziz H, Kang DM, Lee J, Lee S, Jung S, Hyeon S, Choo H, Nam G, Kim YK, Lim S, Min SJ. Synthesis and biological evaluation of indane-based fluorescent probes for detection of amyloid-β aggregates in Alzheimer's disease. Bioorg Med Chem 2023; 95:117513. [PMID: 37931520 DOI: 10.1016/j.bmc.2023.117513] [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: 09/26/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
In this article, the development of fluorescent imaging probes for the detection of Alzheimer's disease (AD)-associated protein aggregates is described. Indane derivatives with a donor-π-acceptor (D-π-A) structure were designed and synthesized. The probes were evaluated for their ability to bind to β-amyloid (Aβ) protein aggregates, which are a key pathological hallmark of AD. The results showed that several probes exhibited significant changes in fluorescence intensity at wavelengths greater than 600 nm when they were bound to Aβ aggregates compared to the Aβ monomeric form. Among the tested probes, four D-π-A type indane derivatives showed promising binding selectivity to Aβ aggregates over non-specific proteins such as bovine serum albumin (BSA). The molecular docking study showed that our compounds were appropriately located along the Aβ fibril axis through the hydrophobic tunnel structure. Further analysis revealed that the most active compound having dimethylaminopyridyl group as an election donor and dicyano group as an electron acceptor could effectively stain Aβ plaques in brain tissue samples from AD transgenic mice. These findings suggest that our indane-based compounds have the potential to serve as fluorescent probes for the detection and monitoring of Aβ aggregation in AD.
Collapse
Affiliation(s)
- Hyunseung Lee
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Yihoon Kim
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hira Aziz
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Dong-Min Kang
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Jaewoon Lee
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea; Center for Bionano Intelligence Education and Research, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Sujin Lee
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea; Center for Bionano Intelligence Education and Research, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Sunhwa Jung
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea; Center for Bionano Intelligence Education and Research, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Suyeon Hyeon
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyunah Choo
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Ghilsoo Nam
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Yun Kyung Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Sungsu Lim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Sun-Joon Min
- Department of Applied Chemistry, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea; Center for Bionano Intelligence Education and Research, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea; Department of Chemical & Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea.
| |
Collapse
|
128
|
Liu CC, Wang N, Chen Y, Inoue Y, Shue F, Ren Y, Wang M, Qiao W, Ikezu TC, Li Z, Zhao J, Martens Y, Doss SV, Rosenberg CL, Jeevaratnam S, Jia L, Raulin AC, Qi F, Zhu Y, Alnobani A, Knight J, Chen Y, Linares C, Kurti A, Fryer JD, Zhang B, Wu LJ, Kim BYS, Bu G. Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer's disease. Nat Immunol 2023; 24:1854-1866. [PMID: 37857825 PMCID: PMC11980647 DOI: 10.1038/s41590-023-01640-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 09/06/2023] [Indexed: 10/21/2023]
Abstract
Microglial involvement in Alzheimer's disease (AD) pathology has emerged as a risk-determining pathogenic event. While apolipoprotein E (APOE) is known to modify AD risk, it remains unclear how microglial apoE impacts brain cognition and AD pathology. Here, using conditional mouse models expressing apoE isoforms in microglia and central nervous system-associated macrophages (CAMs), we demonstrate a cell-autonomous effect of apoE3-mediated microglial activation and function, which are negated by apoE4. Expression of apoE3 in microglia/CAMs improves cognitive function, increases microglia surrounding amyloid plaque and reduces amyloid pathology and associated toxicity, whereas apoE4 expression either compromises or has no effects on these outcomes by impairing lipid metabolism. Single-cell transcriptomic profiling reveals increased antigen presentation and interferon pathways upon apoE3 expression. In contrast, apoE4 expression downregulates complement and lysosomal pathways, and promotes stress-related responses. Moreover, in the presence of mouse endogenous apoE, microglial apoE4 exacerbates amyloid pathology. Finally, we observed a reduction in Lgals3-positive responsive microglia surrounding amyloid plaque and an increased accumulation of lipid droplets in APOE4 human brains and induced pluripotent stem cell-derived microglia. Our findings establish critical isoform-dependent effects of microglia/CAM-expressed apoE in brain function and the development of amyloid pathology, providing new insight into how apoE4 vastly increases AD risk.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA.
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yuanxin Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuka Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sydney V Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Alla Alnobani
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
129
|
Lee J, Weerasinghe-Mudiyanselage PDE, Kim B, Kang S, Kim JS, Moon C. Particulate matter exposure and neurodegenerative diseases: A comprehensive update on toxicity and mechanisms. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115565. [PMID: 37832485 DOI: 10.1016/j.ecoenv.2023.115565] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/30/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
Exposure to particulate matter (PM) has been associated with a range of health impacts, including neurological abnormalities that affect neurodevelopment, neuroplasticity, and behavior. Recently, there has been growing interest in investigating the possible relationship between PM exposure and the onset and progression of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. However, the precise mechanism by which PM affects neurodegeneration is still unclear, even though several epidemiological and animal model studies have provided mechanistic insights. This article presents a review of the current research on the neurotoxicity of PM and its impact on neurodegenerative diseases. This review summarizes findings from epidemiological and animal model studies collected through searches in Google Scholar, PubMed, Web of Science, and Scopus. This review paper also discusses the reported effects of PM exposure on the central nervous system and highlights research gaps and future directions. The information presented in this review may inform public health policies aimed at reducing PM exposure and may contribute to the development of new treatments for neurodegenerative diseases. Further mechanistic and therapeutic research will be needed to fully understand the relationship between PM exposure and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jeongmin Lee
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea
| | - Poornima D E Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea
| | - Bohye Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
130
|
Yuan J, He M, Dai X, Huo Q, Chang P, Zhang J, Wang S, Sun Y. 3,6'-Disinapoyl sucrose alleviates cognitive deficits in APP/PS1 transgenic mice. J Neurophysiol 2023; 130:1174-1182. [PMID: 37702542 DOI: 10.1152/jn.00067.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/21/2023] [Accepted: 09/01/2023] [Indexed: 09/14/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with insidious onset and progressive development. There is an urgent need to find drugs that prevent and slow AD progression. We focus our attention on 3,6'-disinapoyl sucrose (DISS), an oligosaccharide with antidepressant and antioxidant activities. In this work, APP/PS1 transgenic mice were used to explore the neuroprotective impact of DISS to provide new applications for prevention and therapy of AD. This study aims to assess DISS's neuroprotective impact on learning and memory deficits in APP/PS1 transgenic mice using behavioral tests (Morris water maze, novel object recognition test, and passive avoidance test). Morphological alterations of hippocampus neurons were observed by Nissl staining and neuronal apoptosis was assessed by TUNEL assay. By using ELISA, the expressions of inflammatory factors were evaluated, and Western blotting was used to measure the protein expressions of neuron-related regulators in the hippocampus. DISS significantly ameliorated the cognitive disorder in APP/PS1 transgenic mice, reduced apoptosis by decreasing the ratio of Bax/B-cell lymphoma/leukemia-2 (Bcl-2) in hippocampal neurons, and restored the abnormal secretion of inflammatory factors (IL-2, TNF-α, IL-1β, and IL-6). Moreover, the gavage of high-dose DISS can boost the expressions of CREB/brain-derived neurotrophic factor (BDNF). Overall, our results indicate that DISS improves cognitive function in APP/PS1 transgenic mice by inhibiting neural apoptosis and activating the CREB/BDNF signal pathway.NEW & NOTEWORTHY In this study, for the first time, DISS was used in APP/PS1 transgenic mice to explore its neuroprotective effect. After gavage DISS for 1 mo, the impairment of learning and spatial memory ability and the loss of neurons in APP/PS1 mice were alleviated. DISS reduced a neuroprotective effect in AD mice via decreasing neuronal apoptosis, enhancing the expressions of CREB phosphorylation and BDNF, pointing to DISS as a new therapeutic target for AD.
Collapse
Affiliation(s)
- Jiaqi Yuan
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Mengjie He
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qing Huo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Ping Chang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Jing Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| |
Collapse
|
131
|
Quintela-López T, Lezmy J. Homeostatic plasticity of axonal excitable sites in Alzheimer's disease. Front Neurosci 2023; 17:1277251. [PMID: 37937068 PMCID: PMC10626477 DOI: 10.3389/fnins.2023.1277251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Affiliation(s)
| | - Jonathan Lezmy
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
132
|
Liu C, Cardenas-Rivera A, Teitelbaum S, Birmingham A, Alfadhel M, Yaseen MA. Neuroinflammation increases oxygen extraction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562353. [PMID: 37905082 PMCID: PMC10614808 DOI: 10.1101/2023.10.16.562353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Neuroinflammation, impaired metabolism, and hypoperfusion are fundamental pathological hallmarks of early Alzheimer's disease (AD). Numerous studies have asserted a close association between neuroinflammation and disrupted cerebral energetics. During AD progression and other neurodegenerative disorders, a persistent state of chronic neuroinflammation reportedly exacerbates cytotoxicity and potentiates neuronal death. Here, we assessed the impact of a neuroinflammatory challenge on metabolic demand and microvascular hemodynamics in the somatosensory cortex of an AD mouse model. We utilized in vivo 2-photon microscopy and the phosphorescent oxygen sensor Oxyphor 2P to measure partial pressure of oxygen (pO2) and capillary red blood cell flux in cortical microvessels of awake mice. Intravascular pO2 and capillary RBC flux measurements were performed in 8-month-old APPswe/PS1dE9 mice and wildtype littermates on days 0, 7, and 14 of a 14-day period of lipopolysaccaride-induced neuroinflammation. Before the induced inflammatory challenge, AD mice demonstrated reduced metabolic demand but similar capillary red blood cell flux as their wild type counterparts. Neuroinflammation provoked significant reductions in cerebral intravascular oxygen levels and elevated oxygen extraction in both animal groups, without significantly altering red blood cell flux in capillaries. This study provides evidence that neuroinflammation alters cerebral oxygen demand at the early stages of AD without substantially altering vascular oxygen supply. The results will guide our understanding of neuroinflammation's influence on neuroimaging biomarkers for early AD diagnosis.
Collapse
|
133
|
Zheng X, Zhao D, Jin Y, Liu Y, Liu D. Role of the NLRP3 inflammasome in gynecological disease. Biomed Pharmacother 2023; 166:115393. [PMID: 37660654 DOI: 10.1016/j.biopha.2023.115393] [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: 07/04/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023] Open
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Collapse
Affiliation(s)
- Xu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Dan Zhao
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Yang Liu
- Acupuncture department,Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
134
|
Papazoglou A, Henseler C, Weickhardt S, Daubner J, Schiffer T, Broich K, Hescheler J, Ehninger D, Scholl C, Haenisch B, Sachinidis A, Weiergräber M. Whole genome transcriptome data from the WT cortex and hippocampus of female and male control and APP/PS1 Alzheimer's disease mice. Data Brief 2023; 50:109594. [PMID: 37767130 PMCID: PMC10520298 DOI: 10.1016/j.dib.2023.109594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
A variety of Alzheimer disease (AD) mouse models has been established and characterized within the last decades. These models are generated to meet the principal criteria of AD isomorphism, homology and predictability to a maximum extent. To get an integrative view of the sophisticated etiopathogenesis of AD, whole genome transcriptome data analysis turns out to be indispensable. Here, we present a microarray-based transcriptome data collection based on RNA extracted from the retrosplenial (RS) cortex and the hippocampus of APP/PS1 AD mice and control animals. Experimental animals were age matched and importantly, both sexes were considered separately. Isolated RNA was purified, quantified und quality controlled prior to the hybridization procedure with SurePrint G3 Mouse Gene Expression v2 8 × 60K microarrays. Following immunofluorescent measurement und preprocessing/extraction of image data, raw transcriptome data were uploaded including differentially expressed gene candidates and related fold changes in APP/PS1 AD mice and controls. Our data allow further insight into alterations in gene transcript levels in APP/PS1 AD mice compared to controls and enable the reader/user to carry out complex transcriptome analysis to characterize potential age-, sex- and brain-region-specific alterations in e.g., neuroinflammatory, immunological, neurodegenerative and ion channel pathways.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Sandra Weickhardt
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Teresa Schiffer
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Jürgen Hescheler
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Dan Ehninger
- Translational Biogerontology, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Catharina Scholl
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
- Center for Translational Medicine, Medical Faculty, University of Bonn, 53113 Bonn, Germany
| | - Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| |
Collapse
|
135
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley KB, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. GeroScience 2023; 45:3019-3043. [PMID: 37393197 PMCID: PMC10643718 DOI: 10.1007/s11357-023-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023] Open
Abstract
Major histocompatibility complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses, but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here, we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating ribosome affinity purification-qPCR analysis of 3-6- and 18-22-month-old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m, H2-D1, H2-K1, H2-M3, H2-Q6, and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I-binding leukocyte immunoglobulin-like (Lilrs) and paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell -autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A, suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M Kellogg
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Adeline H Machalinski
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Hunter L Porter
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Harris E Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
136
|
Na D, Yang Y, Xie L, Piekna-Przybylska D, Bunn D, Shamambo M, White P. Neuroinflammation in a Mouse Model of Alzheimer's Disease versus Auditory Dysfunction: Machine Learning Interpretation and Analysis. RESEARCH SQUARE 2023:rs.3.rs-3370200. [PMID: 37841847 PMCID: PMC10571613 DOI: 10.21203/rs.3.rs-3370200/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Background Auditory dysfunction, including central auditory hyperactivity, hearing loss and hearing in noise deficits, has been reported in 5xFAD Alzheimer's disease (AD) mice, suggesting a causal relationship between amyloidosis and auditory dysfunction. Central auditory hyperactivity correlated in time with small amounts of plaque deposition in the inferior colliculus and medial geniculate body, which are the auditory midbrain and thalamus, respectively. Neuroinflammation has been associated with excitation to inhibition imbalance in the central nervous system, and therefore has been proposed as a link between central auditory hyperactivity and AD in our previous report. However, neuroinflammation in the auditory pathway has not been investigated in mouse amyloidosis models. Methods Machine learning was used to classify the previously obtained auditory brainstem responses (ABRs) from 5xFAD mice and their wild type (WT) littermates. Neuroinflammation was assessed in six auditory-related regions of the cortex, thalamus, and brainstem. Cochlear pathology was assessed in cryosection and whole mount. Behavioral changes were assessed with fear conditioning, open field testing and novel objection recognition. Results Reliable machine learning classification of 5xFAD and WT littermate ABRs were achieved for 6M and 12M, but not 3M. The top features for accurate classification at 6 months of age were characteristics of Waves IV and V. Microglial and astrocytic activation were pronounced in 5xFAD inferior colliculus and medial geniculate body at 6 months, two neural centers that are thought to contribute to these waves. Lower regions of the brainstem were unaffected, and cortical auditory centers also displayed inflammation beginning at 6 months. No losses were seen in numbers of spiral ganglion neurons (SGNs), auditory synapses, or efferent synapses in the cochlea. 5xFAD mice had reduced responses to tones in fear conditioning compared to WT littermates beginning at 6 months. Conclusions Serial use of ABR in early AD patients represents a promising approach for early and inexpensive detection of neuroinflammation in higher auditory brainstem processing centers. As changes in auditory processing are strongly linked to AD progression, central auditory hyperactivity may serve as a biomarker for AD progression and/or stratify AD patients into distinct populations.
Collapse
Affiliation(s)
| | | | - Li Xie
- University of Rochester Medical Center
| | | | | | | | | |
Collapse
|
137
|
Toader C, Eva L, Covache-Busuioc RA, Costin HP, Glavan LA, Corlatescu AD, Ciurea AV. Unraveling the Multifaceted Role of the Golgi Apparatus: Insights into Neuronal Plasticity, Development, Neurogenesis, Alzheimer's Disease, and SARS-CoV-2 Interactions. Brain Sci 2023; 13:1363. [PMID: 37891732 PMCID: PMC10605100 DOI: 10.3390/brainsci13101363] [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: 08/01/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
This article critically evaluates the multifunctional role of the Golgi apparatus within neurological paradigms. We succinctly highlight its influence on neuronal plasticity, development, and the vital trafficking and sorting mechanisms for proteins and lipids. The discourse further navigates to its regulatory prominence in neurogenesis and its implications in Alzheimer's Disease pathogenesis. The emerging nexus between the Golgi apparatus and SARS-CoV-2 underscores its potential in viral replication processes. This consolidation accentuates the Golgi apparatus's centrality in neurobiology and its intersections with both neurodegenerative and viral pathologies. In essence, understanding the Golgi's multifaceted functions harbors profound implications for future therapeutic innovations in neurological and viral afflictions.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Lucian Eva
- Faculty of Medicine, “Dunarea de Jos” University of Galati, 800201 Galați, Romania
- Emergency Clinical Hospital “Prof. dr. N. Oblu”, 700309 Iasi, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
138
|
Lou K, Liu S, Zhang F, Sun W, Su X, Bi W, Yin Q, Qiu Y, Zhang Z, Jing M, Ma S. The effect of hyperthyroidism on cognitive function, neuroinflammation, and necroptosis in APP/PS1 mice. J Transl Med 2023; 21:657. [PMID: 37740205 PMCID: PMC10517505 DOI: 10.1186/s12967-023-04511-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Increasing evidence has linked the thyroid dysfunction to the pathogenesis of dementia. Evidence from clinical studies has demonstrated that hypothyroidism is related to an increased risk of dementia. But the association of hyperthyroidism with dementia is largely unknown. METHODS We used the adenovirus containing thyrotropin receptor (TSHR) amino acid residues 1-289 (Ad-TSHR289)-induced Graves' disease (GD) phenotype in Alzheimer's disease (AD) model mice (APP/PS1 mice) to evaluate the effect of hyperthyroidism on the cognitive function and β-amyloid (Aβ) accumulation. RESULTS GD mice exhibited a stable long-term hyperthyroidism and cognitive deficits. Single Cell RNA-sequencing analysis indicated that microglia function played a critical role in the pathophysiological processes in GD mice. Neuroinflammation and polarization of microglia (M1/M2 phenotype) and activated receptor-interacting serine/threonine protein kinase 3 (RIPK3)/mixed lineage kinase domain-like pseudo-kinase (MLKL)-mediated necroptosis contributed to the pathological process, including Aβ deposition and neuronal loss. RIPK3 inhibitor could inhibit GD-mediated Aβ accumulation and neuronal loss. CONCLUSIONS Our findings reveal that GD hyperthyroidism aggravates cognitive deficits in AD mice and induces Aβ deposition and neuronal loss by inducing neuroinflammation and RIPK3/MLKL-mediated necroptosis.
Collapse
Affiliation(s)
- Kai Lou
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Shudong Liu
- Department of Endocrinology, Shandong Rongjun General Hospital, Jinan, 250013, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Wenxiu Sun
- Department of Nursing, Taishan Vocational College of Nursing, Taian, 271000, Shandong, China
| | - Xinhuan Su
- Department of Geriatrics Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaxin Qiu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Zhenyuan Zhang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Mengzhe Jing
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Shizhan Ma
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, 250021, Shandong, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China.
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
| |
Collapse
|
139
|
Cisternas P, Gherardelli C, Gutierrez J, Salazar P, Mendez-Orellana C, Wong GW, Inestrosa NC. Adiponectin and resistin modulate the progression of Alzheimer´s disease in a metabolic syndrome model. Front Endocrinol (Lausanne) 2023; 14:1237796. [PMID: 37732123 PMCID: PMC10507329 DOI: 10.3389/fendo.2023.1237796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
Metabolic syndrome (MetS), a cluster of metabolic conditions that include obesity, hyperlipidemia, and insulin resistance, increases the risk of several aging-related brain diseases, including Alzheimer's disease (AD). However, the underlying mechanism explaining the link between MetS and brain function is poorly understood. Among the possible mediators are several adipose-derived secreted molecules called adipokines, including adiponectin (ApN) and resistin, which have been shown to regulate brain function by modulating several metabolic processes. To investigate the impact of adipokines on MetS, we employed a diet-induced model to induce the various complications associated with MetS. For this purpose, we administered a high-fat diet (HFD) to both WT and APP/PSN1 mice at a pre-symptomatic disease stage. Our data showed that MetS causes a fast decline in cognitive performance and stimulates Aβ42 production in the brain. Interestingly, ApN treatment restored glucose metabolism and improved cognitive functions by 50% while decreasing the Aβ42/40 ratio by approximately 65%. In contrast, resistin exacerbated Aβ pathology, increased oxidative stress, and strongly reduced glucose metabolism. Together, our data demonstrate that ApN and resistin alterations could further contribute to AD pathology.
Collapse
Affiliation(s)
- Pedro Cisternas
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua, Chile
| | - Camila Gherardelli
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Joel Gutierrez
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Mendez-Orellana
- Carrera de Fonoaudiología, Departamento Ciencias de la Salud, facultad Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - G. William Wong
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
140
|
Alves SS, Servilha-Menezes G, Rossi L, da Silva Junior RMP, Garcia-Cairasco N. Evidence of disturbed insulin signaling in animal models of Alzheimer's disease. Neurosci Biobehav Rev 2023; 152:105326. [PMID: 37479008 DOI: 10.1016/j.neubiorev.2023.105326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 06/02/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
Since glucose reuptake by neurons is mostly independent of insulin, it has been an intriguing question whether insulin has or not any roles in the brain. Consequently, the identification of insulin receptors in the central nervous system has fueled investigations of insulin functions in the brain. It is also already known that insulin can influence glucose reuptake by neurons, mostly during activities that have the highest energy demand. The identification of high density of insulin receptors in the hippocampus also suggests that insulin may present important roles related to memory. In this context, studies have reported worse performance in cognitive tests among diabetic patients. In addition, alterations in the regulation of central insulin pathways have been observed in the brains of Alzheimer's disease (AD) patients. In fact, some authors have proposed AD as a third type of diabetes and recently, our group proposed insulin resistance as a common link between different AD hypotheses. Therefore, in the present narrative review, we intend to revise and gather the evidence of disturbed insulin signaling in experimental animal models of AD.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil
| | - Leticia Rossi
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil
| | - Rui Milton Patrício da Silva Junior
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil; Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Brazil; Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil.
| |
Collapse
|
141
|
Pinto-Hernandez P, Castilla-Silgado J, Coto-Vilcapoma A, Fernández-Sanjurjo M, Fernández-García B, Tomás-Zapico C, Iglesias-Gutiérrez E. Modulation of microRNAs through Lifestyle Changes in Alzheimer's Disease. Nutrients 2023; 15:3688. [PMID: 37686720 PMCID: PMC10490103 DOI: 10.3390/nu15173688] [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: 07/28/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Lifestyle factors, including diet and physical activity (PA), are known beneficial strategies to prevent and delay Alzheimer's disease (AD) development. Recently, microRNAs have emerged as potential biomarkers in multiple diseases, including AD. The aim of this review was to analyze the available information on the modulatory effect of lifestyle on microRNA expression in AD. Few studies have addressed this question, leaving important gaps and limitations: (1) in human studies, only circulating microRNAs were analyzed; (2) in mice studies, microRNA expression was only analyzed in brain tissue; (3) a limited number of microRNAs was analyzed; (4) no human nutritional intervention studies were conducted; and (5) PA interventions in humans and mice were poorly detailed and only included aerobic training. Despite this, some conclusions could be drawn. Circulating levels of let-7g-5p, miR-107, and miR-144-3p were associated with overall diet quality in mild cognitive impairment patients. In silico analysis showed that these microRNAs are implicated in synapse formation, microglia activation, amyloid beta accumulation, and pro-inflammatory pathways, the latter also being targeted by miR-129-5p and miR-192-5p, whose circulating levels are modified by PA in AD patients. PA also modifies miR-132, miR-15b-5p, miR-148b-3p, and miR-130a-5p expression in mice brains, which targets are related to the regulation of neuronal activity, ageing, and pro-inflammatory pathways. This supports the need to further explore lifestyle-related miRNA changes in AD, both as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Paola Pinto-Hernandez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Juan Castilla-Silgado
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Almudena Coto-Vilcapoma
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Manuel Fernández-Sanjurjo
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Benjamín Fernández-García
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
- Department of Morphology and Cell Biology, Anatomy, University of Oviedo, 33006 Asturias, Spain
| | - Cristina Tomás-Zapico
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Eduardo Iglesias-Gutiérrez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| |
Collapse
|
142
|
van Heusden FC, van Nifterick AM, Souza BC, França ASC, Nauta IM, Stam CJ, Scheltens P, Smit AB, Gouw AA, van Kesteren RE. Neurophysiological alterations in mice and humans carrying mutations in APP and PSEN1 genes. Alzheimers Res Ther 2023; 15:142. [PMID: 37608393 PMCID: PMC10464047 DOI: 10.1186/s13195-023-01287-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Studies in animal models of Alzheimer's disease (AD) have provided valuable insights into the molecular and cellular processes underlying neuronal network dysfunction. Whether and how AD-related neurophysiological alterations translate between mice and humans remains however uncertain. METHODS We characterized neurophysiological alterations in mice and humans carrying AD mutations in the APP and/or PSEN1 genes, focusing on early pre-symptomatic changes. Longitudinal local field potential recordings were performed in APP/PS1 mice and cross-sectional magnetoencephalography recordings in human APP and/or PSEN1 mutation carriers. All recordings were acquired in the left frontal cortex, parietal cortex, and hippocampus. Spectral power and functional connectivity were analyzed and compared with wildtype control mice and healthy age-matched human subjects. RESULTS APP/PS1 mice showed increased absolute power, especially at higher frequencies (beta and gamma) and predominantly between 3 and 6 moa. Relative power showed an overall shift from lower to higher frequencies over almost the entire recording period and across all three brain regions. Human mutation carriers, on the other hand, did not show changes in power except for an increase in relative theta power in the hippocampus. Mouse parietal cortex and hippocampal power spectra showed a characteristic peak at around 8 Hz which was not significantly altered in transgenic mice. Human power spectra showed a characteristic peak at around 9 Hz, the frequency of which was significantly reduced in mutation carriers. Significant alterations in functional connectivity were detected in theta, alpha, beta, and gamma frequency bands, but the exact frequency range and direction of change differed for APP/PS1 mice and human mutation carriers. CONCLUSIONS Both mice and humans carrying APP and/or PSEN1 mutations show abnormal neurophysiological activity, but several measures do not translate one-to-one between species. Alterations in absolute and relative power in mice should be interpreted with care and may be due to overexpression of amyloid in combination with the absence of tau pathology and cholinergic degeneration. Future studies should explore whether changes in brain activity in other AD mouse models, for instance, those also including tau pathology, provide better translation to the human AD continuum.
Collapse
Affiliation(s)
- Fran C van Heusden
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Anne M van Nifterick
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Bryan C Souza
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, 6525AJ, The Netherlands
| | - Arthur S C França
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, 6525AJ, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, 1105 BA, The Netherlands
| | - Ilse M Nauta
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Cornelis J Stam
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Alida A Gouw
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands.
| |
Collapse
|
143
|
Pérez-Martín E, Coto-Vilcapoma A, Castilla-Silgado J, Rodríguez-Cañón M, Prado C, Álvarez G, Álvarez-Vega MA, Fernández-García B, Menéndez-González M, Tomás-Zapico C. Refining Stereotaxic Neurosurgery Techniques and Welfare Assessment for Long-Term Intracerebroventricular Device Implantation in Rodents. Animals (Basel) 2023; 13:2627. [PMID: 37627418 PMCID: PMC10452028 DOI: 10.3390/ani13162627] [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: 07/25/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Stereotaxic surgeries enable precise access to specific brain regions, being of particular interest for chronic intracerebroventricular drug delivery. However, the challenge of long-term studies at this level is to allow the implantation of drug storage devices and their correct intrathecal connection while guaranteeing animal welfare during the entire study period. In this study, we propose an optimized method for safe intrathecal device implantation, focusing on preoperative, intraoperative, and postoperative procedures, following the 3Rs principle and animal welfare regulations. Our optimized protocol introduces three main refinements. Firstly, we modify the dimensions of the implantable devices, notably diminishing the device-to-mouse weight ratio. Secondly, we use a combination of cyanoacrylate tissue adhesive and UV light-curing resin, which decreases surgery time, improves healing, and notably minimizes cannula detachment or adverse effects. Thirdly, we develop a customized welfare assessment scoresheet to accurately monitor animal well-being during long-term implantations. Taken together, these refinements positively impacted animal welfare by minimizing the negative effects on body weight, surgery-related complications, and anxiety-like behaviors. Overall, the proposed refinements have the potential to reduce animal use, enhance experimental data quality, and improve reproducibility. Additionally, these improvements can be extended to other neurosurgical techniques, thereby advancing neuroscience research, and benefiting the scientific community.
Collapse
Affiliation(s)
- Ester Pérez-Martín
- Neuroscience Innovative Technologies S.L., Neurostech, 33428 Llanera, Spain (C.P.)
| | - Almudena Coto-Vilcapoma
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Juan Castilla-Silgado
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | | | - Catuxa Prado
- Neuroscience Innovative Technologies S.L., Neurostech, 33428 Llanera, Spain (C.P.)
| | - Gabriel Álvarez
- Neuroscience Innovative Technologies S.L., Neurostech, 33428 Llanera, Spain (C.P.)
| | - Marco Antonio Álvarez-Vega
- Departamento de Cirugía, Área de Cirugía, Universidad de Oviedo, 33006 Oviedo, Spain
- Servicio de Neurocirugía, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Benjamín Fernández-García
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Departamento de Morfología y Biología Celular, Área de Anatomía, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Manuel Menéndez-González
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, 33011 Oviedo, Spain
| | - Cristina Tomás-Zapico
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
144
|
Olufunmilayo EO, Holsinger RMD. Roles of Non-Coding RNA in Alzheimer's Disease Pathophysiology. Int J Mol Sci 2023; 24:12498. [PMID: 37569871 PMCID: PMC10420049 DOI: 10.3390/ijms241512498] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is accompanied by deficits in memory and cognitive functions. The disease is pathologically characterised by the accumulation and aggregation of an extracellular peptide referred to as amyloid-β (Aβ) in the form of amyloid plaques and the intracellular aggregation of a hyperphosphorelated protein tau in the form of neurofibrillary tangles (NFTs) that cause neuroinflammation, synaptic dysfunction, and oxidative stress. The search for pathomechanisms leading to disease onset and progression has identified many key players that include genetic, epigenetic, behavioural, and environmental factors, which lend support to the fact that this is a multi-faceted disease where failure in various systems contributes to disease onset and progression. Although the vast majority of individuals present with the sporadic (non-genetic) form of the disease, dysfunctions in numerous protein-coding and non-coding genes have been implicated in mechanisms contributing to the disease. Recent studies have provided strong evidence for the association of non-coding RNAs (ncRNAs) with AD. In this review, we highlight the current findings on changes observed in circular RNA (circRNA), microRNA (miRNA), short interfering RNA (siRNA), piwi-interacting RNA (piRNA), and long non-coding RNA (lncRNA) in AD. Variations in these ncRNAs could potentially serve as biomarkers or therapeutic targets for the diagnosis and treatment of Alzheimer's disease. We also discuss the results of studies that have targeted these ncRNAs in cellular and animal models of AD with a view for translating these findings into therapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Edward O. Olufunmilayo
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
- Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 200212, Nigeria
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
145
|
Gomez‐Gutierrez R, Ghosh U, Yau W, Gamez N, Do K, Kramm C, Shirani H, Vegas‐Gomez L, Schulz J, Moreno‐Gonzalez I, Gutierrez A, Nilsson KPR, Tycko R, Soto C, Morales R. Two structurally defined Aβ polymorphs promote different pathological changes in susceptible mice. EMBO Rep 2023; 24:e57003. [PMID: 37424505 PMCID: PMC10398671 DOI: 10.15252/embr.202357003] [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: 02/14/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/11/2023] Open
Abstract
Misfolded Aβ is involved in the progression of Alzheimer's disease (AD). However, the role of its polymorphic variants or conformational strains in AD pathogenesis is not fully understood. Here, we study the seeding properties of two structurally defined synthetic misfolded Aβ strains (termed 2F and 3F) using in vitro and in vivo assays. We show that 2F and 3F strains differ in their biochemical properties, including resistance to proteolysis, binding to strain-specific dyes, and in vitro seeding. Injection of these strains into a transgenic mouse model produces different pathological features, namely different rates of aggregation, formation of different plaque types, tropism to specific brain regions, differential recruitment of Aβ40 /Aβ42 peptides, and induction of microglial and astroglial responses. Importantly, the aggregates induced by 2F and 3F are structurally different as determined by ssNMR. Our study analyzes the biological properties of purified Aβ polymorphs that have been characterized at the atomic resolution level and provides relevant information on the pathological significance of misfolded Aβ strains.
Collapse
Affiliation(s)
- Ruben Gomez‐Gutierrez
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‐IBIMA, Facultad de CienciasUniversidad de MálagaMálagaSpain
| | - Ujjayini Ghosh
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Wai‐Ming Yau
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Nazaret Gamez
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‐IBIMA, Facultad de CienciasUniversidad de MálagaMálagaSpain
| | - Katherine Do
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Carlos Kramm
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Hamid Shirani
- Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - Laura Vegas‐Gomez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‐IBIMA, Facultad de CienciasUniversidad de MálagaMálagaSpain
| | - Jonathan Schulz
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Ines Moreno‐Gonzalez
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‐IBIMA, Facultad de CienciasUniversidad de MálagaMálagaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA)Universidad Bernardo O'HigginsSantiagoChile
| | - Antonia Gutierrez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‐IBIMA, Facultad de CienciasUniversidad de MálagaMálagaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - K Peter R Nilsson
- Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - Robert Tycko
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Claudio Soto
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Rodrigo Morales
- Department of NeurologyThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA)Universidad Bernardo O'HigginsSantiagoChile
| |
Collapse
|
146
|
Portalés A, Chamero P, Jurado S. Natural and Pathological Aging Distinctively Impacts the Pheromone Detection System and Social Behavior. Mol Neurobiol 2023; 60:4641-4658. [PMID: 37129797 PMCID: PMC10293359 DOI: 10.1007/s12035-023-03362-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Normal aging and many age-related disorders such as Alzheimer's disease cause deficits in olfaction; however, it is currently unknown how natural and pathological aging impacts the detection of social odors which might contribute to the impoverishment of social behavior at old age further worsening overall health. Analysis of the vomeronasal organ, the main gateway to pheromone-encoded information, indicated that natural and pathological aging distinctively affects the neurogenic ability of the vomeronasal sensory epithelium. Whereas cell proliferation remained majorly preserved in 1-year-old APP/PS1 mice, naturally aged animals exhibited significant deficiencies in the number of mature, proliferative, and progenitor cells. These alterations may support age-related deficits in the recognition of social cues and the display of social behavior. Our findings indicate that aging disrupts the processing of social olfactory cues decreasing social odor exploration, discrimination, and habituation in both wild-type senescent (2-year-old) mice and in 1-year-old double mutant model of Alzheimer's disease (APP/PS1). Furthermore, social novelty was diminished in 1-year-old APP/PS1 mice, indicating that alterations in the processing of social cues are accelerated during pathological aging. This study reveals fundamental differences in the cellular processes by which natural and pathological aging disrupts the exploration of social information and social behavior.
Collapse
Affiliation(s)
- Adrián Portalés
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández (CSIC-UMH), 03550, Sant Joan d´Alacant, Spain
| | - Pablo Chamero
- Laboratoire de Physiologie de La Reproduction Et Des Comportements, CNRS, IFCE, INRAE, University of Tours, 37380, Nouzilly, France
| | - Sandra Jurado
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández (CSIC-UMH), 03550, Sant Joan d´Alacant, Spain.
| |
Collapse
|
147
|
Soto PL, Young ME, DiMarco GM, George B, Melnikova T, Savonenko AV, Harris BN. Longitudinal assessment of cognitive function in the APPswe/PS1dE9 mouse model of Alzheimer's-related beta-amyloidosis. Neurobiol Aging 2023; 128:85-99. [PMID: 37120419 PMCID: PMC10239324 DOI: 10.1016/j.neurobiolaging.2023.03.010] [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: 01/13/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 05/01/2023]
Abstract
Preclinical models of Alzheimer's disease (AD)-related cognitive decline can be useful for developing therapeutics. The current study longitudinally assessed short-term memory, using a delayed matching-to-position (DMTP) task, and attention, using a 3-choice serial reaction time (3CSRT) task, from approximately 18 weeks of age through death or 72 weeks of age in APPswe/PS1dE9 mice, a widely used mouse model of AD-related amyloidosis. Both transgenic (Tg) and non-Tg mice exhibited improvements in DMTP accuracy over time. Breaks in testing reduced DMTP accuracy but accuracy values quickly recovered in both Tg and non-Tg mice. Both Tg and non-Tg mice exhibited high accuracy in the 3CSRT task with breaks in testing briefly reducing accuracy values equivalently in the 2 genotypes. The current results raise the possibility that deficits in Tg APPswe/PS1dE9 mice involve impairments in learning rather than declines in established performances. A better understanding of the factors that determine whether deficits develop will be useful for designing evaluations of potential pharmacotherapeutics and may reveal interventions for clinical application.
Collapse
Affiliation(s)
- Paul L Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA; Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | - Michael E Young
- Department of Psychology, Kansas State University, Manhattan, KS, USA
| | - Giuliana M DiMarco
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ, USA
| | - Brianna George
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alena V Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Breanna N Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
148
|
Wang C, Liu H, Xu S, Deng Y, Xu B, Yang T, Liu W. Ferroptosis and Neurodegenerative Diseases: Insights into the Regulatory Roles of SLC7A11. Cell Mol Neurobiol 2023; 43:2627-2642. [PMID: 36988772 PMCID: PMC11410137 DOI: 10.1007/s10571-023-01343-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Programed cell death plays a key role in promoting human development and maintaining homeostasis. Ferroptosis is a recently identified pattern of programmed cell death that is closely associated with the onset and progression of neurodegenerative diseases. Ferroptosis is mainly caused by the intracellular accumulation of iron-dependent lipid peroxides. The cysteine/glutamate antibody Solute carrier family 7 member 11 (SLC7A11, also known as xCT) functions to import cysteine for glutathione biosynthesis and antioxidant defense. SLC7A11 has a significant impact on ferroptosis, and inhibition of SLC7A11 expression promotes ferroptosis. Moreover, SLC7A11 is also closely associated with neurodegenerative diseases. In this paper, we summarize the relationship between ferroptosis and neurodegenerative diseases and the role of SLC7A11 during this process. The various regulatory mechanisms of SLC7A11 are also discussed. In conclusion, we are looking forward to a theoretical basis for further understanding the occurrence and development of ferroptosis in SLC7A11 and neurodegenerative diseases, and to seek new clues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Chen Wang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Haihui Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Si Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
149
|
Rodríguez-Ruiz ER, Herrero-Labrador R, Fernández-Fernández AP, Serrano-Masa J, Martínez-Montero JA, González-Nieto D, Hana-Vaish M, Benchekroun M, Ismaili L, Marco-Contelles J, Martínez-Murillo R. The Proof-of-Concept of MBA121, a Tacrine-Ferulic Acid Hybrid, for Alzheimer's Disease Therapy. Int J Mol Sci 2023; 24:12254. [PMID: 37569630 PMCID: PMC10419016 DOI: 10.3390/ijms241512254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/26/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Great effort has been devoted to the synthesis of novel multi-target directed tacrine derivatives in the search of new treatments for Alzheimer's disease (AD). Herein we describe the proof of concept of MBA121, a compound designed as a tacrine-ferulic acid hybrid, and its potential use in the therapy of AD. MBA121 shows good β-amyloid (Aβ) anti-aggregation properties, selective inhibition of human butyrylcholinesterase, good neuroprotection against toxic insults, such as Aβ1-40, Aβ1-42, and H2O2, and promising ADMET properties that support translational developments. A passive avoidance task in mice with experimentally induced amnesia was carried out, MBA121 being able to significantly decrease scopolamine-induced learning deficits. In addition, MBA121 reduced the Aβ plaque burden in the cerebral cortex and hippocampus in APPswe/PS1ΔE9 transgenic male mice. Our in vivo results relate its bioavailability with the therapeutic response, demonstrating that MBA121 is a promising agent to treat the cognitive decline and neurodegeneration underlying AD.
Collapse
Affiliation(s)
- Emelina R. Rodríguez-Ruiz
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| | - Raquel Herrero-Labrador
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| | - Ana P. Fernández-Fernández
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| | - Julia Serrano-Masa
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| | - José A. Martínez-Montero
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| | - Daniel González-Nieto
- Experimental Neurology Unit, Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain;
| | - Mayuri Hana-Vaish
- UT Southwestern Medical Center, Department of Neurosurgery, School of Medicine, Baylor College of Medicine, Rice University, Houston, TX 77005, USA;
| | - Mohamed Benchekroun
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive de Besançon, Groupe Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France;
| | - Lhassane Ismaili
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive de Besançon, Groupe Chimie Médicinale, Université de Franche-Comté, F-25000 Besançon, France;
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of Organic Chemistry (CSIC), C/Juan de la Cierva, 3, 28006 Madrid, Spain;
- Center for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, 28029 Madrid, Spain
| | - Ricardo Martínez-Murillo
- Neurovascular Research Group, Instituto Cajal (CSIC), Ave. Doctor Arce 37, 28002 Madrid, Spain; (E.R.R.-R.); (R.H.-L.); (A.P.F.-F.); (J.S.-M.); (J.A.M.-M.)
| |
Collapse
|
150
|
Cazzaro S, Woo JAA, Wang X, Liu T, Rego S, Kee TR, Koh Y, Vázquez-Rosa E, Pieper AA, Kang DE. Slingshot homolog-1-mediated Nrf2 sequestration tips the balance from neuroprotection to neurodegeneration in Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2217128120. [PMID: 37463212 PMCID: PMC10374160 DOI: 10.1073/pnas.2217128120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Oxidative damage in the brain is one of the earliest drivers of pathology in Alzheimer's disease (AD) and related dementias, both preceding and exacerbating clinical symptoms. In response to oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) is normally activated to protect the brain from oxidative damage. However, Nrf2-mediated defense against oxidative stress declines in AD, rendering the brain increasingly vulnerable to oxidative damage. Although this phenomenon has long been recognized, its mechanistic basis has been a mystery. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that Slingshot homolog-1 (SSH1) drives this effect by acting as a counterweight to neuroprotective Nrf2 in response to oxidative stress and disease. Specifically, oxidative stress-activated SSH1 suppresses nuclear Nrf2 signaling by sequestering Nrf2 complexes on actin filaments and augmenting Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 interaction, independently of SSH1 phosphatase activity. We also show that Ssh1 elimination in AD models increases Nrf2 activation, which mitigates tau and amyloid-β accumulation and protects against oxidative injury, neuroinflammation, and neurodegeneration. Furthermore, loss of Ssh1 preserves normal synaptic function and transcriptomic patterns in tauP301S mice. Importantly, we also show that human AD brains exhibit highly elevated interactions of Nrf2 with both SSH1 and Keap1. Thus, we demonstrate here a unique mode of Nrf2 blockade that occurs through SSH1, which drives oxidative damage and ensuing pathogenesis in AD. Strategies to inhibit SSH1-mediated Nrf2 suppression while preserving normal SSH1 catalytic function may provide new neuroprotective therapies for AD and related dementias.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Jung-A A. Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Tian Liu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Shanon Rego
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Teresa R. Kee
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Yeojung Koh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
- Brain Health Medicines, Center Harrington Discovery Institute, Cleveland, OH44106
| | - David E. Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
| |
Collapse
|