1
|
Liu Y, Aquili L, Wong KH, Lu Z, Lim LW. Past, present, and future of serotonin-targeting therapeutics for Alzheimer's disease: Perspectives from DNA methylation. Ageing Res Rev 2025; 108:102755. [PMID: 40239871 DOI: 10.1016/j.arr.2025.102755] [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: 06/18/2024] [Revised: 03/02/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
With population aging, Alzheimer's disease (AD) is becoming increasingly prevalent, causing great health and economic burdens worldwide. Despite decades of research, there are still no effective disease-modifying treatments for AD, highlighting the urgent need for more in-depth understanding of the disease-causing mechanisms. The brain serotonin (5-HT) neurotransmission system undergoes structural and functional changes in aging and AD, which contributes to cognitive decline and comorbid mood disturbances. This review discusses the critical involvement of the brain 5-HT system in aging and AD. Existing findings on the changes in projection fiber innervation and receptor/transporter expression in AD are reviewed. Preclinical and clinical progress on the development of 5-HT-modulating drugs for AD and the obstacles faced by these development efforts are discussed. Epigenetic control of the brain 5-HT system and the potential of modulating 5-HT transmission via DNA methylation are also examined.
Collapse
Affiliation(s)
- Yanzhi Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Luca Aquili
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China; School of Management, Ritsumeikan Asia Pacific University, Beppu, Oita, Japan.
| | - Kah Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia.
| | - Zhiliang Lu
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, and Suzhou Municipal Key Laboratory of Cancer Biology and Chronic Disease, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| |
Collapse
|
2
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
3
|
Xiong C, Yu Z, Yin Y, Zhu Q, Ba R, Shen Y, Li H, Wei Z, Lin Z, Wu D. Longitudinal changes of blood-brain barrier and transcytolemmal water exchange permeability in Alzheimer's disease mice: A non-contrast MRI study. Neuroimage 2025; 310:121141. [PMID: 40089221 DOI: 10.1016/j.neuroimage.2025.121141] [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: 11/25/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Growing evidence suggests that Alzheimer's disease (AD) has been linked with the dysfunction of glymphatic system. Previous studies were primarily cross-sectional and focused on only one specific component, hindering the understanding of overall glymphatic function in AD. We evaluated the longitudinal changes in multiple components of glymphatic system (blood-brain barrier (BBB) and transcytolemmal water exchange (TWE) permeability) in AD mice. Five female wild-type and four 3 × Tg-AD mice from 5 to 13 months of age were scanned monthly using two non-contrast MRI techniques, water-extraction-with-phase-contrast-arterial-spin-tagging (WEPCAST) and diffusion-time-dependent kurtosis imaging (tDKI), yielding BBB and TWE permeability. Immunostaining was used to evaluate tight junction proteins associated with BBB structural integrity, aquaporin 4 (AQP4) related to TWE, and AQP4 perivascular space (PVS) polarization that might represent PVS-parenchyma water exchange. The relationship between glymphatic function and AD pathology, as measured by amyloid beta (Aβ) and tau deposition, was also explored. Our results revealed significantly increased BBB and hippocampal TWE permeability in AD mouse brains, consistent with the histological findings of reduced tight junction proteins and upregulated AQP4, which were correlated with each other and can be predictive of Aβ and tau deposition. Impaired AQP4 PVS polarization was also found in AD mice. In conclusion, water exchange in multiple components of glymphatic system altered in AD mice, and these in vivo MRI findings were validated pathologically, which might affect the waste clearance in the glymphatic neurofluid.
Collapse
Affiliation(s)
- Chuhan Xiong
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Ziyang Yu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Yu Yin
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Qinfeng Zhu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Ruicheng Ba
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Yao Shen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Haotian Li
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Zhiliang Wei
- Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zixuan Lin
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China.
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China.
| |
Collapse
|
4
|
Matias C, Yamada C, Movila A, Brault JJ. Optimizing Confocal Imaging Protocols for Muscle Fiber Typing in the Mouse Masseter Muscle. Bio Protoc 2025; 15:e5267. [PMID: 40224663 PMCID: PMC11986697 DOI: 10.21769/bioprotoc.5267] [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: 12/17/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
The masseter muscle, a key orofacial muscle, demonstrates unique anatomical and functional properties, including sexual dimorphism in myosin heavy chain (MyHC) expression and complex fiber architecture. Despite its importance in mastication and relevance to various disorders, phenotypic characterization of the masseter remains limited. Conventional fluorescence microscopy has been a cornerstone in muscle fiber typing, reliably identifying MyHC isoforms and measuring fiber cross-sectional areas. Building on this foundation, confocal microscopy offers complementary advantages, such as enhanced resolution, increased flexibility for multiplexing, and the ability to visualize complex structures in three dimensions. This study presents a detailed protocol for using confocal microscopy to achieve high-resolution imaging and molecular characterization of masseter muscle cryosections. By leveraging advanced technologies such as white light lasers and extended z-length imaging, this method ensures precise spectral separation, simultaneous multichannel fluorescence detection, and the ability to capture muscle architecture in three dimensions. The protocol includes tissue preparation, immunostaining for MyHC isoforms, and postprocessing for fiber segmentation and quantification. The imaging setup was optimized for minimizing signal bleed through, improving the signal-to-noise ratio, and enabling detailed visualization of muscle fibers and molecular markers. Image postprocessing allows for quantification of the cross-sectional area of individual fibers, nuclei location measurements, and identification of MyHC isoforms within each fiber. This confocal microscopy-based protocol provides similar resolution and contrast compared to conventional techniques, enabling robust multiplexed imaging and 3D reconstruction of muscle structures. These advantages make it a valuable tool for studying complex muscle architecture, offering broad applications in muscle physiology and pathology research. Key features • Enables high-resolution imaging of muscle fiber architecture, capturing detailed spatial relationships using extended z-length and advanced spectral separation techniques. • Supports simultaneous detection of multiple molecular markers for robust muscle fiber typing and molecular localization. • Allows for the generation of three-dimensional models to analyze muscle structures such as neuromuscular junctions, extracellular matrix, and mitochondrial organization. • Adaptable to various skeletal muscles and species, providing valuable insights into muscle physiology, regeneration, and disease processes. Graphical overview Analyzing muscle fiber composition and morphology in mice's masseter muscle using confocal microscopy. Workflow for characterizing rodent masseter muscle fibers using advanced confocal microscopy. Confocal microscopy, equipped with white light laser technology and optimized z-stack imaging, allows precise spectral unmixing to reduce bleed through and enhance signal detection. The z-length is extended beyond the physical thickness of the sample to account for potential variations in tissue flatness and ensure complete imaging of all focal planes. The resulting high-resolution images provide detailed insights into fiber architecture, molecular composition, and cross-sectional areas, ensuring robust and reproducible data for analyzing the complex phenotypic characteristics of the masseter and other muscles.
Collapse
Affiliation(s)
- Catalina Matias
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chiaki Yamada
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Alexandru Movila
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Jeffrey J. Brault
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
5
|
Liang C, Okamoto AA, Karim F, Kawauchi S, Melkonyan L, Danh TB, Mukherjee J. Disruption of normal brain distribution of [ 18F]Nifene to α4β2* nicotinic acetylcholinergic receptors in old B6129SF2/J mice and transgenic 3xTg-AD mice model of Alzheimer's disease: In Vivo PET/CT imaging studies. Neuroimage 2025; 309:121092. [PMID: 39978704 DOI: 10.1016/j.neuroimage.2025.121092] [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: 09/03/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
The 3xTg-AD transgenic mouse model develops Aβ plaque and tau pathology and is purported to closely resemble pathological development in the human Alzheimer's disease (AD) brain. Nicotinic acetylcholine receptors (nAChRs) α4β2* subtype, was studied in this mouse model using [18F]nifene PET/CT and compared with non-transgenic B6129SF2/J mice (male and female). Young 2-month old B6129SF2/J exhibited normal [18F]nifene distribution (measured as standard uptake volume ratios, SUVR with cerebellum as reference) thalamus (TH) 3.12> medial prefrontal cortex (mPFC) 2.33> frontal cortex (FC) 2.06> hippocampus-subiculum (HP-SUB) 1.6. At 11-months of age, B6129SF2/J exhibited high, irreversible and non-saturable [18F]nifene binding in mPFC higher than in TH (mPFC 3.8> TH 2.82> FC 1.79> HP-SUB 1.73). The 3xTg-AD also exhibited high mPFC binding, although the region of highest binding within the mPFC was different compared to B6129SF2/J mice (mPFC 2.44> TH 2.27> FC 1.61> HP-SUB 1.48). [125I]IBETA and immunohistochemistry in 3xTg-AD brain slices confirmed Aβ plaques. The TH of 3xTg-AD mice had lower [18F]nifene binding (reduced by approximately 20 %) compared to both, young and old B6129SF2/J, and was significant. The mPFC [18F]nifene binding was significantly higher in the old B6129SF2/J compared to both the young B6129SF2/J and the 3xTg-AD mice (>150 %). Overall, 3xTg-AD transgenic mice had reduced [18F]nifene binding compared to B6129SF2/J controls, suggesting possible effects of Aβ plaques and Tau on α4β2* nAChRs.
Collapse
Affiliation(s)
- Christopher Liang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States
| | - Atsumi A Okamoto
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States
| | - Fariha Karim
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States
| | - Shimako Kawauchi
- Transgenic Mouse Facility, University Laboratory Animal Resources, Office of Research, University of California-Irvine, Irvine, CA, United States
| | - Lusine Melkonyan
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States
| | - Tram B Danh
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, United States.
| |
Collapse
|
6
|
Zhao X, Zhou Q, Zhang H, Ono M, Furuyama T, Yamamoto R, Ishikura T, Kumai M, Nakamura Y, Shiga H, Miwa T, Kato N. Olfactory deprivation promotes amyloid β deposition in a mouse model of Alzheimer's disease. Brain Res 2025; 1851:149500. [PMID: 39922408 DOI: 10.1016/j.brainres.2025.149500] [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: 08/22/2024] [Revised: 01/12/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Olfactory dysfunction is regarded as an early marker for Alzheimer's disease (AD). Slowly progressing AD pathology is interpreted to impair cognition and olfactory sensation independently, while olfactory deficits emerge earlier. The present experiments tested the possibility that olfactory impairment may worsen cognition or AD pathology using 3xTg AD model mice with olfactory bulbectomy (OBX). In open-field tests, OBX was shown to increase anxiety-like behavior in both wild-type (WT) and AD model mice, and hyperactivity was induced in WT mice only. Spatial memory, assessed by the Morris water maze (MWM) test, was impaired in WT but not AD mice. Object memory, assessed by the novel object recognition test, was not changed by OBX either in WT or AD mice. Densitometry of Aβ plaques stained with 6E10 and anti-Aβ42 antibodies was carried out in sections containing the hippocampal formation obtained from AD mice aged 12 and 18 months. The plaque area was larger in the OBX than in the sham group at 12 months. At 18 months, there was also difference in the plaque area. Given that Aβ plaques emerge in 3xTg mice relatively later (>9 months of age) than in other models, OBX in 3xTg mice appears to exacerbate Aβ pathology at the early phase of Aβ emergence, implying a causative link of smell loss to AD pathogenesis. The accelerated Aβ plaque formation by OBX was accompanied by microglial activation. Early intervention to smell loss may be beneficial for AD control.
Collapse
Affiliation(s)
- Xirun Zhao
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Qing Zhou
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Huan Zhang
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Munenori Ono
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Takafumi Furuyama
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Tomoko Ishikura
- Department of Otorhinolaryngology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Masami Kumai
- Department of Otorhinolaryngology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Yukari Nakamura
- Department of Otorhinolaryngology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Hideaki Shiga
- Department of Otorhinolaryngology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Takaki Miwa
- Department of Otorhinolaryngology, Kanazawa Medical University, Uchinada 920-0293 Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Uchinada 920-0293 Japan.
| |
Collapse
|
7
|
Soelter TM, Howton TC, Wilk EJ, Whitlock JH, Clark AD, Birnbaum A, Patterson DC, Cortes CJ, Lasseigne BN. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points. J Cell Commun Signal 2025; 19:e70006. [PMID: 40026671 PMCID: PMC11870853 DOI: 10.1002/ccs3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss and cognitive decline, affecting behavior, speech, and motor abilities. The neuropathology of AD includes the formation of extracellular amyloid-β plaques and intracellular neurofibrillary tangles of phosphorylated tau, along with neuronal loss. Although neuronal loss is an AD hallmark, cell-cell communication between neuronal and non-neuronal cell populations maintains neuronal health and brain homeostasis. To study changes in cell-cell communication during disease progression, we performed snRNA-sequencing of the hippocampus from female 3xTg-AD and wild-type littermates at 6 and 12 months. We inferred differential cell-cell communication between 3xTg-AD and wild-type mice across time points and between senders (astrocytes, microglia, oligodendrocytes, and OPCs) and receivers (excitatory and inhibitory neurons) of interest. We also assessed the downstream effects of altered glia-neuron communication using pseudobulk differential gene expression, functional enrichment, and gene regulatory analyses. We found that glia-neuron communication is increasingly dysregulated in 12-month 3xTg-AD mice. We also identified 23 AD-associated ligand-receptor pairs that are upregulated in the 12-month-old 3xTg-AD hippocampus. Our results suggest increased AD association of interactions originating from microglia. Signaling mediators were not significantly differentially expressed but showed altered gene regulation and transcription factor activity. Our findings indicate that altered glia-neuron communication is increasingly dysregulated and affects the gene regulatory mechanisms in neurons of 12-month-old 3xTg-AD mice.
Collapse
Affiliation(s)
- Tabea M. Soelter
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jordan H. Whitlock
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Allison Birnbaum
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Molecular, Cell and Developmental BiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Dalton C. Patterson
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Constanza J. Cortes
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
8
|
Jimenez-Harrison DM, Butler MJ, Ijaz H, Alsabbagh R, Bettes MN, DeMarsh JW, Mackey-Alfonso SE, Muscat SM, Alvarez BD, Blackwell JA, Taylor A, Jantsch J, Sanchez AA, Peters SB, Barrientos RM. Ligature-induced periodontitis in a transgenic mouse model of Alzheimer's disease dysregulates neuroinflammation, exacerbates cognitive impairment, and accelerates amyloid pathology. Brain Behav Immun Health 2025; 44:100969. [PMID: 40094122 PMCID: PMC11909722 DOI: 10.1016/j.bbih.2025.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/19/2025] Open
Abstract
A growing body of literature has identified periodontal disease among the modifiable risk factors for Alzheimer's disease (AD), but the mechanisms underlying this relationship is unknown. This study investigated this relationship using a ligature-induced preclinical periodontitis (Pd) model in non-transgenic (non-Tg) and 3xTg-AD mice. We found that ligature placement caused significant alveolar bone loss, with 3xTg-AD mice exhibiting exacerbated bone loss, suggesting AD-related genetic risk may amplify disease progression. Pd induced robust local inflammatory gene expression in both genotypes, but 3xTg-AD mice indicated a dysregulated immune response. Cognitive deficits were observed only in Pd-afflicted 3xTg-AD mice, specifically in hippocampus-mediated spatial memory and perirhinal cortex-mediated object recognition memory, while non-Tg mice remained unaffected. Neuroinflammatory responses varied by brain region, with the hippocampus and prefrontal cortex (PFC) showing the most pronounced changes. In these regions, 3xTg-AD mice exhibited significantly altered cytokine gene expression compared to non-Tg mice, particularly at later time points. Synaptic markers revealed vulnerabilities in 3xTg-AD mice, including reduced baseline Syp expression and dysregulated Synpo post-ligature. Pd transiently reduced glutamate receptor gene expression in both genotypes, with non-Tg mice showing persistent changes, potentially linked to preserved memory. Pd also accelerated amyloid-β (Aβ) deposition and sustained neurodegeneration in 3xTg-AD mice. Overall, this study shows that combining Pd and AD-related genetic risk exacerbates inflammation, cognitive impairment, synaptic dysfunction, Aβ pathology, and neurodegeneration. Neither insult alone was sufficient to produce these effects, highlighting the synergistic impact. These findings emphasize the need to explore anti-inflammatory interventions and downstream mechanisms to mitigate the confluence of these diseases.
Collapse
Affiliation(s)
- Daniela M. Jimenez-Harrison
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, The Ohio State University College of Medicine, USA
- Neuroscience Graduate Program, The Ohio State University, USA
| | - Michael J. Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Haanya Ijaz
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Rami Alsabbagh
- Division of Periodontology, The Ohio State University College of Dentistry, USA
- Division of Biosciences, The Ohio State University College of Dentistry, USA
| | - Menaz N. Bettes
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - James W. DeMarsh
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Sabrina E. Mackey-Alfonso
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- Medical Scientist Training Program, The Ohio State University College of Medicine, USA
- Neuroscience Graduate Program, The Ohio State University, USA
| | - Stephanie M. Muscat
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Bryan D. Alvarez
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- Neuroscience Graduate Program, The Ohio State University, USA
| | - Jade A. Blackwell
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- MCDB Graduate Program, The Ohio State University, USA
| | - Ashton Taylor
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Jeferson Jantsch
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Andrew A. Sanchez
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Sarah B. Peters
- Division of Biosciences, The Ohio State University College of Dentistry, USA
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ruth M. Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH, USA
- Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
9
|
Varada S, Chamberlin SR, Bui L, Brandes MS, Gladen-Kolarsky N, Harris CJ, Hack W, Neff CJ, Brumbach BH, Soumyanath A, Quinn JF, Gray NE. Oral Asiatic Acid Improves Cognitive Function and Modulates Antioxidant and Mitochondrial Pathways in Female 5xFAD Mice. Nutrients 2025; 17:729. [PMID: 40005058 PMCID: PMC11858387 DOI: 10.3390/nu17040729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Extracts of the plant Centella asiatica can enhance mitochondrial function, promote antioxidant activity and improve cognitive deficits. Asiatic acid (AA) is one of the constituent triterpene compounds present in the plant. In this study, we explore the effects of AA on brain mitochondrial function, antioxidant response and cognition in a beta-amyloid (Aβ)-overexpressing 5xFAD mouse line. Methods: Six- to seven-month-old 5xFAD mice were treated with 1% AA for 4 weeks. In the last week of treatment, associative memory was assessed along with mitochondrial bioenergetics and the expression of mitochondrial and antioxidant response genes from isolated cortical synaptosomes. The Aβ plaque burden was also evaluated. Results: AA treatment resulted in improvements in associative memory in female 5xFAD mice without altering the Aβ plaque burden. Cortical mitochondrial function and mitochondrial gene expression were increased in the AA-treated female 5xFAD mice, as was the expression of antioxidant genes. More modest effects of AA on cortical mitochondrial function and mitochondrial and antioxidant gene expression were observed in male 5xFAD mice. Conclusions: Oral AA treatment improved cognitive and mitochondrial function and activated antioxidant in Aβ-overexpressing mice. These changes occurred independent of alterations in Aβ plaque burden, suggesting that AA could have translational therapeutic relevance in later-stage AD when plaques are well established.
Collapse
Affiliation(s)
- Samantha Varada
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Stephen R. Chamberlin
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Lillie Bui
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Mikah S. Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Noah Gladen-Kolarsky
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Christopher J. Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Wyatt Hack
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Cody J. Neff
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Barbara H. Brumbach
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
- Department of Neurology and Parkinson’s Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR 97239, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| |
Collapse
|
10
|
Martínez-Orozco H, Bencomo-Martínez A, Maya-Arteaga JP, Rubio-De Anda PF, Sanabria-Romero F, Casas ZGM, Rodríguez-Vargas I, Hernández-Puga AG, Sablón-Carrazana M, Menéndez-Soto del Valle R, Rodríguez-Tanty C, Díaz-Cintra S. CNEURO-201, an Anti-amyloidogenic Agent and σ1-Receptor Agonist, Improves Cognition in the 3xTg Mouse Model of Alzheimer's Disease by Multiple Actions in the Pathology. Int J Mol Sci 2025; 26:1301. [PMID: 39941068 PMCID: PMC11818425 DOI: 10.3390/ijms26031301] [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: 12/16/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The complexity of Alzheimer's disease (AD) pathophysiology represents a significant challenge in the development of effective therapeutic agents for its treatment. CNEURO-201 (CN, also Amylovis-201) is a novel pharmaceutical agent with dual activity as an anti-amyloid-β (Aβ) agent and σ1 receptor agonist. CN exhibits great efficacy at very low doses, delaying cognitive impairment and alleviating Aβ load in animal models of AD. However, CN functions on other remains related to this pathology remain to be investigated. The present study sought to evaluate the effects of CN treatment at a dosage of 0.1 mg kg-1 (p.o) over an eight-week period in the 3xTg-AD mouse model. In silico studies, as well as biochemical and immunofluorescence assays, were conducted on brain tissue to investigate the CN effects on acetylcholine metabolism, redox system, and glial cell activation-related biomarkers in brain regions that are relevant for memory. The results demonstrated that CN effectively rescues cognitive impairment of 3xTg-AD mice by influencing glial activity to reduce existing Aβ plaques but also modulating acetylcholine metabolism and the enzymatic response of proteins involved in the redox system. Our outcomes reinforced the potential of CN in treating AD by acting on multiple pathways altered in this disease.
Collapse
Affiliation(s)
- Humberto Martínez-Orozco
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Alberto Bencomo-Martínez
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Juan Pablo Maya-Arteaga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Pedro Francisco Rubio-De Anda
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Fausto Sanabria-Romero
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Zyanya Gloria Mena Casas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Isaac Rodríguez-Vargas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Ana Gabriela Hernández-Puga
- Centro de Investigación Biomédica Avanzada, Facultad de Medicina, Universidad Autónoma de Querétaro, Carretera a Chichimequillas S/N, Santiago de Querétaro 76140, Querétaro, Mexico;
| | - Marquiza Sablón-Carrazana
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Roberto Menéndez-Soto del Valle
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Chryslaine Rodríguez-Tanty
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| |
Collapse
|
11
|
Chevalier C, Tournier BB, Marizzoni M, Park R, Paquis A, Ceyzériat K, Badina AM, Lathuiliere A, Saleri S, Cillis FD, Cattaneo A, Millet P, Frisoni GB. Fecal Microbiota Transplantation (FMT) From a Human at Low Risk for Alzheimer's Disease Improves Short-Term Recognition Memory and Increases Neuroinflammation in a 3xTg AD Mouse Model. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70012. [PMID: 39801363 PMCID: PMC11725982 DOI: 10.1111/gbb.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/15/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025]
Abstract
Human microbiota-associated murine models, using fecal microbiota transplantation (FMT) from human donors, help explore the microbiome's role in diseases like Alzheimer's disease (AD). This study examines how gut bacteria from donors with protective factors against AD influence behavior and brain pathology in an AD mouse model. Female 3xTgAD mice received weekly FMT for 2 months from (i) an 80-year-old AD patient (AD-FMT), (ii) a cognitively healthy 73-year-old with the protective APOEe2 allele (APOEe2-FMT), (iii) a 22-year-old healthy donor (Young-FMT), and (iv) untreated mice (Mice-FMT). Behavioral assessments included novel object recognition (NOR), Y-maze, open-field, and elevated plus maze tests; brain pathology (amyloid and tau), neuroinflammation (in situ autoradiography of the 18 kDa translocator protein in the hippocampus); and gut microbiota were analyzed. APOEe2-FMT improved short-term memory in the NOR test compared to AD-FMT, without significant changes in other behavioral tests. This was associated with increased neuroinflammation in the hippocampus, but no effect was detected on brain amyloidosis and tauopathy. Specific genera, such as Parabacteroides and Prevotellaceae_UGC001, were enriched in the APOEe2-FMT group and associated with neuroinflammation, while genera like Desulfovibrio were reduced and linked to decreased neuroinflammation. Gut microbiota from a donor with a protective factor against AD improved short-term memory and induced neuroinflammation in regions strategic to AD. The association of several genera with neuroinflammation in the APOEe2-FMT group suggests a collegial effect of the transplanted microbiome rather than a single-microbe driver effect. These data support an association between gut bacteria, glial cell activation, and cognitive function in AD.
Collapse
Affiliation(s)
- Claire Chevalier
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | | | - Moira Marizzoni
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Rahel Park
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Arthur Paquis
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| | - Kelly Ceyzériat
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | | | | | - Samantha Saleri
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Floriana De Cillis
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Annamaria Cattaneo
- Biological Psychiatry UnitIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Philippe Millet
- Département de PsychiatrieUniversity of GenevaGenevaSwitzerland
| | - Giovanni B. Frisoni
- Département de Readaptation et gériatrieUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
12
|
Tran KM, Kwang NE, Butler CA, Gomez-Arboledas A, Kawauchi S, Mar C, Chao D, Barahona RA, Da Cunha C, Tsourmas KI, Shi Z, Wang S, Collins S, Walker A, Shi KX, Alcantara JA, Neumann J, Duong DM, Seyfried NT, Tenner AJ, LaFerla FM, Hohsfield LA, Swarup V, MacGregor GR, Green KN. APOE Christchurch enhances a disease-associated microglial response to plaque but suppresses response to tau pathology. Mol Neurodegener 2025; 20:9. [PMID: 39844286 PMCID: PMC11752804 DOI: 10.1186/s13024-024-00793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Apolipoprotein E ε4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). A recent case report identified a rare variant in APOE, APOE3-R136S (Christchurch), proposed to confer resistance to autosomal dominant Alzheimer's Disease (AD). However, it remains unclear whether and how this variant exerts its protective effects. METHODS We introduced the R136S variant into mouse Apoe (ApoeCh) and investigated its effect on the development of AD-related pathology using the 5xFAD model of amyloidosis and the PS19 model of tauopathy. We used immunohistochemical and biochemical analysis along with single-cell spatial omics and bulk proteomics to explore the impact of the ApoeCh variant on AD pathological development and the brain's response to plaques and tau. RESULTS In 5xFAD mice, ApoeCh enhances a Disease-Associated Microglia (DAM) phenotype in microglia surrounding plaques, and reduces plaque load, dystrophic neurites, and plasma neurofilament light chain. By contrast, in PS19 mice, ApoeCh suppresses the microglial and astrocytic responses to tau-laden neurons and does not reduce tau accumulation or phosphorylation, but partially rescues tau-induced synaptic and myelin loss. We compared how microglia responses differ between the two mouse models to elucidate the distinct DAM signatures induced by ApoeCh. We identified upregulation of antigen presentation-related genes in the DAM response in a PS19 compared to a 5xFAD background, suggesting a differential response to amyloid versus tau pathology that is modulated by the presence of ApoeCh. Bulk proteomics show upregulated mitochondrial protein abundance with ApoeCh in 5xFAD mice, but reductions in mitochondrial and translation associated proteins in PS19 mice. CONCLUSIONS These findings highlight the ability of the ApoeCh variant to modulate microglial responses based on the type of pathology, enhancing DAM reactivity in amyloid models and dampening neuroinflammation to promote protection in tau models. This suggests that the Christchurch variant's protective effects likely involve multiple mechanisms, including changes in receptor binding and microglial programming.
Collapse
Affiliation(s)
- Kristine M Tran
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Claire A Butler
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Angela Gomez-Arboledas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Cassandra Mar
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Donna Chao
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Rocio A Barahona
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Kai-Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Joshua A Alcantara
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | | | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Andrea J Tenner
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology & Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, 92697, USA
| | - Frank M LaFerla
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA.
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA.
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
13
|
Udo MSB, Zaccarelli-Magalhães J, Clemons GA, Citadin CT, Langman J, Smith DJ, Matuguma LH, Tesic V, Lin HW. Blockade of A 2AR improved brain perfusion and cognitive function in a mouse model of Alzheimer's disease. GeroScience 2025:10.1007/s11357-025-01526-8. [PMID: 39843732 DOI: 10.1007/s11357-025-01526-8] [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: 11/20/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects more than 6.2 million Americans aged 65 and older, particularly women. Along with AD's main hallmarks (formation of β-amyloid plaques and tau neurofibrillary tangles), there are vascular alterations that occurs in AD pathology. Adenosine A2 receptor (A2AR) is one of the key factors of brain vascular autoregulation and is overexpressed in AD patients. Our previous findings suggest that protein arginine methyltransferase 4 (PRMT4) is overexpressed in AD, which leads to decrease in cerebral blood flow in aged female 3xTg mice. We aimed to investigate the mechanism behind A2AR signaling in the regulation of brain perfusion and blood-brain barrier integrity in age and sex-dependent 3xTg mice, and if it is related to PRMT4. Istradefylline, a highly selective A2AR antagonist, was used to modulate A2AR signaling. Aged female 3xTg and C57BL/6 J mice were evaluated for brain perfusion (via laser speckle) and cognitive function (via open field, T-maze and novel object recognition). Our results suggest that modulation of A2AR signaling in aged female 3xTg increased cerebral perfusion by decreasing PRMT4 expression, restored the levels of APP and tau, maintained blood-brain barrier integrity by maintaining the expression of tight junction proteins, and preserved functional learning/memory.
Collapse
Affiliation(s)
- Mariana Sayuri Berto Udo
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Julia Zaccarelli-Magalhães
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Garrett Alan Clemons
- Department of Biomedical Science, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Cristiane Teresinha Citadin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Julia Langman
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Drew James Smith
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Luiz Henrique Matuguma
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Vetere LM, Galas AM, Vaughan N, Feng Y, Wick ZC, Philipsberg PA, Liobimova O, Fernandez-Ruiz A, Cai DJ, Shuman T. Medial entorhinal-hippocampal desynchronization parallels the emergence of memory impairment in a mouse model of Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633171. [PMID: 39868201 PMCID: PMC11761809 DOI: 10.1101/2025.01.15.633171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive impairments in episodic and spatial memory, as well as circuit and network-level dysfunction. While functional impairments in medial entorhinal cortex (MEC) and hippocampus (HPC) have been observed in patients and rodent models of AD, it remains unclear how communication between these regions breaks down in disease, and what specific physiological changes are associated with the onset of memory impairment. We used silicon probes to simultaneously record neural activity in MEC and hippocampus before or after the onset of spatial memory impairment in the 3xTg mouse model of AD pathology. We found that reduced hippocampal theta power, reduced MEC-CA1 theta coherence, and altered phase locking of MEC and hippocampal neurons all coincided with the emergence of spatial memory impairment in 3xTg mice. Together, these findings indicate that disrupted temporal coordination of neural activity in the MEC-hippocampal system parallels the emergence of memory impairment in a model of AD pathology.
Collapse
Affiliation(s)
| | | | - Nick Vaughan
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yu Feng
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | | | - Denise J Cai
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | |
Collapse
|
15
|
Mohd Murshid N, Mohd Sahardi NFN, Makpol S. Advancing Alzheimer's Disease Modelling by Developing a Refined Biomimetic Brain Microenvironment for Facilitating High-Throughput Screening of Pharmacological Treatment Strategies. Int J Mol Sci 2024; 26:241. [PMID: 39796097 PMCID: PMC11719782 DOI: 10.3390/ijms26010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) poses a significant worldwide health challenge, requiring novel approaches for improved models and treatment development. This comprehensive review emphasises the systematic development and improvement of a biomimetic brain environment to address the shortcomings of existing AD models and enhance the efficiency of screening potential drug treatments. We identify drawbacks in traditional models and emphasise the necessity for more physiologically accurate systems through an in-depth analysis of current literature. This review aims to study the development of an advanced AD model that accurately replicates key AD pathophysiological aspects using cutting-edge biomaterials and microenvironment design. Incorporating biomolecular elements like Tau proteins and beta-amyloid (Aβ) plaques improve the accuracy of illustrating disease mechanisms. The expected results involve creating a solid foundation for high-throughput screening with enhanced scalability, translational significance, and the possibility of speeding up drug discovery. Thus, this review fills the gaps in AD modelling and shows potential for creating precise and efficient drug treatments for AD.
Collapse
Affiliation(s)
- Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Nur Fatin Nabilah Mohd Sahardi
- Secretariat of Research and Innovation, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
16
|
Bartholomew SK, Winslow W, Sharma R, Pathak KV, Tallino S, Judd JM, Leon H, Turk J, Pirrotte P, Velazquez R. Glyphosate exposure exacerbates neuroinflammation and Alzheimer's disease-like pathology despite a 6-month recovery period in mice. J Neuroinflammation 2024; 21:316. [PMID: 39633366 PMCID: PMC11619132 DOI: 10.1186/s12974-024-03290-6] [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: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Glyphosate use in the United States (US) has increased each year since the introduction of glyphosate-tolerant crops in 1996, yet little is known about its effects on the brain. We recently found that C57BL/6J mice dosed with glyphosate for 14 days showed glyphosate and its major metabolite aminomethylphosphonic acid present in brain tissue, with corresponding increases in pro-inflammatory cytokine tumor necrosis factor-⍺ (TNF-⍺) in the brain and peripheral blood plasma. Since TNF-⍺ is elevated in neurodegenerative disorders such as Alzheimer's Disease (AD), in this study, we asked whether glyphosate exposure serves as an accelerant of AD pathogenesis. Additionally, whether glyphosate and aminomethylphosphonic acid remain in the brain after a recovery period has yet to be examined. METHODS We hypothesized that glyphosate exposure would induce neuroinflammation in control mice, while exacerbating neuroinflammation in AD mice, causing elevated Amyloid-β and tau pathology and worsening spatial cognition after recovery. We dosed 4.5-month-old 3xTg-AD and non-transgenic (NonTg) control mice with either 0, 50 or 500 mg/kg of glyphosate daily for 13 weeks followed by a 6-month recovery period. RESULTS We found that aminomethylphosphonic acid was detectable in the brains of 3xTg-AD and NonTg glyphosate-dosed mice despite the 6-month recovery. Glyphosate-dosed 3xTg-AD mice showed reduced survival, increased thigmotaxia in the Morris water maze, significant increases in the beta secretase enzyme (BACE-1) of amyloidogenic processing, amyloid-β (Aβ) 42 insoluble fractions, Aβ 42 plaque load and plaque size, and phosphorylated tau (pTau) at epitopes Threonine 181, Serine 396, and AT8 (Serine 202, Threonine 205). Notably, we found increased pro- and anti-inflammatory cytokines and chemokines persisting in both 3xTg-AD and NonTg brain tissue and in 3xTg-AD peripheral blood plasma. CONCLUSION Taken together, our results are the first to demonstrate that despite an extended recovery period, exposure to glyphosate elicits long-lasting pathological consequences. As glyphosate use continues to rise, more research is needed to elucidate the impact of this herbicide and its metabolites on the human brain, and their potential to contribute to dysfunctions observed in neurodegenerative diseases.
Collapse
Affiliation(s)
- Samantha K Bartholomew
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ritin Sharma
- Integrated Mass Spectrometry Shared Resources, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Khyatiben V Pathak
- Integrated Mass Spectrometry Shared Resources, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Savannah Tallino
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jessica M Judd
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Hector Leon
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Julie Turk
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Patrick Pirrotte
- Integrated Mass Spectrometry Shared Resources, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA.
| | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
17
|
Adlimoghaddam A, Fontaine KM, Albensi BC. Age- and sex-associated alterations in hypothalamic mitochondrial bioenergetics and inflammatory-associated signaling in the 3xTg mouse model of Alzheimer's disease. Biol Sex Differ 2024; 15:95. [PMID: 39587693 PMCID: PMC11587679 DOI: 10.1186/s13293-024-00671-7] [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: 08/28/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
Mitochondrial dysfunction and associated inflammatory signaling are pivotal in both aging and in Alzheimer's disease (AD). Studies have also shown that hypothalamic function is affected in AD. The hypothalamus may be a target for AD drugs given that mitochondrial alterations are observed in the hypothalamus. This study investigated how age and sex affect mitochondrial bioenergetics and inflammatory signaling in the hypothalamic mitochondria of 3xTg and control mice at 2, 6, and 13 months, aiming to enhance our understanding of these processes in aging and AD. Parameters included oxygen consumption rates, expression levels of subunits comprising mitochondrial complexes I-V, the enzymatic activity of cytochrome c oxidase (COX), transcription factors associated with inflammation such as NF-κB, pIκB-α, Nrf2, and other inflammatory biomarkers. Hypothalamic mitochondrial dysfunction was observed in 3xTg females as early as 2 months, but no changes were detected in 3xTg males until 6 months of age. In 3xTg mice, subunit expression levels for mitochondrial complexes I-II were significantly reduced in both sexes. Significant sex-based differences in COX activity were also observed at 13 months of age, with levels being lower in females compared to males. In addition, significant sex differences were indicated in NF-κB, pIκB-α, Nrf2, and other inflammatory biomarkers at different age groups during normal aging and AD progression. These findings highlight important sex differences in hypothalamic bioenergetics and inflammation, offering insights into potential new targets for preventing and/or treating AD.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Kyle M Fontaine
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Benedict C Albensi
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
18
|
Blankenship HE, Carter KA, Pham KD, Cassidy NT, Markiewicz AN, Thellmann MI, Sharpe AL, Freeman WM, Beckstead MJ. VTA dopamine neurons are hyperexcitable in 3xTg-AD mice due to casein kinase 2-dependent SK channel dysfunction. Nat Commun 2024; 15:9673. [PMID: 39516200 PMCID: PMC11549218 DOI: 10.1038/s41467-024-53891-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) patients exhibit neuropsychiatric symptoms that extend beyond classical cognitive deficits, suggesting involvement of subcortical areas. Here, we investigated the role of midbrain dopamine (DA) neurons in AD using the amyloid + tau-driven 3xTg-AD mouse model. We found deficits in reward-based operant learning in AD mice, suggesting possible VTA DA neuron dysregulation. Physiological assessment revealed hyperexcitability and disrupted firing in DA neurons caused by reduced activity of small-conductance calcium-activated potassium (SK) channels. RNA sequencing from contents of single patch-clamped DA neurons (Patch-seq) identified up-regulation of the SK channel modulator casein kinase 2 (CK2), which we corroborated by immunohistochemical protein analysis. Pharmacological inhibition of CK2 restored SK channel activity and normal firing patterns in 3xTg-AD mice. These findings identify a mechanism of ion channel dysregulation in VTA DA neurons that could contribute to behavioral abnormalities in AD, paving the way for novel treatment strategies.
Collapse
Affiliation(s)
- Harris E Blankenship
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelsey A Carter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kevin D Pham
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Nina T Cassidy
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Andrea N Markiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael I Thellmann
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
19
|
De Plano LM, Saitta A, Oddo S, Caccamo A. Navigating Alzheimer's Disease Mouse Models: Age-Related Pathology and Cognitive Deficits. Biomolecules 2024; 14:1405. [PMID: 39595581 PMCID: PMC11592094 DOI: 10.3390/biom14111405] [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: 09/27/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Since the mid-1990s, scientists have been generating mouse models of Alzheimer's disease to elucidate key mechanisms underlying the onset and progression of the disease and aid in developing potential therapeutic approaches. The first successful mouse model of Alzheimer's disease was reported in 1995 with the generation of the PDAPP mice, which were obtained by the overexpression of gene coding for the amyloid precursor protein (APP). Since then, scientists have used different approaches to develop other APP overexpression mice, mice overexpressing tau, or a combination of them. More recently, Saito and colleagues generated a mouse model by knocking in mutations associated with familial Alzheimer's disease into the APP gene. In this review, we will describe the most used animal models and provide a practical guide for the disease's age of onset and progression. We believe that this guide will be valuable for the planning and experimental design of studies utilizing these mouse models.
Collapse
Affiliation(s)
| | | | | | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (L.M.D.P.); (A.S.); (S.O.)
| |
Collapse
|
20
|
Colín-Martínez E, Espino-de-la-Fuente C, Arias C. Age- and Sex-Associated Wnt Signaling Dysregulation is Exacerbated from the Early Stages of Neuropathology in an Alzheimer's Disease Model. Neurochem Res 2024; 49:3094-3104. [PMID: 39167347 PMCID: PMC11449975 DOI: 10.1007/s11064-024-04224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024]
Abstract
Emerging studies suggest that Wnt signaling is dysregulated in the brains of AD patients, suggesting that this pathway may also contribute to disease progression. However, it remains to be determined whether alterations in the Wnt pathway are the cause or consequence of this disease and which elements of Wnt signaling mainly contribute to the appearance of AD histopathological markers early in disease compared to what occurs during normal aging. The present study aimed to describe the status of several canonical Wnt pathway components and the expression of the AD marker p-tau in the hippocampi of female and male 3xTg-AD mice during disease progression compared to those during normal aging. We analyzed the levels of the canonical Wnt components Wnt7a, Dkk-1, LRP6 and GSK3β as well as the levels of p-tau and BDNF at 3, 6, 9-12 and 18 months of age. We found a gradual increase in Dkk-1 levels during aging prior to Wnt7a and LRP5/6 depletion, which was strongly exacerbated in 3xTg-AD mice even at young ages and correlated with GSK3β activation and p-tau-S202/Thr205 expression. Dkk-1 upregulation, as well as the level of p-tau, was significantly greater in females than in males. Our results suggest that Dkk-1 upregulation is involved in the expression of several features of AD at early stages, which supports the possibility of positively modulating the canonical Wnt pathway as a therapeutic tool to delay this disease at early stages.
Collapse
Affiliation(s)
- Elizabeth Colín-Martínez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - César Espino-de-la-Fuente
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México.
| |
Collapse
|
21
|
Bostick JW, Connerly TJ, Thron T, Needham BD, de Castro Fonseca M, Kaddurah-Daouk R, Knight R, Mazmanian SK. The microbiome shapes immunity in a sex-specific manner in mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593011. [PMID: 38766238 PMCID: PMC11100721 DOI: 10.1101/2024.05.07.593011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
INTRODUCTION : Preclinical studies reveal that the microbiome broadly affects immune responses and deposition and/or clearance of amyloid-beta (Aβ) in mouse models of Alzheimer's disease (AD). Whether the microbiome shapes central and peripheral immune profiles in AD models remains unknown. METHODS : We examined adaptive immune responses in two mouse models containing AD- related genetic predispositions (3xTg and 5xFAD) in the presence or absence of the microbiome. RESULTS : T and B cells were altered in brain-associated and systemic immune tissues between genetic models and wildtype mice, with earlier signs of immune activity in females. Systemic immune responses were modulated by the microbiome and differed by sex. Further, the absence of a microbiome in germ-free mice resulted in reduced cognitive deficits, primarily in females. DISCUSSION : These data reveal sexual dimorphism in early signs of immune activity and microbiome effects, and highlight an interesting interaction between sex and the microbiome in mouse models of AD.
Collapse
|
22
|
Kong Y, Maschio CA, Shi X, Xie F, Zuo C, Konietzko U, Shi K, Rominger A, Xiao J, Huang Q, Nitsch RM, Guan Y, Ni R. Relationship Between Reactive Astrocytes, by [ 18F]SMBT-1 Imaging, with Amyloid-Beta, Tau, Glucose Metabolism, and TSPO in Mouse Models of Alzheimer's Disease. Mol Neurobiol 2024; 61:8387-8401. [PMID: 38502413 DOI: 10.1007/s12035-024-04106-7] [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: 09/25/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Reactive astrocytes play an important role in the development of Alzheimer's disease (AD). Here, we aimed to investigate the temporospatial relationships among monoamine oxidase-B, tau and amyloid-β (Aβ), translocator protein, and glucose metabolism by using multitracer imaging in AD transgenic mouse models. Positron emission tomography (PET) imaging with [18F]SMBT-1 (monoamine oxidase-B), [18F]florbetapir (Aβ), [18F]PM-PBB3 (tau), [18F]fluorodeoxyglucose (FDG), and [18F]DPA-714 (translocator protein) was carried out in 5- and 10-month-old APP/PS1, 11-month-old 3×Tg mice, and aged-matched wild-type mice. The brain regional referenced standard uptake value (SUVR) was computed with the cerebellum as the reference region. Immunofluorescence staining was performed on mouse brain tissue slices. [18F]SMBT-1 and [18F]florbetapir SUVRs were greater in the cortex and hippocampus of 10-month-old APP/PS1 mice than in those of 5-month-old APP/PS1 mice and wild-type mice. No significant difference in the regional [18F]FDG or [18F]DPA-714 SUVRs was observed in the brains of 5- or 10-month-old APP/PS1 mice or wild-type mice. No significant difference in the SUVRs of any tracer was observed between 11-month-old 3×Tg mice and age-matched wild-type mice. A positive correlation between the SUVRs of [18F]florbetapir and [18F]DPA-714 in the cortex and hippocampus was observed among the transgenic mice. Immunostaining validated the distribution of MAO-B and limited Aβ and tau pathology in 11-month-old 3×Tg mice; and Aβ deposits in brain tissue from 10-month-old APP/PS1 mice. In summary, these findings provide in vivo evidence that an increase in astrocyte [18F]SMBT-1 accompanies Aβ accumulation in APP/PS1 models of AD amyloidosis.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland
| | - Xuefeng Shi
- Qinghai Provincial People's Hospital, Xining, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland.
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Geigenmüller JN, Tari AR, Wisloff U, Walker TL. The relationship between adult hippocampal neurogenesis and cognitive impairment in Alzheimer's disease. Alzheimers Dement 2024; 20:7369-7383. [PMID: 39166771 PMCID: PMC11485317 DOI: 10.1002/alz.14179] [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: 05/14/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Neurogenesis persists throughout adulthood in the hippocampus and contributes to specific cognitive functions. In Alzheimer's disease (AD), the hippocampus is affected by pathology and functional impairment early in the disease. Human AD patients have reduced adult hippocampal neurogenesis (AHN) levels compared to age-matched healthy controls. Similarly, rodent AD models show a decrease in AHN before the onset of the classical hallmarks of AD pathology. Conversely, enhancement of AHN can protect against AD pathology and ameliorate memory deficits in both rodents and humans. Therefore, impaired AHN may be a contributing factor of AD-associated cognitive decline, rather than an effect of it. In this review we outline the regulation and function of AHN in healthy individuals, and highlight the relationship between AHN dysfunction and cognitive impairments in AD. The existence of AHN in humans and its relevance in AD patients will also be discussed, with an outlook toward future research directions. HIGHLIGHTS: Adult hippocampal neurogenesis occurs in the brains of mammals including humans. Adult hippocampal neurogenesis is reduced in Alzheimer's disease in humans and animal models.
Collapse
Affiliation(s)
| | - Atefe R. Tari
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Department of Neurology and Clinical NeurophysiologySt. Olavs University Hospital, Trondheim University HospitalTrondheimNorway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Tara L. Walker
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneAustralia
| |
Collapse
|
24
|
Mackey-Alfonso SE, Butler MJ, Taylor AM, Williams-Medina AR, Muscat SM, Fu H, Barrientos RM. Short-term high fat diet impairs memory, exacerbates the neuroimmune response, and evokes synaptic degradation via a complement-dependent mechanism in a mouse model of Alzheimer's disease. Brain Behav Immun 2024; 121:56-69. [PMID: 39043341 DOI: 10.1016/j.bbi.2024.07.021] [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: 04/01/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disease characterized by profound memory impairments, synaptic loss, neuroinflammation, and hallmark pathological markers. High-fat diet (HFD) consumption increases the risk of developing AD even after controlling for metabolic syndrome, pointing to a role of the diet itself in increasing risk. In AD, the complement system, an arm of the immune system which normally tags redundant or damaged synapses for pruning, becomes pathologically overactivated leading to tagging of healthy synapses. While the unhealthy diet to AD link is strong, the underlying mechanisms are not well understood in part due to confounding variables associated with long-term HFD which can independently influence the brain. Therefore, we experimented with a short-term diet regimen to isolate the diet's impact on brain function without causing obesity. This project investigated the effect of short-term HFD on 1) memory, 2) neuroinflammation including complement, 3) AD pathology markers, 4) synaptic markers, and 5) in vitro microglial synaptic phagocytosis in the 3xTg-AD mouse model. Following the consumption of either standard chow or HFD, 3xTg-AD and non-Tg mice were tested for memory impairments. In a separate cohort of mice, levels of hippocampal inflammatory markers, complement proteins, AD pathology markers, and synaptic markers were measured. For the last set of experiments, BV2 microglial phagocytosis of synapses was evaluated. Synaptoneurosomes isolated from the hippocampus of 3xTg-AD mice fed chow or HFD were incubated with equal numbers of BV2 microglia. The number of BV2 microglia that phagocytosed synaptoneurosomes was tracked over time with a live-cell imaging assay. Finally, we incubated BV2 microglia with a complement receptor inhibitor (NIF) and repeated the assay. Behavioral analysis showed 3xTg-AD mice had significantly impaired long-term contextual and cued fear memory compared to non-Tg mice that was further impaired by HFD. HFD significantly increased inflammatory markers and complement expression while decreasing synaptic marker expression only in 3xTg-AD mice, without altering AD pathology markers. Synaptoneurosomes from HFD-fed 3xTg-AD mice were phagocytosed at a significantly higher rate than those from chow-fed mice, suggesting the synapses were altered by HFD. The complement receptor inhibitor blocked this effect in a dose-dependent manner, demonstrating the HFD-mediated increase in phagocytosis was complement dependent. This study indicates HFD consumption increases neuroinflammation and over-activates the complement cascade in 3xTg-AD mice, resulting in poorer memory. The in vitro data point to complement as a potential mechanistic culprit and therapeutic target underlying HFD's influence in increasing cognitive vulnerability to AD.
Collapse
Affiliation(s)
- Sabrina E Mackey-Alfonso
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Ashton M Taylor
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | | | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
25
|
Soelter TM, Howton TC, Wilk EJ, Whitlock JH, Clark AD, Birnbaum A, Patterson DC, Cortes CJ, Lasseigne BN. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595199. [PMID: 38826305 PMCID: PMC11142088 DOI: 10.1101/2024.05.21.595199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss and cognitive decline, affecting behavior, speech, and motor abilities. The neuropathology of AD includes the formation of extracellular amyloid-β plaque and intracellular neurofibrillary tangles of phosphorylated tau, along with neuronal loss. While neuronal loss is an AD hallmark, cell-cell communication between neuronal and non-neuronal cell populations maintains neuronal health and brain homeostasis. To study changes in cellcell communication during disease progression, we performed snRNA-sequencing of the hippocampus from female 3xTg-AD and wild-type littermates at 6 and 12 months. We inferred differential cell-cell communication between 3xTg-AD and wild-type mice across time points and between senders (astrocytes, microglia, oligodendrocytes, and OPCs) and receivers (excitatory and inhibitory neurons) of interest. We also assessed the downstream effects of altered glia-neuron communication using pseudobulk differential gene expression, functional enrichment, and gene regulatory analyses. We found that glia-neuron communication is increasingly dysregulated in 12-month 3xTg-AD mice. We also identified 23 AD-associated ligand-receptor pairs that are upregulated in the 12-month-old 3xTg-AD hippocampus. Our results suggest increased AD association of interactions originating from microglia. Signaling mediators were not significantly differentially expressed but showed altered gene regulation and TF activity. Our findings indicate that altered glia-neuron communication is increasingly dysregulated and affects the gene regulatory mechanisms in neurons of 12-month-old 3xTg-AD mice.
Collapse
Affiliation(s)
- Tabea M. Soelter
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jordan H. Whitlock
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Allison Birnbaum
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Dalton C. Patterson
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Constanza J. Cortes
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
26
|
Michaud F, Francavilla R, Topolnik D, Iloun P, Tamboli S, Calon F, Topolnik L. Altered firing output of VIP interneurons and early dysfunctions in CA1 hippocampal circuits in the 3xTg mouse model of Alzheimer's disease. eLife 2024; 13:RP95412. [PMID: 39264364 PMCID: PMC11392531 DOI: 10.7554/elife.95412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive memory decline, and alterations in hippocampal function are among the earliest pathological features observed in human and animal studies. GABAergic interneurons (INs) within the hippocampus coordinate network activity, among which type 3 interneuron-specific (I-S3) cells expressing vasoactive intestinal polypeptide and calretinin play a crucial role. These cells provide primarily disinhibition to principal excitatory cells (PCs) in the hippocampal CA1 region, regulating incoming inputs and memory formation. However, it remains unclear whether AD pathology induces changes in the activity of I-S3 cells, impacting the hippocampal network motifs. Here, using young adult 3xTg-AD mice, we found that while the density and morphology of I-S3 cells remain unaffected, there were significant changes in their firing output. Specifically, I-S3 cells displayed elongated action potentials and decreased firing rates, which was associated with a reduced inhibition of CA1 INs and their higher recruitment during spatial decision-making and object exploration tasks. Furthermore, the activation of CA1 PCs was also impacted, signifying early disruptions in CA1 network functionality. These findings suggest that altered firing patterns of I-S3 cells might initiate early-stage dysfunction in hippocampal CA1 circuits, potentially influencing the progression of AD pathology.
Collapse
Affiliation(s)
- Felix Michaud
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Ruggiero Francavilla
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Dimitry Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Parisa Iloun
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Suhel Tamboli
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Frederic Calon
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| |
Collapse
|
27
|
Lin X, Chen C, Chen J, Zhu C, Zhang J, Su R, Chen S, Weng S, Chang X, Lin S, Chen Y, Li J, Lin L, Zhou J, Guo Z, Yu G, Shao W, Hu H, Wu S, Zhang Q, Li H, Zheng F. Long Noncoding RNA NR_030777 Alleviates Cobalt Nanoparticles-Induced Neurodegenerative Damage by Promoting Autophagosome-Lysosome Fusion. ACS NANO 2024; 18:24872-24897. [PMID: 39197041 PMCID: PMC11394346 DOI: 10.1021/acsnano.4c05249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/30/2024]
Abstract
Potential exposure to cobalt nanoparticles (CoNPs) occurs in various fields, including hard alloy industrial production, the increasing use of new energy lithium-ion batteries, and millions of patients with metal-on-metal joint prostheses. Evidence from human, animal, and in vitro experiments suggests a close relationship between CoNPs and neurotoxicity. However, a systematic assessment of central nervous system (CNS) impairment due to CoNPs exposure and the underlying molecular mechanisms is lacking. In this study, we found that CoNPs induced neurodegenerative damage both in vivo and in vitro, including cognitive impairment, β-amyloid deposition and Tau hyperphosphorylation. CoNPs promoted the formation of autophagosomes and impeding autophagosomal-lysosomal fusion in vivo and in vitro, leading to toxic protein accumulation. Moreover, CoNPs exposure reduced the level of transcription factor EB (TFEB) and the abundance of lysosome, causing a blockage in autophagosomal-lysosomal fusion. Interestingly, overexpression of long noncoding RNA NR_030777 mitigated CoNPs-induced neurodegenerative damage in both in vivo and in vitro models. Fluorescence in situ hybridization assay revealed that NR_030777 directly binds and stabilizes TFEB mRNA, alleviating the blockage of autophagosomal-lysosomal fusion and ultimately restoring neurodegeneration induced by CoNPs in vivo and in vitro. In summary, our study demonstrates that autophagic dysfunction is the main toxic mechanism of neurodegeneration upon CoNPs exposure and NR_030777 plays a crucial role in CoNPs-induced autophagic dysfunction. Additionally, the proposed adverse outcome pathway contributes to a better understanding of CNS toxicity assessment of CoNPs.
Collapse
Affiliation(s)
- Xinpei Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Cheng Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jinxiang Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Canlin Zhu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiajun Zhang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ruiqi Su
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shujia Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shucan Weng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Xiangyu Chang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shengsong Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Yilong Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiamei Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ling Lin
- Public
Technology Service Center, Fujian Medical
University, Fuzhou, Fujian Province 350122, China
| | - Jinfu Zhou
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Medical
Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health
Hospital College of Clinical Medicine for Obstetrics & Gynecology
and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province 350001, China
| | - Zhenkun Guo
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Guangxia Yu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Wenya Shao
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Hong Hu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Siying Wu
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Department
of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Qunwei Zhang
- Department
of Epidemiology and Population Health, School of Public Health and
Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, Kentucky 40292, United States
| | - Huangyuan Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Fuli Zheng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| |
Collapse
|
28
|
Park G, Kadyan S, Hochuli N, Salazar G, Laitano O, Chakrabarty P, Efron PA, Zafar MA, Wilber A, Nagpal R. An Enteric Bacterial Infection Triggers Neuroinflammation and Neurobehavioral Impairment in 3xTg-AD Transgenic Mice. J Infect Dis 2024; 230:S95-S108. [PMID: 39255397 PMCID: PMC11385593 DOI: 10.1093/infdis/jiae165] [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: 01/14/2024] [Accepted: 03/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae is infamous for hospital-acquired infections and sepsis, which have also been linked to Alzheimer disease (AD)-related neuroinflammatory and neurodegenerative impairment. However, its causative and mechanistic role in AD pathology remains unstudied. METHODS A preclinical model of K. pneumoniae enteric infection and colonization is developed in an AD model (3xTg-AD mice) to investigate whether and how K. pneumoniae pathogenesis exacerbates neuropathogenesis via the gut-blood-brain axis. RESULTS K. pneumoniae, particularly under antibiotic-induced dysbiosis, was able to translocate from the gut to the bloodstream by penetrating the gut epithelial barrier. Subsequently, K. pneumoniae infiltrated the brain by breaching the blood-brain barrier. Significant neuroinflammatory phenotype was observed in mice with K. pneumoniae brain infection. K. pneumoniae-infected mice also exhibited impaired neurobehavioral function and elevated total tau levels in the brain. Metagenomic analyses revealed an inverse correlation of K. pneumoniae with gut biome diversity and commensal bacteria, highlighting how antibiotic-induced dysbiosis triggers an enteroseptic "pathobiome" signature implicated in gut-brain perturbations. CONCLUSIONS The findings demonstrate how infectious agents following hospital-acquired infections and consequent antibiotic regimen may induce gut dysbiosis and pathobiome and increase the risk of sepsis, thereby increasing the predisposition to neuroinflammatory and neurobehavioral impairments via breaching the gut-blood-brain barrier.
Collapse
Affiliation(s)
- Gwoncheol Park
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
| | - Saurabh Kadyan
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
| | - Nathaniel Hochuli
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
| | - Gloria Salazar
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Philip A Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - M Ammar Zafar
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Aaron Wilber
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Ravinder Nagpal
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
29
|
Chen YY, Chang CJ, Liang YW, Tseng HY, Li SJ, Chang CW, Wu YT, Shao HH, Chen PC, Lai ML, Deng WC, Hsu R, Lo YC. Utilizing diffusion tensor imaging as an image biomarker in exploring the therapeutic efficacy of forniceal deep brain stimulation in a mice model of Alzheimer's disease. J Neural Eng 2024; 21:056003. [PMID: 39230033 DOI: 10.1088/1741-2552/ad7322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
Objective.With prolonged life expectancy, the incidence of memory deficits, especially in Alzheimer's disease (AD), has increased. Although multiple treatments have been evaluated, no promising treatment has been found to date. Deep brain stimulation (DBS) of the fornix area was explored as a possible treatment because the fornix is intimately connected to memory-related areas that are vulnerable in AD; however, a proper imaging biomarker for assessing the therapeutic efficiency of forniceal DBS in AD has not been established.Approach.This study assessed the efficacy and safety of DBS by estimating the optimal intersection volume between the volume of tissue activated and the fornix. Utilizing a gold-electroplating process, the microelectrode's surface area on the neural probe was increased, enhancing charge transfer performance within potential water window limits. Bilateral fornix implantation was conducted in triple-transgenic AD mice (3 × Tg-AD) and wild-type mice (strain: B6129SF1/J), with forniceal DBS administered exclusively to 3 × Tg-AD mice in the DBS-on group. Behavioral tasks, diffusion tensor imaging (DTI), and immunohistochemistry (IHC) were performed in all mice to assess the therapeutic efficacy of forniceal DBS.Main results.The results illustrated that memory deficits and increased anxiety-like behavior in 3 × Tg-AD mice were rescued by forniceal DBS. Furthermore, forniceal DBS positively altered DTI indices, such as increasing fractional anisotropy (FA) and decreasing mean diffusivity (MD), together with reducing microglial cell and astrocyte counts, suggesting a potential causal relationship between revised FA/MD and reduced cell counts in the anterior cingulate cortex, hippocampus, fornix, amygdala, and entorhinal cortex of 3 × Tg-AD mice following forniceal DBS.Significance.The efficacy of forniceal DBS in AD can be indicated by alterations in DTI-based biomarkers reflecting the decreased activation of glial cells, suggesting reduced neural inflammation as evidenced by improvements in memory and anxiety-like behavior.
Collapse
Affiliation(s)
- You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| | - Chih-Ju Chang
- Department of Neurosurgery, Cathay General Hospital, No. 280, Sec. 4, Renai Rd., Taipei 10629, Taiwan, Republic of China
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., New Taipei City 242062, Taiwan, Republic of China
| | - Yao-Wen Liang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Hsin-Yi Tseng
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| | - Ssu-Ju Li
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Ching-Wen Chang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Yen-Ting Wu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Huai-Hsuan Shao
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong St., Taipei 11221, Taiwan, Republic of China
| | - Po-Chun Chen
- Department of Materials and Mineral Resources Engineering, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan, Republic of China
| | - Ming-Liang Lai
- Graduate Institute of Intellectual Property, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan, Republic of China
| | - Wen-Chun Deng
- Departments of Neurosurgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, No.222, Maijin Rd., Keelung 20400, Taiwan, Republic of China
| | - RuSiou Hsu
- Department of Ophthalmology, Stanford University, 1651 Page Mill Rd., Palo Alto, CA 94304, United States of America
| | - Yu-Chun Lo
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, 12F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., New Taipei City 23564, Taiwan, Republic of China
| |
Collapse
|
30
|
Han J, Sul JH, Lee J, Kim E, Kim HK, Chae M, Lim J, Kim J, Kim C, Kim JS, Cho Y, Park JH, Cho YW, Jo DG. Engineered exosomes with a photoinducible protein delivery system enable CRISPR-Cas-based epigenome editing in Alzheimer's disease. Sci Transl Med 2024; 16:eadi4830. [PMID: 39110781 DOI: 10.1126/scitranslmed.adi4830] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 01/24/2024] [Accepted: 07/18/2024] [Indexed: 02/16/2025]
Abstract
Effective intracellular delivery of therapeutic proteins can potentially treat a wide array of diseases. However, efficient delivery of functional proteins across the cell membrane remains challenging. Exosomes are nanosized vesicles naturally secreted by various types of cells and may serve as promising nanocarriers for therapeutic biomolecules. Here, we engineered exosomes equipped with a photoinducible cargo protein release system, termed mMaple3-mediated protein loading into and release from exosome (MAPLEX), in which cargo proteins can be loaded into the exosomes by fusing them with photocleavable protein (mMaple3)-conjugated exosomal membrane markers and subsequently released from the exosomal membrane by inducing photocleavage with blue light illumination. Using this system, we first induced transcriptional regulation by delivering octamer-binding transcription factor 4 and SRY-box transcription factor 2 to fibroblasts in vitro. Second, we induced in vivo gene recombination in Cre reporter mice by delivering Cre recombinase. Last, we achieved targeted epigenome editing in the brains of 5xFAD and 3xTg-AD mice, two models of Alzheimer's disease. Administration of MAPLEXs loaded with β-site amyloid precursor protein cleaving enzyme 1 (Bace1)-targeting single guide RNA-incorporated dCas9 ribonucleoprotein complexes, coupled with the catalytic domain of DNA methyltransferase 3A, resulted in successful methylation of the targeted CpG sites within the Bace1 promoter. This approach led to a significant reduction in Bace1 expression, improved recognition memory impairment, and reduced amyloid pathology in 5xFAD and 3xTg-AD mice. These results suggest that MAPLEX is an efficient intracellular protein delivery system that can deliver diverse therapeutic proteins for multiple diseases.
Collapse
Affiliation(s)
- Jihoon Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Hoon Sul
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Eunae Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Hark Kyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Minshik Chae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jeein Lim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jongho Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Chanhee Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jun-Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- ExoStem Tech Inc., Ansan 15588, Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea
| | - Yong Woo Cho
- ExoStem Tech Inc., Ansan 15588, Korea
- Department of Chemical Engineering, Hanyang University, Ansan 15588, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- ExoStem Tech Inc., Ansan 15588, Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
31
|
Uchiumi O, Zou J, Yamaki S, Hori Y, Ono M, Yamamoto R, Kato N. Disruption of sphingomyelin synthase 2 gene alleviates cognitive impairment in a mouse model of Alzheimer's disease. Brain Res 2024; 1835:148934. [PMID: 38609029 DOI: 10.1016/j.brainres.2024.148934] [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: 02/15/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
The membrane raft accommodates the key enzymes synthesizing amyloid β (Aβ). One of the two characteristic components of the membrane raft, cholesterol, is well known to promote the key enzymes that produce amyloid-β (Aβ) and exacerbate Alzheimer's disease (AD) pathogenesis. Given that the raft is a physicochemical platform for the sound functioning of embedded bioactive proteins, the other major lipid component sphingomyelin may also be involved in AD. Here we knocked out the sphingomyelin synthase 2 gene (SMS2) in 3xTg AD model mice by hybridization, yielding SMS2KO mice (4S mice). The novel object recognition test in 9/10-month-old 4S mice showed that cognitive impairment in 3xTg mice was alleviated by SMS2KO, though performance in the Morris water maze (MWM) was not improved. The tail suspension test detected a depressive trait in 4S mice, which may have hindered the manifestation of performance in the wet, stressful environment of MWM. In the hippocampal CA1, hyperexcitability in 3xTg was also found alleviated by SMS2KO. In the hippocampal dentate gyrus of 4S mice, the number of neurons positive with intracellular Aβ or its precursor proteins, the hallmark of young 3xTg mice, is reduced to one-third, suggesting an SMS2KO-led suppression of syntheses of those peptides in the dentate gyrus. Although we previously reported that large-conductance calcium-activated potassium (BK) channels are suppressed in 3xTg mice and their recovery relates to cognitive amelioration, no changes occurred by hybridization. Sphingomyelin in the membrane raft may serve as a novel target for AD drugs.
Collapse
Affiliation(s)
- Osamu Uchiumi
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Jingyu Zou
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan; First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan.
| |
Collapse
|
32
|
Butler CA, Mendoza Arvilla A, Milinkeviciute G, Da Cunha C, Kawauchi S, Rezaie N, Liang HY, Javonillo D, Thach A, Wang S, Collins S, Walker A, Shi K, Neumann J, Gomez‐Arboledas A, Henningfield CM, Hohsfield LA, Mapstone M, Tenner AJ, LaFerla FM, Mortazavi A, MacGregor GR, Green KN. The Abca7 V1613M variant reduces Aβ generation, plaque load, and neuronal damage. Alzheimers Dement 2024; 20:4914-4934. [PMID: 38506634 PMCID: PMC11247689 DOI: 10.1002/alz.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/21/2024]
Abstract
BACKGROUND Variants in ABCA7, a member of the ABC transporter superfamily, have been associated with increased risk for developing late onset Alzheimer's disease (LOAD). METHODS CRISPR-Cas9 was used to generate an Abca7V1613M variant in mice, modeling the homologous human ABCA7V1599M variant, and extensive characterization was performed. RESULTS Abca7V1613M microglia show differential gene expression profiles upon lipopolysaccharide challenge and increased phagocytic capacity. Homozygous Abca7V1613M mice display elevated circulating cholesterol and altered brain lipid composition. When crossed with 5xFAD mice, homozygous Abca7V1613M mice display fewer Thioflavin S-positive plaques, decreased amyloid beta (Aβ) peptides, and altered amyloid precursor protein processing and trafficking. They also exhibit reduced Aβ-associated inflammation, gliosis, and neuronal damage. DISCUSSION Overall, homozygosity for the Abca7V1613M variant influences phagocytosis, response to inflammation, lipid metabolism, Aβ pathology, and neuronal damage in mice. This variant may confer a gain of function and offer a protective effect against Alzheimer's disease-related pathology. HIGHLIGHTS ABCA7 recognized as a top 10 risk gene for developing Alzheimer's disease. Loss of function mutations result in increased risk for LOAD. V1613M variant reduces amyloid beta plaque burden in 5xFAD mice. V1613M variant modulates APP processing and trafficking in 5xFAD mice. V1613M variant reduces amyloid beta-associated damage in 5xFAD mice.
Collapse
Affiliation(s)
- Claire A. Butler
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Adrian Mendoza Arvilla
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Narges Rezaie
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Heidi Y. Liang
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dominic Javonillo
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Annie Thach
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kai‐Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Lindsay A. Hohsfield
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Mark Mapstone
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology & BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Ali Mortazavi
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAR, Office of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
33
|
Jong Huat T, Camats-Perna J, Newcombe EA, Onraet T, Campbell D, Sucic JT, Martini A, Forner S, Mirzaei M, Poon W, LaFerla FM, Medeiros R. The impact of astrocytic NF-κB on healthy and Alzheimer's disease brains. Sci Rep 2024; 14:14305. [PMID: 38906984 PMCID: PMC11192733 DOI: 10.1038/s41598-024-65248-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
Astrocytes play a role in healthy cognitive function and Alzheimer's disease (AD). The transcriptional factor nuclear factor-κB (NF-κB) drives astrocyte diversity, but the mechanisms are not fully understood. By combining studies in human brains and animal models and selectively manipulating NF-κB function in astrocytes, we deepened the understanding of the role of astrocytic NF-κB in brain health and AD. In silico analysis of bulk and cell-specific transcriptomic data revealed the association of NF-κB and astrocytes in AD. Confocal studies validated the higher level of p50 NF-κB and phosphorylated-p65 NF-κB in glial fibrillary acidic protein (GFAP)+-astrocytes in AD versus non-AD subjects. In the healthy mouse brain, chronic activation of astrocytic NF-κB disturbed the proteomic milieu, causing a loss of mitochondrial-associated proteins and the rise of inflammatory-related proteins. Sustained NF-κB signaling also led to microglial reactivity, production of pro-inflammatory mediators, and buildup of senescence-related protein p16INK4A in neurons. However, in an AD mouse model, NF-κB inhibition accelerated β-amyloid and tau accumulation. Molecular biology studies revealed that astrocytic NF-κB activation drives the increase in GFAP and inflammatory proteins and aquaporin-4, a glymphatic system protein that assists in mitigating AD. Our investigation uncovered fundamental mechanisms by which NF-κB enables astrocytes' neuroprotective and neurotoxic responses in the brain.
Collapse
Affiliation(s)
- Tee Jong Huat
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, QLD, Australia
| | - Judith Camats-Perna
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Estella A Newcombe
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Tessa Onraet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel Campbell
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Josiah T Sucic
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Alessandra Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Mehdi Mirzaei
- Clinical Medicine Department, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Wayne Poon
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA, 92697-4545, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Rodrigo Medeiros
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA, 92697-4545, USA.
| |
Collapse
|
34
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
35
|
Kong Y, Cao L, Wang J, Zhuang J, Liu Y, Bi L, Qiu Y, Hou Y, Huang Q, Xie F, Yang Y, Shi K, Rominger A, Guan Y, Jin H, Ni R. Increased Cerebral Level of P2X7R in a Tauopathy Mouse Model by PET Using [ 18F]GSK1482160. ACS Chem Neurosci 2024; 15:2112-2120. [PMID: 38776461 PMCID: PMC11157487 DOI: 10.1021/acschemneuro.4c00067] [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: 01/29/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Neuroinflammation plays an important role in Alzheimer's disease and primary tauopathies. The aim of the current study was to map [18F]GSK1482160 for imaging of purinergic P2X7R in Alzheimer's disease and primary tauopathy mouse models. Small animal PET was performed using [18F]GSK1482160 in widely used mouse models of Alzheimer's disease (APP/PS1, 5×FAD, and 3×Tg), 4-repeat tauopathy (rTg4510) mice, and age-matched wild-type mice. Increased uptake of [18F]GSK1482160 was observed in the brains of 7-month-old rTg4510 mice compared to wild-type mice and compared to 3-month-old rTg4510 mice. A positive correlation between hippocampal tau [18F]APN-1607 and [18F]GSK1482160 uptake was found in rTg4510 mice. No significant differences in the uptake of [18F]GSK1482160 was observed for APP/PS1 mice, 5×FAD mice, or 3×Tg mice. Immunofluorescence staining further indicated the distribution of P2X7Rs in the brains of 7-month-old rTg4510 mice with accumulation of tau inclusion. These findings provide in vivo imaging evidence for an increased level of P2X7R in the brains of tauopathy mice.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Lei Cao
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
- Institute
for Regenerative Medicine, University of
Zurich, Zurich 8952, Switzerland
| | - Jiao Wang
- Lab
of Molecular
Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Junyi Zhuang
- Lab
of Molecular
Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yongshan Liu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Lei Bi
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Yifan Qiu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Yuyi Hou
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Qi Huang
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Fang Xie
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Yunhao Yang
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Kuangyu Shi
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
| | - Axel Rominger
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
| | - Yihui Guan
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Hongjun Jin
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Ruiqing Ni
- Institute
for Regenerative Medicine, University of
Zurich, Zurich 8952, Switzerland
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
- Institute
for Biomedical Engineering, University of
Zurich & ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
36
|
Tran KM, Kwang N, Gomez-Arboledas A, Kawauchi S, Mar C, Chao D, Da Cunha C, Wang S, Collins S, Walker A, Shi KX, Alcantara JA, Neumann J, Tenner AJ, LaFerla FM, Hohsfield LA, Swarup V, MacGregor GR, Green KN. APOE Christchurch enhances a disease-associated microglial response to plaque but suppresses response to tau pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597211. [PMID: 38895362 PMCID: PMC11185750 DOI: 10.1101/2024.06.03.597211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Apolipoprotein E ε4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). A recent case report identified a rare variant in APOE, APOE3-R136S (Christchurch), proposed to confer resistance to autosomal dominant Alzheimer's Disease (AD). However, it remains unclear whether and how this variant exerts its protective effects. Methods We introduced the R136S variant into mouse Apoe (ApoeCh) and investigated its effect on the development of AD-related pathology using the 5xFAD model of amyloidosis and the PS19 model of tauopathy. We used immunohistochemical and biochemical analysis along with single-cell spatial transcriptomics and proteomics to explore the impact of the ApoeCh variant on AD pathological development and the brain's response to plaques and tau. Results In 5xFAD mice, ApoeCh enhances a Disease-Associated Microglia (DAM) phenotype in microglia surrounding plaques, and reduces plaque load, dystrophic neurites, and plasma neurofilament light chain. By contrast, in PS19 mice, ApoeCh suppresses the microglial and astrocytic responses to tau-laden neurons and does not reduce tau accumulation or phosphorylation, but partially rescues tau-induced synaptic and myelin loss. We compared how microglia responses differ between the two mouse models to elucidate the distinct DAM signatures induced by ApoeCh. We identified upregulation of antigen presentation-related genes in the DAM response in a PS19 compared to a 5xFAD background, suggesting a differential response to amyloid versus tau pathology that is modulated by the presence of ApoeCh. Conclusions These findings highlight the ability of the ApoeCh variant to modulate microglial responses based on the type of pathology, enhancing DAM reactivity in amyloid models and dampening neuroinflammation to promote protection in tau models. This suggests that the Christchurch variant's protective effects likely involve multiple mechanisms, including changes in receptor binding and microglial programming.
Collapse
Affiliation(s)
- Kristine M. Tran
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Nellie Kwang
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Angela Gomez-Arboledas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Cassandra Mar
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Donna Chao
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Kai-Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Joshua A. Alcantara
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
| | - Andrea J. Tenner
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Frank M. LaFerla
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| |
Collapse
|
37
|
Brown RE. Measuring the replicability of our own research. J Neurosci Methods 2024; 406:110111. [PMID: 38521128 DOI: 10.1016/j.jneumeth.2024.110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
In the study of transgenic mouse models of neurodevelopmental and neurodegenerative disorders, we use batteries of tests to measure deficits in behaviour and from the results of these tests, we make inferences about the mental states of the mice that we interpret as deficits in "learning", "memory", "anxiety", "depression", etc. This paper discusses the problems of determining whether a particular transgenic mouse is a valid mouse model of disease X, the problem of background strains, and the question of whether our behavioural tests are measuring what we say they are. The problem of the reliability of results is then discussed: are they replicable between labs and can we replicate our results in our own lab? This involves the study of intra- and inter- experimenter reliability. The variables that influence replicability and the importance of conducting a complete behavioural phenotype: sensory, motor, cognitive and social emotional behaviour are discussed. Then the thorny question of failure to replicate is examined: Is it a curse or a blessing? Finally, the role of failure in research and what it tells us about our research paradigms is examined.
Collapse
Affiliation(s)
- Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
38
|
Yamada C, Ho A, Garcia C, Oblak AL, Bissel S, Porosencova T, Porosencov E, Uncuta D, Ngala B, Shepilov D, Skibo G, Mascarenhas AK, Akkaoui J, Lakshmana MK, Sankar U, Nichols F, Lamb BT, Groppa S, Movila A. Dementia exacerbates periodontal bone loss in females. J Periodontal Res 2024; 59:512-520. [PMID: 38243688 DOI: 10.1111/jre.13227] [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: 03/09/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease defined by the pathologic loss of the periodontal ligament and alveolar bone in relation to aging. Although clinical cohort studies reported that periodontitis is significantly elevated in males compared to females, emerging evidence indicates that females with dementia are at a greater risk for periodontitis and decreased alveolar bone. OBJECTIVE This study aimed to evaluate whether dementia is a potential sex-dependent risk factor for periodontal bone loss using an experimental model of periodontitis induced in the triple transgenic (3x-Tg) dementia-like mice and clinical samples collected from senior 65 plus age patients with diagnosed dementia. MATERIALS AND METHODS We induced periodontitis in dementia-like triple-transgenic (3x-Tg) male and female mice and age-matched wild-type (WT) control mice by ligature placement. Then, alveolar bone loss and osteoclast activity were evaluated using micro-CT and in situ imaging assays. In addition, we performed dental examinations on patients with diagnosed dementia. Finally, dementia-associated Aβ42 and p-Tau (T181) and osteoclastogenic receptor activator of nuclear factor kappa-Β ligand (RANKL) in gingival crevicular fluid (GCF) collected from mice and clinical samples were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS Alveolar bone loss and in situ osteoclast activity were significantly elevated in periodontal lesions of 3x-Tg females but not males, compared to wild-type control mice. In addition, we also observed that the probing pocket depth (PPD) was also significantly elevated in female patients with dementia. Using ELISA assay, we observed that females had elevated levels of osteoclastogenic RANKL and dementia-associated Aβ42 and p-Tau (T181) in the GCF collected from experimental periodontitis lesions and clinical samples. CONCLUSION Altogether, we demonstrate that females with dementia have an increased risk for periodontal bone loss compared to males.
Collapse
Affiliation(s)
- Chiaki Yamada
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Anny Ho
- Institute of Neuro-immune Medicine, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - Christopher Garcia
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Adrian L Oblak
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stephanie Bissel
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tatiana Porosencova
- Faculty of Dentistry, "Nicolae Testemițanu" State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Egor Porosencov
- Faculty of Dentistry, "Nicolae Testemițanu" State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Diana Uncuta
- Faculty of Dentistry, "Nicolae Testemițanu" State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Bidii Ngala
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dmytro Shepilov
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | - Galyna Skibo
- Department of Cytology, Bogomoletz Institute of Physiology, NAS of Ukraine, Kyiv, Ukraine
| | | | - Juliet Akkaoui
- Institute of Neuro-immune Medicine, Nova Southeastern University, Ft. Lauderdale, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Madepalli K Lakshmana
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Uma Sankar
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Frank Nichols
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stanislav Groppa
- Department of Neurology, Institute of Emergency Medicine, Chisinau, Moldova
- Department of Neurology, "Nicolae Testemițanu" State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, Indiana, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Institute of Neuro-immune Medicine, Nova Southeastern University, Ft. Lauderdale, Florida, USA
- Department of Oral Sciences and Translational Research, College of Dental Medicine, Nova Southeastern University, Davie, Florida, USA
| |
Collapse
|
39
|
Fraile-Ramos J, Reig-Vilallonga J, Giménez-Llort L. Glomerular Hypertrophy and Splenic Red Pulp Degeneration Concurrent with Oxidative Stress in 3xTg-AD Mice Model for Alzheimer's Disease and Its Exacerbation with Sex and Social Isolation. Int J Mol Sci 2024; 25:6112. [PMID: 38892297 PMCID: PMC11172848 DOI: 10.3390/ijms25116112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
The continuously expanding field of Alzheimer's disease (AD) research is now beginning to defocus the brain to take a more systemic approach to the disease, as alterations in the peripheral organs could be related to disease progression. One emerging hypothesis is organ involvement in the process of Aβ clearance. In the present work, we aimed to examine the status and involvement of the kidney as a key organ for waste elimination and the spleen, which is in charge of filtering the blood and producing lymphocytes, and their influence on AD. The results showed morphological and structural changes due to acute amyloidosis in the kidney (glomeruli area) and spleen (red pulp area and red/white pulp ratio) together with reduced antioxidant defense activity (GPx) in 16-month-old male and female 3xTg-AD mice when compared to their age- and sex-matched non-transgenic (NTg) counterparts. All these alterations correlated with the anxious-like behavioral phenotype of this mouse model. In addition, forced isolation, a cause of psychological stress, had a negative effect by intensifying genotype differences and causing differences to appear in NTg animals. This study further supports the relevance of a more integrative view of the complex interplay between systems in aging, especially at advanced stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Juan Fraile-Ramos
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Josep Reig-Vilallonga
- Department of Anatomy, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
40
|
O'Leary TP, Brown RE. Age-related changes in species-typical behaviours in the 5xFAD mouse model of Alzheimer's disease from 4 to 16 months of age. Behav Brain Res 2024; 465:114970. [PMID: 38531510 DOI: 10.1016/j.bbr.2024.114970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) patients show age-related decreases in the ability to perform activities of daily living and the decline in these activities is related to the severity of neurobiological deterioration underlying the disease. The 5xFAD mouse model of AD shows age-related impairments in sensory- motor and cognitive function, but little is known about changes in species-typical behaviours that may model activities of daily living in AD patients. Therefore, we examined species-typical behaviours used as indices of exploration (rearing) and compulsivity (grooming) across six tests of anxiety-like behaviour or motor function in female 5xFAD mice from 3 to 16 months of age. Robust decreases in rearing were found in 5xFAD mice across all tests after 9 months of age, although few differences were observed in grooming. A fine-scale analysis of grooming, however, revealed a previously unresolved and spatially restricted pattern of grooming in 5xFAD mice at 13-16 months of age. We then examined changes in species-typical behaviours in the home-cage, and show impaired nest building in 5xFAD mice at all ages tested. Lastly, we examined the relationship between reduced species typical behaviours in 5xFAD mice and the presentation of freezing behaviour, a commonly used measure of memory for conditioned fear. These results showed that along with cognitive and sensory-motor behaviour, 5xFAD mice have robust age-related impairments in species-typical behaviours. Therefore, species typical behaviours in 5xFAD mice may help to model the decline in activities of daily living observed in AD patients, and may provide useful behavioural phenotypes for evaluating the pre-clinical efficacy of novel therapeutics for AD.
Collapse
Affiliation(s)
- Timothy P O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
41
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
42
|
Kiss E, Kins S, Gorgas K, Venczel Szakács KH, Kirsch J, Kuhse J. Another Use for a Proven Drug: Experimental Evidence for the Potential of Artemisinin and Its Derivatives to Treat Alzheimer's Disease. Int J Mol Sci 2024; 25:4165. [PMID: 38673751 PMCID: PMC11049906 DOI: 10.3390/ijms25084165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Plant-derived multitarget compounds may represent a promising therapeutic strategy for multifactorial diseases, such as Alzheimer's disease (AD). Artemisinin and its derivatives were indicated to beneficially modulate various aspects of AD pathology in different AD animal models through the regulation of a wide range of different cellular processes, such as energy homeostasis, apoptosis, proliferation and inflammatory pathways. In this review, we aimed to provide an up-to-date overview of the experimental evidence documenting the neuroprotective activities of artemi-sinins to underscore the potential of these already-approved drugs for treating AD also in humans and propose their consideration for carefully designed clinical trials. In particular, the benefits to the main pathological hallmarks and events in the pathological cascade throughout AD development in different animal models of AD are summarized. Moreover, dose- and context-dependent effects of artemisinins are noted.
Collapse
Affiliation(s)
- Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, 69120 Kaiserslautern, Germany;
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Kinga Hajnal Venczel Szakács
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| |
Collapse
|
43
|
Datta D, Perone I, Wijegunawardana D, Liang F, Morozov YM, Arellano J, Duque A, Xie Z, van Dyck CH, Joyce MKP, Arnsten AFT. Nanoscale imaging of pT217-tau in aged rhesus macaque entorhinal and dorsolateral prefrontal cortex: Evidence of interneuronal trafficking and early-stage neurodegeneration. Alzheimers Dement 2024; 20:2843-2860. [PMID: 38445818 PMCID: PMC11032534 DOI: 10.1002/alz.13737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Tau phosphorylated at threonine-217 (pT217-tau) is a novel fluid-based biomarker that predicts onset of Alzheimer's disease (AD) symptoms, but little is known about how pT217-tau arises in the brain, as soluble pT217-tau is dephosphorylated post mortem in humans. METHODS We used multilabel immunofluorescence and immunoelectron microscopy to examine the subcellular localization of early-stage pT217-tau in entorhinal and prefrontal cortices of aged macaques with naturally occurring tau pathology and assayed pT217-tau levels in plasma. RESULTS pT217-tau was aggregated on microtubules within dendrites exhibiting early signs of degeneration, including autophagic vacuoles. It was also seen trafficking between excitatory neurons within synapses on spines, where it was exposed to the extracellular space, and thus accessible to cerebrospinal fluid (CSF)/blood. Plasma pT217-tau levels increased across the age span and thus can serve as a biomarker in macaques. DISCUSSION These data help to explain why pT217-tau predicts degeneration in AD and how it gains access to CSF and plasma to serve as a fluid biomarker.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Isabella Perone
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | | | - Feng Liang
- Department of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yury M. Morozov
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Jon Arellano
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Alvaro Duque
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Zhongcong Xie
- Department of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Mary Kate P. Joyce
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Amy F. T. Arnsten
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| |
Collapse
|
44
|
Garcia‐Agudo LF, Shi Z, Smith IF, Kramár EA, Tran K, Kawauchi S, Wang S, Collins S, Walker A, Shi K, Neumann J, Liang HY, Da Cunha C, Milinkeviciute G, Morabito S, Miyoshi E, Rezaie N, Gomez‐Arboledas A, Arvilla AM, Ghaemi DI, Tenner AJ, LaFerla FM, Wood MA, Mortazavi A, Swarup V, MacGregor GR, Green KN. BIN1 K358R suppresses glial response to plaques in mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:2922-2942. [PMID: 38460121 PMCID: PMC11032570 DOI: 10.1002/alz.13767] [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: 11/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 03/11/2024]
Abstract
INTRODUCTION The BIN1 coding variant rs138047593 (K358R) is linked to Late-Onset Alzheimer's Disease (LOAD) via targeted exome sequencing. METHODS To elucidate the functional consequences of this rare coding variant on brain amyloidosis and neuroinflammation, we generated BIN1K358R knock-in mice using CRISPR/Cas9 technology. These mice were subsequently bred with 5xFAD transgenic mice, which serve as a model for Alzheimer's pathology. RESULTS The presence of the BIN1K358R variant leads to increased cerebral amyloid deposition, with a dampened response of astrocytes and oligodendrocytes, but not microglia, at both the cellular and transcriptional levels. This correlates with decreased neurofilament light chain in both plasma and brain tissue. Synaptic densities are significantly increased in both wild-type and 5xFAD backgrounds homozygous for the BIN1K358R variant. DISCUSSION The BIN1 K358R variant modulates amyloid pathology in 5xFAD mice, attenuates the astrocytic and oligodendrocytic responses to amyloid plaques, decreases damage markers, and elevates synaptic densities. HIGHLIGHTS BIN1 rs138047593 (K358R) coding variant is associated with increased risk of LOAD. BIN1 K358R variant increases amyloid plaque load in 12-month-old 5xFAD mice. BIN1 K358R variant dampens astrocytic and oligodendrocytic response to plaques. BIN1 K358R variant decreases neuronal damage in 5xFAD mice. BIN1 K358R upregulates synaptic densities and modulates synaptic transmission.
Collapse
Affiliation(s)
| | - Zechuan Shi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Ian F. Smith
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Enikö A. Kramár
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Kai‐Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Heidi Yahan Liang
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Samuel Morabito
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Emily Miyoshi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Narges Rezaie
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Adrian Mendoza Arvilla
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Daryan Iman Ghaemi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology & BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Ali Mortazavi
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Vivek Swarup
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
45
|
Soni N, Hohsfield LA, Tran KM, Kawauchi S, Walker A, Javonillo D, Phan J, Matheos D, Da Cunha C, Uyar A, Milinkeviciute G, Gomez‐Arboledas A, Tran K, Kaczorowski CC, Wood MA, Tenner AJ, LaFerla FM, Carter GW, Mortazavi A, Swarup V, MacGregor GR, Green KN. Genetic diversity promotes resilience in a mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:2794-2816. [PMID: 38426371 PMCID: PMC11032575 DOI: 10.1002/alz.13753] [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: 11/17/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder with multifactorial etiology, including genetic factors that play a significant role in disease risk and resilience. However, the role of genetic diversity in preclinical AD studies has received limited attention. METHODS We crossed five Collaborative Cross strains with 5xFAD C57BL/6J female mice to generate F1 mice with and without the 5xFAD transgene. Amyloid plaque pathology, microglial and astrocytic responses, neurofilament light chain levels, and gene expression were assessed at various ages. RESULTS Genetic diversity significantly impacts AD-related pathology. Hybrid strains showed resistance to amyloid plaque formation and neuronal damage. Transcriptome diversity was maintained across ages and sexes, with observable strain-specific variations in AD-related phenotypes. Comparative gene expression analysis indicated correlations between mouse strains and human AD. DISCUSSION Increasing genetic diversity promotes resilience to AD-related pathogenesis, relative to an inbred C57BL/6J background, reinforcing the importance of genetic diversity in uncovering resilience in the development of AD. HIGHLIGHTS Genetic diversity's impact on AD in mice was explored. Diverse F1 mouse strains were used for AD study, via the Collaborative Cross. Strain-specific variations in AD pathology, glia, and transcription were found. Strains resilient to plaque formation and plasma neurofilament light chain (NfL) increases were identified. Correlations with human AD transcriptomics were observed.
Collapse
Affiliation(s)
- Neelakshi Soni
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kristine M. Tran
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dominic Javonillo
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dina Matheos
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Asli Uyar
- The Jackson LaboratoryBar HarborMaineUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | | | - Ali Mortazavi
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological SystemsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Vivek Swarup
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAROffice of ResearchUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cellular BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
46
|
Puris E, Saveleva L, Auriola S, Gynther M, Kanninen KM, Fricker G. Sex-specific changes in protein expression of membrane transporters in the brain cortex of 5xFAD mouse model of Alzheimer's disease. Front Pharmacol 2024; 15:1365051. [PMID: 38572427 PMCID: PMC10989684 DOI: 10.3389/fphar.2024.1365051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Membrane transporters playing an important role in the passage of drugs, metabolites and nutrients across the membranes of the brain cells have been shown to be involved in pathogenesis of Alzheimer's disease (AD). However, little is known about sex-specific changes in transporter protein expression at the brain in AD. Here, we investigated sex-specific alterations in protein expression of three ATP-binding cassette (ABC) and five solute carriers (SLC) transporters in the prefrontal cortex of a commonly used model of familial AD (FAD), 5xFAD mice. Sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic analysis was applied for absolute quantification of transporter protein expression. We compared the changes in transporter protein expressions in 7-month-old male and female 5xFAD mice versus sex-matched wild-type mice. The study revealed a significant sex-specific increase in protein expression of ABCC1 (p = 0.007) only in male 5xFAD mice as compared to sex-matched wild-type animals. In addition, the increased protein expression of glucose transporter 1 (p = 0.01), 4F2 cell-surface antigen heavy chain (p = 0.01) and long-chain fatty acid transport protein 1 (p = 0.02) were found only in female 5xFAD mice as compared to sex-matched wild-type animals. Finally, protein expression of alanine/serine/cysteine/threonine transporter 1 was upregulated in both male (p = 0.02) and female (p = 0.002) 5xFAD mice. The study provides important information about sex-specific changes in brain cortical transporter expression in 5xFAD mice, which will facilitate drug development of therapeutic strategies for AD targeting these transporters and drug delivery research.
Collapse
Affiliation(s)
- Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
47
|
Jagadeesan N, Roules GC, Chandrashekar DV, Yang J, Kolluru S, Sumbria RK. Modulation of hippocampal protein expression by a brain penetrant biologic TNF-α inhibitor in the 3xTg Alzheimer's disease mice. J Transl Med 2024; 22:291. [PMID: 38500108 PMCID: PMC10946165 DOI: 10.1186/s12967-024-05008-x] [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: 09/29/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Biologic TNF-α inhibitors (bTNFIs) can block cerebral TNF-α in Alzheimer's disease (AD) if these macromolecules can cross the blood-brain barrier (BBB). Thus, a model bTNFI, the extracellular domain of type II TNF-α receptor (TNFR), which can bind to and sequester TNF-α, was fused with a mouse transferrin receptor antibody (TfRMAb) to enable brain delivery via BBB TfR-mediated transcytosis. Previously, we found TfRMAb-TNFR to be protective in a mouse model of amyloidosis (APP/PS1) and tauopathy (PS19), and herein we investigated its effects in mice that combine both amyloidosis and tauopathy (3xTg-AD). METHODS Eight-month-old female 3xTg-AD mice were injected intraperitoneally with saline (n = 11) or TfRMAb-TNFR (3 mg/kg; n = 11) three days per week for 12 weeks. Age-matched wild-type (WT) mice (n = 9) were treated similarly with saline. Brains were processed for immunostaining and high-resolution multiplex NanoString GeoMx spatial proteomics. RESULTS We observed regional differences in proteins relevant to Aβ, tau, and neuroinflammation in the hippocampus of 3xTg-AD mice compared with WT mice. From 64 target proteins studied using spatial proteomics, a comparison of the Aβ-plaque bearing vs. plaque-free regions in the 3xTg-AD mice yielded 39 differentially expressed proteins (DEP) largely related to neuroinflammation (39% of DEP) and Aβ and tau pathology combined (31% of DEP). Hippocampal spatial proteomics revealed that the majority of the proteins modulated by TfRMAb-TNFR in the 3xTg-AD mice were relevant to microglial function (⁓ 33%). TfRMAb-TNFR significantly reduced mature Aβ plaques and increased Aβ-associated microglia around larger Aβ deposits in the 3xTg-AD mice. Further, TfRMAb-TNFR increased mature Aβ plaque-associated microglial TREM2 in 3xTg-AD mice. CONCLUSION Overall, despite the low visual Aβ load in the 11-month-old female 3xTg-AD mice, our results highlight region-specific AD-relevant DEP in the hippocampus of these mice. Chronic TfRMAb-TNFR dosing modulated several DEP involved in AD pathology and showed a largely microglia-centric mechanism of action in the 3xTg-AD mice.
Collapse
Affiliation(s)
- Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Sanjana Kolluru
- Rancho Cucamonga High School, 11801 Lark Dr, Rancho Cucamonga, CA, 91701, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
48
|
Duggan MR, Steinberg Z, Peterson T, Francois TJ, Parikh V. Cognitive trajectories in longitudinally trained 3xTg-AD mice. Physiol Behav 2024; 275:114435. [PMID: 38103626 PMCID: PMC10872326 DOI: 10.1016/j.physbeh.2023.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Preclinical studies in Alzheimer's disease (AD) often rely on cognitively naïve animal models in cross-sectional designs that can fail to reflect the cognitive exposures across the lifespan and heterogeneous neurobehavioral features observed in humans. To determine whether longitudinal cognitive training may affect cognitive capacities in a well-characterized AD mouse model, 3xTg and wild-type mice (n = 20) were exposed daily to a training variant of the Go-No-Go (GNG) operant task from 3 to 9 months old. At 3, 6, and 9 months, performance on a testing variant of the GNG task and anxiety-like behaviors were measured, while long-term recognition memory was also assessed at 9 months. In general, GNG training improved performance with increasing age across genotypes. At 3 months old, 3xTg mice showed slight deficits in inhibitory control that were accompanied by minor improvements in signal detection and decreased anxiety-like behavior, but these differences did not persist at 6 and 9 months old. At 9 months old, 3xTg mice displayed minor deficits in signal detection, and long-term recognition memory capacity was comparable with wild-type subjects. Our findings indicate that longitudinal cognitive training can render 3xTg mice with cognitive capacities that are on par with their wild-type counterparts, potentially reflecting functional compensation in subjects harboring AD genetic mutations.
Collapse
Affiliation(s)
- Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Zoe Steinberg
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Tara Peterson
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Tara-Jade Francois
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| |
Collapse
|
49
|
Martín-Rodríguez A, Gostian-Ropotin LA, Beltrán-Velasco AI, Belando-Pedreño N, Simón JA, López-Mora C, Navarro-Jiménez E, Tornero-Aguilera JF, Clemente-Suárez VJ. Sporting Mind: The Interplay of Physical Activity and Psychological Health. Sports (Basel) 2024; 12:37. [PMID: 38275986 PMCID: PMC10819297 DOI: 10.3390/sports12010037] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The symbiotic relationship between sports practice and psychological well-being has, in recent times, surged to the forefront of academic and public attention. The aim of this narrative review is to comprehensively explore the intricate pathways linking physical engagement in sports to its subsequent impacts on mental health and synthesize the multifarious effects of sports on psychological health, offering insights for integrating physical and psychological strategies to enhance well-being. From neurobiological underpinnings to therapeutic applications, this comprehensive manuscript provides an in-depth dive into the multifaceted world of sports and psychology. Highlighting evidence-based interventions, this review aspires to offer actionable insights for practitioners, athletes, and individuals alike, advocating for a holistic approach to mental well-being. This manuscript highlights the profound impact of sports on mental health, emphasizing its role in emotional regulation, resilience, cognitive function, and treating psychological conditions. It details how sports induce neurochemical changes, enhance brain functions like memory and learning, and aid against cognitive decline. This review also notes the benefits of regular exercise in mood improvement, stress management, and social skill enhancement, particularly when combined with mindfulness practices. It underscores the importance of considering cultural and gender perspectives in sports psychology, advocating for an integrated physical-psychological approach to promote overall well-being.
Collapse
Affiliation(s)
- Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Laura Augusta Gostian-Ropotin
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | | | - Noelia Belando-Pedreño
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Juan Antonio Simón
- Department Ciencias Sociales Act Fis Deporte & Ocio, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Clara López-Mora
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Valencia, Pg. de l’Albereda, 7, 46010 València, Spain;
| | | | - José Francisco Tornero-Aguilera
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
50
|
Lamichhane G, Liu J, Lee SJ, Lee DY, Zhang G, Kim Y. Curcumin Mitigates the High-Fat High-Sugar Diet-Induced Impairment of Spatial Memory, Hepatic Metabolism, and the Alteration of the Gut Microbiome in Alzheimer's Disease-Induced (3xTg-AD) Mice. Nutrients 2024; 16:240. [PMID: 38257133 PMCID: PMC10818691 DOI: 10.3390/nu16020240] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
The escalating prevalence of metabolic diseases and an aging demographic has been correlated with a concerning rise in Alzheimer's disease (AD) incidence. This study aimed to access the protective effects of curcumin, a bioactive flavonoid from turmeric, on spatial memory, metabolic functions, and the regulation of the gut microbiome in AD-induced (3xTg-AD) mice fed with either a normal chow diet (NCD) or a high-fat high-sugar diet (HFHSD). Our findings revealed an augmented susceptibility of the HFHSD-fed 3xTg-AD mice for weight gain and memory impairment, while curcumin supplementation demonstrated a protective effect against these changes. This was evidenced by significantly reduced body weight gain and improved behavioral and cognitive function in the curcumin-treated group. These improvements were substantiated by diminished fatty acid synthesis, altered cholesterol metabolism, and suppressed adipogenesis-related pathways in the liver, along with modified synaptic plasticity-related pathways in the brain. Moreover, curcumin enriched beneficial gut microbiota, including Oscillospiraceae and Rikenellaceae at the family level, and Oscillibacter, Alistipes, Pseudoflavonifractor, Duncaniella, and Flintibacter at the genus level. The observed alteration in these gut microbiota profiles suggests a potential crosswalk in the liver and brain for regulating metabolic and cognitive functions, particularly in the context of obesity-associated cognitive disfunction, notably AD.
Collapse
Affiliation(s)
- Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Jing Liu
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Su-Jeong Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| |
Collapse
|