1
|
Long Z, Ge C, Zhao Y, Liu Y, Zeng Q, Tang Q, Dong Z, He G. Enhanced autophagic clearance of amyloid-β via histone deacetylase 6-mediated V-ATPase assembly and lysosomal acidification protects against Alzheimer's disease in vitro and in vivo. Neural Regen Res 2025; 20:2633-2644. [PMID: 38993141 DOI: 10.4103/nrr.nrr-d-23-01633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/29/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00025/figure1/v/2024-11-05T132919Z/r/image-tiff Recent studies have suggested that abnormal acidification of lysosomes induces autophagic accumulation of amyloid-β in neurons, which is a key step in senile plaque formation. Therefore, restoring normal lysosomal function and rebalancing lysosomal acidification in neurons in the brain may be a new treatment strategy for Alzheimer's disease. Microtubule acetylation/deacetylation plays a central role in lysosomal acidification. Here, we show that inhibiting the classic microtubule deacetylase histone deacetylase 6 with an histone deacetylase 6 shRNA or thehistone deacetylase 6 inhibitor valproic acid promoted lysosomal reacidification by modulating V-ATPase assembly in Alzheimer's disease. Furthermore, we found that treatment with valproic acid markedly enhanced autophagy, promoted clearance of amyloid-β aggregates, and ameliorated cognitive deficits in a mouse model of Alzheimer's disease. Our findings demonstrate a previously unknown neuroprotective mechanism in Alzheimer's disease, in which histone deacetylase 6 inhibition by valproic acid increases V-ATPase assembly and lysosomal acidification.
Collapse
Affiliation(s)
- Zhimin Long
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Chuanhua Ge
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yuanjie Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qinghua Zeng
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qing Tang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Forny Germano L, Koehler JA, Baggio LL, Cui F, Wong CK, Rittig N, Cao X, Matthews D, Drucker DJ. The GLP-1 medicines semaglutide and tirzepatide do not alter disease-related pathology, behaviour or cognitive function in 5XFAD and APP/PS1 mice. Mol Metab 2024; 89:102019. [PMID: 39216535 PMCID: PMC11408156 DOI: 10.1016/j.molmet.2024.102019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/17/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE The development of glucagon-like peptide-1 receptor (GLP-1R) agonists for the treatment of type 2 diabetes and obesity has been accompanied by evidence for anti-inflammatory and cytoprotective actions in the heart, blood vessels, kidney, and brain. Whether GLP-1R agonists might be useful clinically for attenuating deterioration of cognitive dysfunction and reducing the progression of Alzheimer's disease remains uncertain. METHODS Here we evaluated the actions of semaglutide and tirzepatide, clinically distinct GLP-1 medicines, in two mouse models of neurodegeneration. RESULTS Semaglutide reduced body weight and improved glucose tolerance in 12-month-old male and female 5XFAD and APP/PS1 mice, consistent with pharmacological engagement of the GLP-1R. Nevertheless, amyloid plaque density was not different in the cerebral cortex, hippocampus, or subiculum of semaglutide-treated 12-month-old 5XFAD and APP/PS1 mice. IBA1 and GFAP expression were increased in the hippocampus of 5XFAD and APP/PS1 mice but were not reduced by semaglutide. Moreover, parameters of neurobehavioral and cognitive function evaluated using Open Field testing or the Morris water maze were not improved following treatment with semaglutide. To explore whether incretin therapies might be more effective in younger mice, we studied semaglutide and tirzepatide action in 6-month-old male and female 5XFAD mice. Neither semaglutide nor tirzepatide modified the extent of plaque accumulation, hippocampal IBA1+ or GFAP+ cells, or parameters of neurobehavioral testing, despite improving glucose tolerance and reducing body weight. mRNA biomarkers of inflammation and neurodegeneration were increased in the hippocampus of male and female 5XFAD mice but were not reduced after treatment with semaglutide or tirzepatide. CONCLUSIONS Collectively, these findings reveal preservation of the metabolic actions of two GLP-1 medicines, semaglutide and tirzepatide, yet inability to detect improvement in structural and functional parameters of neurodegeneration in two mouse models of Alzheimer's disease.
Collapse
Affiliation(s)
| | - Jacqueline A Koehler
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Laurie L Baggio
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Fiona Cui
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Chi Kin Wong
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Nikolaj Rittig
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Xiemin Cao
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Dianne Matthews
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, M5G1X5, Canada.
| |
Collapse
|
3
|
Bulgart HR, Lopez Perez MA, Tucker A, Giarrano GN, Banford K, Miller O, Bonser SW, Wold LE, Scharre D, Weisleder N. Plasma membrane repair defect in Alzheimer's disease neurons is driven by the reduced dysferlin expression. FASEB J 2024; 38:e70099. [PMID: 39400395 PMCID: PMC11486262 DOI: 10.1096/fj.202401731rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and a defect in neuronal plasma membrane repair could exacerbate neurotoxicity, neuronal death, and disease progression. In this study, application of AD patient cerebrospinal fluid (CSF) and recombinant human Aβ to otherwise healthy neurons induces defective neuronal plasma membrane repair in vitro and ex vivo. We identified Aβ as the biochemical component in patient CSF leading to compromised repair capacity and depleting Aβ rescued repair capacity. These elevated Aβ levels reduced expression of dysferlin, a protein that facilitates membrane repair, by altering autophagy and reducing dysferlin trafficking to sites of membrane injury. Overexpression of dysferlin and autophagy inhibition rescued membrane repair. Overall, these findings indicate an AD pathogenic mechanism where Aβ impairs neuronal membrane repair capacity and increases susceptibility to cell death. This suggests that membrane repair could be therapeutically targeted in AD to restore membrane integrity and reduce neurotoxicity and neuronal death.
Collapse
Affiliation(s)
- Hannah R Bulgart
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Miguel A Lopez Perez
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Alexis Tucker
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Gianni N Giarrano
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Kassidy Banford
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Olivia Miller
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Sidney W.G. Bonser
- Department of Applied Statistics and Research Methods, University of Northern Colorado, Greeley, CO USA
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Douglas Scharre
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Noah Weisleder
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY USA
| |
Collapse
|
4
|
Yarbro JM, Han X, Dasgupta A, Yang K, Liu D, Shrestha HK, Zaman M, Wang Z, Yu K, Lee DG, Vanderwall D, Niu M, Sun H, Xie B, Chen PC, Jiao Y, Zhang X, Wu Z, Fu Y, Li Y, Yuan ZF, Wang X, Poudel S, Vagnerova B, He Q, Tang A, Ronaldson PT, Chang R, Yu G, Liu Y, Peng J. Human-mouse proteomics reveals the shared pathways in Alzheimer's disease and delayed protein turnover in the amyloidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620263. [PMID: 39484428 PMCID: PMC11527136 DOI: 10.1101/2024.10.25.620263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Murine models of Alzheimer's disease (AD) are crucial for elucidating disease mechanisms but have limitations in fully representing AD molecular complexities. We comprehensively profiled age-dependent brain proteome and phosphoproteome ( n > 10,000 for both) across multiple mouse models of amyloidosis. We identified shared pathways by integrating with human metadata, and prioritized novel components by multi-omics analysis. Collectively, two commonly used models (5xFAD and APP-KI) replicate 30% of the human protein alterations; additional genetic incorporation of tau and splicing pathologies increases this similarity to 42%. We dissected the proteome-transcriptome inconsistency in AD and 5xFAD mouse brains, revealing that inconsistent proteins are enriched within amyloid plaque microenvironment (amyloidome). Determining the 5xFAD proteome turnover demonstrates that amyloid formation delays the degradation of amyloidome components, including Aβ-binding proteins and autophagy/lysosomal proteins. Our proteomic strategy defines shared AD pathways, identify potential new targets, and underscores that protein turnover contributes to proteome-transcriptome discrepancies during AD progression.
Collapse
|
5
|
Zhang R, Ohshima M, Brodin D, Wang Y, Morancé A, Schultzberg M, Chen G, Johansson J. Intravenous chaperone treatment of late-stage Alzheimer´s disease (AD) mouse model affects amyloid plaque load, reactive gliosis and AD-related genes. Transl Psychiatry 2024; 14:453. [PMID: 39448576 PMCID: PMC11502864 DOI: 10.1038/s41398-024-03161-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Treatment strategies that are efficient against established Alzheimer's disease (AD) are needed. BRICHOS is a molecular chaperone domain that prevents amyloid fibril formation and associated cellular toxicity. In this study, we treated an AD mouse model seven months after pathology onset, using intravenous administration of recombinant human (rh) Bri2 BRICHOS R221E. Two injections of rh Bri2 BRICHOS R221E per week for three months in AD mice reduced amyloid β (Aβ) burden, and mitigated astro- and microgliosis, as determined by glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (Iba1) immunohistochemistry. Sequencing of RNA from cortical microglia cells showed that BRICHOS treatment normalized the expression of identified plaque-induced genes in mice and humans, including clusterin and GFAP. Rh Bri2 BRICHOS R221E passed the blood-brain barrier (BBB) in age-matched wild-type mice as efficiently as in the AD mice, but then had no effect on measures of AD-like pathology, and mainly affected the expression of genes that affect cellular shape and movement. These results indicate a potential of rh Bri2 BRICHOS against advanced AD and underscore the ability of BRICHOS to target amyloid-induced pathology.
Collapse
Affiliation(s)
- Ruixin Zhang
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Makiko Ohshima
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden
| | - David Brodin
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Yu Wang
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Antonin Morancé
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden
- Department of Neuroscience, University of Mons (UMONS), Mons, Belgium
| | - Marianne Schultzberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden.
| | - Gefei Chen
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden.
| | - Jan Johansson
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
6
|
Zhang L, Lin J, Xiang K, Shi T, Guo B. Omnidirectional improvement of mitochondrial health in Alzheimer's disease by multi-targeting engineered activated neutrophil exosomes. J Control Release 2024; 376:470-487. [PMID: 39433157 DOI: 10.1016/j.jconrel.2024.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Alzheimer's disease (AD) is one kind of devasting neurodegenerative disorders affecting over 50 million people worldwide. Multi-targeted therapy has emerged as a new treatment for diagnosing and alleviating the pathogenesis process of AD; however, the current strategy is limited by its unsatisfactory efficiency. In our study, engineered activated neutrophil-derived exosomes (MP@Cur-MExo) were developed to improve the mitochondrial function in neurons by targeting and alleviating Aβ-induced neurotoxicity. MP@Cur-MExo are exosomes derived from IL-8-stimulated neutrophils decorated with mitochondria targeting ligand and Aβ targeted ligand modified SPION. Engineered exosomes can be cleaved by matrix metallopeptidase-2, which is overexpressed in the AD brain. Consequently, the released SPION and Curcumin-loaded engineered exosomes collaboratively protected neuron cells against Aβ-induced mitochondrial deficiency. In addition, MP@Cur-MExo effectively accumulated in the inflamed region of AD brain at an early stage, allowing early diagnosis of AD through bimodal (MRI/IVIS) imaging. Importantly, in a mouse model at an early stage of AD, intravenously injected MP@Cur-MExo restored mitochondrial function and reduced Aβ-induced mitochondrial damage, thereby attenuating AD progression. In conclusion, our designed engineered exosomes demonstrated that omnidirectional improvement of mitochondrial function can serve as a novel and practical approach for the diagnosis and treatment of neurodegenerative diseases. This study also reveals a promising therapeutic agent for impeding AD progression for future clinical applications.
Collapse
Affiliation(s)
- Lei Zhang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Jiaquan Lin
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Kai Xiang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Tianshu Shi
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| | - Baosheng Guo
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
7
|
Lu W, Shue F, Kurti A, Jeevaratnam S, Macyczko JR, Roy B, Izhar T, Wang N, Bu G, Kanekiyo T, Li Y. Amyloid pathology and cognitive impairment in hAβ-KI and APP SAA-KI mouse models of Alzheimer's disease. Neurobiol Aging 2024; 145:13-23. [PMID: 39447490 DOI: 10.1016/j.neurobiolaging.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
The hAβ-KI and APPSAA-KI are two amyloid models that harbor mutations in the endogenous mouse App gene. Both hAβ-KI and APPSAA-KI mice contain a humanized Aβ sequence, and APPSAA-KI mice carry three additional familial AD mutations. We herein report that the Aβ levels and Aβ42/Aβ40 ratio in APPSAA-KI homozygotes are dramatically higher than those in hAβ-KI homozygotes at 14 months of age. In addition, APPSAA-KI mice display a widespread distribution of amyloid plaques in the brain, whereas the plaques are undetectable in hAβ-KI mice. Moreover, there are no sex differences in amyloid pathology in APPSAA-KI mice. Both APPSAA-KI and hAβ-KI mice exhibit cognitive impairments, wherein no significant differences are found between these two models, although APPSAA KI mice show a trend towards worse cognitive function. Notably, female hAβ-KI and APPSAA-KI mice have a more pronounced cognitive impairments compared to their respective males. Our findings suggest that Aβ humanization contributes to cognitive deficits in APPSAA-KI mice, and that amyloid deposition might not be closely associated with cognitive impairments in APPSAA-KI mice.
Collapse
Affiliation(s)
- Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Suren Jeevaratnam
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jesse R Macyczko
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bhaskar Roy
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Taha Izhar
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ni Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
8
|
Miller SR, Luxem K, Lauderdale K, Nambiar P, Honma PS, Ly KK, Bangera S, Bullock M, Shin J, Kaliss N, Qiu Y, Cai C, Shen K, Mallen KD, Yan Z, Mendiola AS, Saito T, Saido TC, Pico AR, Thomas R, Roberson ED, Akassoglou K, Bauer P, Remy S, Palop JJ. Machine learning reveals prominent spontaneous behavioral changes and treatment efficacy in humanized and transgenic Alzheimer's disease models. Cell Rep 2024; 43:114870. [PMID: 39427315 DOI: 10.1016/j.celrep.2024.114870] [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: 10/09/2023] [Revised: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Computer-vision and machine-learning (ML) approaches are being developed to provide scalable, unbiased, and sensitive methods to assess mouse behavior. Here, we used the ML-based variational animal motion embedding (VAME) segmentation platform to assess spontaneous behavior in humanized App knockin and transgenic APP models of Alzheimer's disease (AD) and to test the role of AD-related neuroinflammation in these behavioral manifestations. We found marked alterations in spontaneous behavior in AppNL-G-F and 5xFAD mice, including age-dependent changes in motif utilization, disorganized behavioral sequences, increased transitions, and randomness. Notably, blocking fibrinogen-microglia interactions in 5xFAD-Fggγ390-396A mice largely prevented spontaneous behavioral alterations, indicating a key role for neuroinflammation. Thus, AD-related spontaneous behavioral alterations are prominent in knockin and transgenic models and sensitive to therapeutic interventions. VAME outcomes had higher specificity and sensitivity than conventional behavioral outcomes. We conclude that spontaneous behavior effectively captures age- and sex-dependent disease manifestations and treatment efficacy in AD models.
Collapse
Affiliation(s)
- Stephanie R Miller
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Kevin Luxem
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Kelli Lauderdale
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pranav Nambiar
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Patrick S Honma
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Katie K Ly
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shreya Bangera
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mary Bullock
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jia Shin
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nick Kaliss
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Yuechen Qiu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Catherine Cai
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Kevin Shen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - K Dakota Mallen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Zhaoqi Yan
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Andrew S Mendiola
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi 351-0198, Japan
| | - Alexander R Pico
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA 94158, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA 94158, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Pavol Bauer
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Stefan Remy
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany; German Center for Mental Health (DZPG), 39118 Magdeburg, Germany
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
9
|
Roberts H, Fang Y, Quinn K, Hill T, Peck MR, Bartke A, Hascup KN, Hascup ER. Lifespan of male and female APP/PS1 and APP NL-F/NL-F mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618508. [PMID: 39464050 PMCID: PMC11507819 DOI: 10.1101/2024.10.15.618508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) disproportionately affects women, yet most preclinical research studies are male-centric. We performed lifespan analyses of male and female AD mouse models (APP/PS1 and APPNL-F/NL-F) and their shared genetic background control (C57BL/6). Survival curves support significant sex differences between within genotypes. Minimal longevity revealed increased age in male APP/PS1, and decreased age in APPNL-F/NL-F mice. Maximal longevity revealed an increased average age in males. Furthermore, median lifespan differed between sex and genotype. This study supports sexual dimorphic survival in two mouse models of AD, emphasizing the need to examine mechanisms and treatments in both sexes.
Collapse
Affiliation(s)
- Hannah Roberts
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Yimin Fang
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Tiarra Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Dept of Internal Medicine; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
10
|
Gu X, Fan M, Zhou Y, Zhang Y, Wang L, Gao W, Li T, Wang H, Si N, Wei X, Bian B, Zhao H. Intestinal endogenous metabolites affect neuroinflammation in 5×FAD mice by mediating "gut-brain" axis and the intervention with Chinese Medicine. Alzheimers Res Ther 2024; 16:222. [PMID: 39396997 PMCID: PMC11472645 DOI: 10.1186/s13195-024-01587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 09/29/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Emerging evidence suggested the association between gut dysbiosis and Alzheimer's disease (AD) progression. However, it remained unclear how the gut microbiome and neuroinflammation in the brain mutually interact or how these interactions affect brain functioning and cognition. Here we hypothesized that "gut-brain" axis mediated by microbial derived metabolites was expected to novel breakthroughs in the fields of AD research and development. METHODS Multiple technologies, such as immunofluorescence, 16s rDNA sequencing, mass spectrometry-based metabolomics (LC-QQQ-MS and GC-MS), were used to reveal potential link between gut microbiota and the metabolism and cognition of the host. RESULTS Microbial depletion induced by the antibiotics mix (ABX) verified that "gut-brain" can transmit information bidirectionally. Short-chain fatty acid-producing (SCFAs-producing) bacteria and amino acid-producing bacteria fluctuated greatly in 5×FAD mice, especially the reduction sharply of the Bifidobacteriaceae and the increase of the Lachnospiraceae family. Concentrations of several Tryptophan-kynurenine intermediates, lactic acid, CD4+ cell, and CD8+ cells were higher in serum of 5×FAD mice, whilst TCA cycle intermediates and Th1/Th2 were lower. In addition, the levels of iso-butyric acid (IBA) in feces, serum, and brain of 5×FAD mice were increased compared with WT-M mice, especially in serum. And IBA in the brain was positively correlated with Aβ and proinflammatory factors. CONCLUSION Together, our finding highlighted that the alternation in gut microbiota affected the effective communication between the "gut-brain" axis in 5×FAD mice by regulating the immune system, carbohydrate, and energy metabolism.
Collapse
Affiliation(s)
- Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- The Neurology Department, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Miaoxuan Fan
- Beijing Drug Package Test Institute, Beijing, 100700, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Linna Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
11
|
Wood JI, Dulewicz M, Ge J, Stringer K, Szadziewska A, Desai S, Koutarapu S, Hajar HB, Blennow K, Zetterberg H, Cummings DM, Savas JN, Edwards FA, Hanrieder J. Isotope Encoded chemical Imaging Identifies Amyloid Plaque Age Dependent Structural Maturation, Synaptic Loss, and Increased Toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617019. [PMID: 39416086 PMCID: PMC11482761 DOI: 10.1101/2024.10.08.617019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
It is of critical importance to our understanding of Alzheimer's disease (AD) pathology to determine how key pathological factors are interconnected and implicated in nerve cell death, clinical symptoms, and disease progression. The formation of extracellular beta-amyloid (Aβ) plaques is the major pathological hallmark of AD and Aβ has been suggested to be a critical inducer of AD, driving disease pathogenesis. Exactly how Aβ plaque formation begins and how ongoing plaque deposition proceeds and initiates subsequent neurotoxic mechanisms is not well understood. The primary aim of our research is to elucidate the biochemical processes underlying early Aβ plaque formation in brain tissue. We recently introduced a chemical imaging paradigm based on mass spectrometry imaging (MSI) and metabolic isotope labelling to follow stable isotope labelling kinetics (iSILK) in vivo to track the in vivo build-up and deposition of Aβ. Herein, knock-in Aβ mouse models (App NL-F ) that develop Aβ pathology gradually are metabolically labeled with stable isotopes. This chemical imaging approach timestamps amyloid plaques during the period of initial deposition allowing the fate of aggregating Aβ species from before and during the earliest events of plaque pathology through plaque maturation to be tracked. To identify the molecular and cellular response to plaque maturation, we integrated iSILK with single plaque transcriptomics performed on adjacent tissue sections. This enabled changes in gene expression to be tracked as a function of plaque age (as encoded in the Aβ peptide isotopologue pattern) distinct from changes due to the chronological age or pathological severity. This approach identified that plaque age correlates negatively with gene expression patterns associated with synaptic function as early as in 10-month-old animals but persists into 18 months. Finally, we integrated hyperspectral confocal microscopy into our multiomic approach to image amyloid structural isomers, revealing a positive correlation between plaque age and amyloid structural maturity. This analysis identified three categories of plaques, each with a distinct impact on the surrounding microenvironment. Here, we identified that older, more compact plaques were associated with the most significant synapse loss and toxicity. These data show how isotope-encoded MS imaging can be used to delineate Aβ toxicity dynamics in vivo. Moreover, we show for the first time a functional integration of dynamic MSI, structural plaque imaging and whole genome-wide spatial transcriptomics at the single plaque level. This multiomic approach offers an unprecedented combination of temporal and spatial resolution enabling a description of the earliest events of precipitating amyloid pathology and how Aβ modulates synaptotoxic mechanisms.
Collapse
Affiliation(s)
- Jack I. Wood
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Junyue Ge
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Katie Stringer
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Alicja Szadziewska
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Sneha Desai
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Srinivas Koutarapu
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Haady B. Hajar
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queens Square, WC1N 3BG London, United Kingdom
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Damian M. Cummings
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Jeffrey N. Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Frances A. Edwards
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queens Square, WC1N 3BG London, United Kingdom
- SciLifeLab, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
12
|
Sekiya M, Sakakibara Y, Hirota Y, Ito N, Chikamatsu S, Takei K, Nishijima R, Iijima KM. Decreased plasma nicotinamide and altered NAD + metabolism in glial cells surrounding Aβ plaques in a mouse model of Alzheimer's disease. Neurobiol Dis 2024; 202:106694. [PMID: 39374707 DOI: 10.1016/j.nbd.2024.106694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and a leading cause of senile dementia. Amyloid-β (Aβ) accumulation triggers chronic neuroinflammation, initiating AD pathogenesis. Recent clinical trials for anti-Aβ immunotherapy underscore that blood-based biomarkers have significant advantages and applicability over conventional diagnostics and are an unmet clinical need. To further advance ongoing clinical trials and identify novel therapeutic targets for AD, developing additional plasma biomarkers closely associated with pathogenic mechanisms downstream of Aβ accumulation is critically important. To identify plasma metabolites reflective of neuroinflammation caused by Aβ pathology, we performed untargeted metabolomic analyses of the plasma by capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS) and analyzed the potential roles of the identified metabolic changes in the brain neuroinflammatory response using the female App knock-in (AppNLGF) mouse model of Aβ amyloidosis. The CE-TOFMS analysis of plasma samples from female wild-type (WT) and AppNLGF mice revealed that plasma levels of nicotinamide, a nicotinamide adenine dinucleotide (NAD+) precursor, were decreased in AppNLGF mice, and altered metabolite profiles were enriched for nicotinate/nicotinamide metabolism. In AppNLGF mouse brains, NAD+ levels were unaltered, but mRNA levels of NAD+-synthesizing nicotinate phosphoribosyltransferase (Naprt) and NAD+-degrading Cd38 genes were increased. These enzymes were induced in reactive astrocytes and microglia surrounding Aβ plaques in the cortex and hippocampus of female AppNLGF mouse brains, suggesting neuroinflammation increases NAD+ metabolism. This study suggests plasma nicotinamide could be indicative of the neuroinflammatory response and that nicotinate and nicotinamide metabolism are potential therapeutic targets for AD, by targeting both neuroinflammation and neuroprotection.
Collapse
Affiliation(s)
- Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Reseach Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Naoki Ito
- Brain-Skeletal Muscle Connection in Aging Project Team, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
13
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
14
|
Bala K, Porel P, Aran KR. Emerging roles of cannabinoid receptor CB2 receptor in the central nervous system: therapeutic target for CNS disorders. Psychopharmacology (Berl) 2024; 241:1939-1954. [PMID: 39264450 DOI: 10.1007/s00213-024-06683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024]
Abstract
RATIONALE The endocannabinoid system (ECS) belongs to the G protein-coupled receptor family of cell membranes and is associated with neuropsychiatric conditions, and neurodegenerative diseases. Cannabinoid 2 receptors (CB2) are expressed in the central nervous system (CNS) on microglia and subgroups of neurons and are involved in various behavioural processes via immunological and neural regulation. OBJECTIVE The objective of this paper is to summarize and explore the impact of CB2 receptors on neuronal modulation, their involvement in various neurological disorders, and their influence on mood, behavior, and cognitive function. RESULTS The activation of CB2 appears to protect the brain and its functions from damage under neuroinflammatory actions, making it an attractive target in a variety of neurological conditions such as Parkinson's disease (PD), multiple sclerosis (MS), Alzheimer's disease (AD), and Huntington's disease (HD). During inflammation, there is an overexpression of CB2 receptors, and CB2 agonists show a strong anti-inflammatory effect. These results have sparked interest in the CB2 receptors as a potential target for neurodegenerative and neuroinflammatory disease treatment. CONCLUSION In conclusion, CB2 receptors signalling shows promise for developing targeted interventions that could positively affect both immune and neuronal functions, ultimately influencing behavioral outcomes in both health and disease.
Collapse
Affiliation(s)
- Kanchan Bala
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Pratyush Porel
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
15
|
Doucette L, Turnbill V, Carlin K, Cavanagh A, Sollinger B, Kuter N, Flock DL, Robinson S, Chavez-Valdez R, Jantzie L, Martin LJ, Northington FJ. Neocortical cholinergic pathology after neonatal brain injury is increased by Alzheimer's disease-related genes in mice. Neurobiol Dis 2024; 200:106629. [PMID: 39111704 DOI: 10.1016/j.nbd.2024.106629] [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: 03/25/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/18/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) in neonates causes mortality and neurologic morbidity, including poor cognition with a complex neuropathology. Injury to the cholinergic basal forebrain and its rich innervation of cerebral cortex may also drive cognitive pathology. It is uncertain whether genes associated with adult cognition-related neurodegeneration worsen outcomes after neonatal HIE. We hypothesized that neocortical damage caused by neonatal HI in mice is ushered by persistent cholinergic innervation and interneuron (IN) pathology that correlates with cognitive outcome and is exacerbated by genes linked to Alzheimer's disease. We subjected non-transgenic (nTg) C57Bl6 mice and mice transgenically (Tg) expressing human mutant amyloid precursor protein (APP-Swedish variant) and mutant presenilin (PS1-ΔE9) to the Rice-Vannucci HI model on postnatal day 10 (P10). nTg and Tg mice with sham procedure were controls. Visual discrimination (VD) was tested for cognition. Cortical and hippocampal cholinergic axonal and IN pathology and Aβ plaques, identified by immunohistochemistry for choline acetyltransferase (ChAT) and 6E10 antibody respectively, were counted at P210. Simple ChAT+ axonal swellings were present in all sham and HI groups; Tg mice had more than their nTg counterparts, but HI did not affect the number of axonal swellings in APP/PS1 Tg mice. In contrast, complex ChAT+ neuritic clusters (NC) occurred only in Tg mice; HI increased that burden. The abundance of ChAT+ clusters in specific regions correlated with decreased VD. The frequency of attritional ChAT+ INs in the entorhinal cortex (EC) was increased in Tg shams relative to their nTg counterparts, but HI obviated this difference. Cholinergic IN pathology in EC correlated with NC number. The Aβ deposition in APP/PS1 Tg mice was not exacerbated by HI, nor did it correlate with other metrics. Adult APP/PS1 Tg mice have significant cortical cholinergic axon and EC ChAT+ IN pathologies; some pathology was exacerbated by neonatal HI and correlated with VD. Mechanisms of neonatal HI induced cognitive deficits and cortical neuropathology may be modulated by genetic risk, perhaps accounting for some of the variability in outcomes.
Collapse
Affiliation(s)
- Leslie Doucette
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Victoria Turnbill
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Katherine Carlin
- US Air Force Medical Corps, US Naval Hospital Okinawa, Okinawa, Japan
| | - Andrew Cavanagh
- Department of Neuroscience, Undergraduate Education, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Benjamin Sollinger
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Nazli Kuter
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Debra L Flock
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Shenandoah Robinson
- Phelps Center for Cerebral Palsy and Neurodevelopmental Medicine, Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD 21287, USA; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Lauren Jantzie
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; Phelps Center for Cerebral Palsy and Neurodevelopmental Medicine, Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD 21287, USA; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Lee J Martin
- Department of Neuroscience, Pathology, and Anesthesiology & Critical Care Medicine, and the Pathobiology Graduate Training Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Frances J Northington
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
16
|
El-Ghazawi K, Eyo UB, Peirce SM. Brain Microvascular Pericyte Pathology Linking Alzheimer's Disease to Diabetes. Microcirculation 2024; 31:e12877. [PMID: 39222475 PMCID: PMC11471384 DOI: 10.1111/micc.12877] [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: 03/13/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 09/04/2024]
Abstract
The brain microvasculature, which delivers oxygen and nutrients and forms a critical barrier protecting the central nervous system via capillaries, is deleteriously affected by both Alzheimer's disease (AD) and type 2 diabetes (T2D). T2D patients have an increased risk of developing AD, suggesting potentially related microvascular pathological mechanisms. Pericytes are an ideal cell type to study for functional links between AD and T2D. These specialized capillary-enwrapping cells regulate capillary density, lumen diameter, and blood flow. Pericytes also maintain endothelial tight junctions to ensure blood-brain barrier integrity, modulation of immune cell extravasation, and clearance of toxins. Changes in these phenomena have been observed in both AD and T2D, implicating "pericyte pathology" as a common feature of AD and T2D. This review examines the mechanisms of AD and T2D from the perspective of the brain microvasculature, highlighting how pericyte pathology contributes to both diseases. Our review identifies voids in understanding how AD and T2D negatively impact the brain microvasculature and suggests future studies to examine the intersections of these diseases.
Collapse
Affiliation(s)
- Kareem El-Ghazawi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Department of Neuroscience, University of Virginia Center for Brain Immunology and Glia School of Medicine, Charlottesville, VA, USA
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
17
|
Granzotto A, Vissel B, Sensi SL. Lost in translation: Inconvenient truths on the utility of mouse models in Alzheimer's disease research. eLife 2024; 13:e90633. [PMID: 39329365 PMCID: PMC11434637 DOI: 10.7554/elife.90633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The recent, controversial approval of antibody-based treatments for Alzheimer's disease (AD) is fueling a heated debate on the molecular determinants of this condition. The discussion should also incorporate a critical revision of the limitations of preclinical mouse models in advancing our understanding of AD. We critically discuss the limitations of animal models, stressing the need for careful consideration of how experiments are designed and results interpreted. We identify the shortcomings of AD models to recapitulate the complexity of the human disease. We dissect these issues at the quantitative, qualitative, temporal, and context-dependent levels. We argue that these models are based on the oversimplistic assumptions proposed by the amyloid cascade hypothesis (ACH) of AD and fail to account for the multifactorial nature of the condition. By shedding light on the constraints of current experimental tools, this review aims to foster the development and implementation of more clinically relevant tools. While we do not rule out a role for preclinical models, we call for alternative approaches to be explored and, most importantly, for a re-evaluation of the ACH.
Collapse
Affiliation(s)
- Alberto Granzotto
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
| | - Bryce Vissel
- St Vincent’s Hospital Centre for Applied Medical Research, St Vincent’s HospitalDarlinghurstAustralia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyAustralia
| | - Stefano L Sensi
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute for Advanced Biomedical Technologies – ITAB, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute of Neurology, SS Annunziata University Hospital, University G. d’Annunzio of Chieti-PescaraChietiItaly
| |
Collapse
|
18
|
Chandrashekar DV, Roules GC, Jagadeesan N, Panchal UR, Oyegbesan A, Imiruaye OE, Zhang H, Garcia J, Kaur K, Win S, Than TA, Kaplowitz N, Roosan MR, Han D, Sumbria RK. Hepatic LRP-1 plays an important role in amyloidosis in Alzheimer's disease mice: Potential role in chronic heavy alcohol feeding. Neurobiol Dis 2024; 199:106570. [PMID: 38885850 DOI: 10.1016/j.nbd.2024.106570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Hepatic lipoprotein receptor-related protein 1 (LRP-1) plays a central role in peripheral amyloid beta (Aβ) clearance, but its importance in Alzheimer's disease (AD) pathology is understudied. Our previous work showed that intragastric alcohol feeding to C57BL/6 J mice reduced hepatic LRP-1 expression which correlated with significant AD-relevant brain changes. Herein, we examined the role of hepatic LRP-1 in AD pathogenesis in APP/PS1 AD mice using two approaches to modulate hepatic LRP-1, intragastric alcohol feeding to model chronic heavy drinking shown by us to reduce hepatic LRP-1, and hepato-specific LRP-1 silencing. METHODS Eight-month-old male APP/PS1 mice were fed ethanol or control diet intragastrically for 5 weeks (n = 7-11/group). Brain and liver Aβ were assessed using immunoassays. Three important mechanisms of brain amyloidosis were investigated: hepatic LRP-1 (major peripheral Aβ regulator), blood-brain barrier (BBB) function (vascular Aβ regulator), and microglia (major brain Aβ regulator) using immunoassays. Spatial LRP-1 gene expression in the periportal versus pericentral hepatic regions was confirmed using NanoString GeoMx Digital Spatial Profiler. Further, hepatic LRP-1 was silenced by injecting LRP-1 microRNA delivered by the adeno-associated virus 8 (AAV8) and the hepato-specific thyroxine-binding globulin (TBG) promoter to 4-month-old male APP/PS1 mice (n = 6). Control male APP/PS1 mice received control AAV8 (n = 6). Spatial memory and locomotion were assessed 12 weeks after LRP-1 silencing using Y-maze and open-field test, respectively, and brain and liver Aβ were measured. RESULTS Alcohol feeding reduced plaque-associated microglia in APP/PS1 mice brains and increased aggregated Aβ (p < 0.05) by ELISA and 6E10-positive Aβ load by immunostaining (p < 0.05). Increased brain Aβ corresponded with a significant downregulation of hepatic LRP-1 (p < 0.01) at the protein and transcript level, primarily in pericentral hepatocytes (zone 3) where alcohol-induced injury occurs. Hepato-specific LRP-1 silencing significantly increased brain Aβ and locomotion hyperactivity (p < 0.05) in APP/PS1 mice. CONCLUSION Chronic heavy alcohol intake reduced hepatic LRP-1 expression and increased brain Aβ. The hepato-specific LRP-1 silencing similarly increased brain Aβ which was associated with behavioral deficits in APP/PS1 mice. Collectively, our results suggest that hepatic LRP-1 is a key regulator of brain amyloidosis in alcohol-dependent AD.
Collapse
Affiliation(s)
- Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Urvashi R Panchal
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Adenike Oyegbesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Oghenetega E Imiruaye
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Hai Zhang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, United States
| | - Jerome Garcia
- Department of Biology, University of La Verne, La Verne, CA, United States
| | - Kamaljit Kaur
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Sanda Win
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Tin A Than
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Neil Kaplowitz
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Moom R Roosan
- Pharmacy Practice, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States.
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States; Department of Neurology, University of California, Irvine, CA, United States.
| |
Collapse
|
19
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024. [PMID: 39219300 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Engineering Science & Mechanics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
20
|
Zhuo L, Pang K, Dai J, Wu B, Wang J, Xu H, Yang S, Liu Z, Niu R, Yu P, Wang W. The neurophysiological mechanisms of medial prefrontal-perirhinal cortex circuit mediating temporal order memory decline in early stage of AD rats. Neurobiol Dis 2024; 199:106584. [PMID: 38945496 DOI: 10.1016/j.nbd.2024.106584] [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: 03/22/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024] Open
Abstract
The temporal component of episodic memory has been recognized as a sensitive behavioral marker in early stage of Alzheimer's disease (AD) patients. However, parallel studies in AD animals are currently lacking, and the underlying neural circuit mechanisms remain poorly understood. Using a novel AppNL-G-F knock-in (APP-KI) rat model, the developmental changes of temporal order memory (TOM) and the relationship with medial prefrontal cortex and perirhinal cortex (mPFC-PRH) circuit were determined through in vivo electrophysiology and microimaging technique. We observed a deficit in TOM performance during the object temporal order memory task (OTOMT) in APP-KI rats at 6 month old, which was not evident at 3 or 4 months of age. Alongside behavioral changes, we identified a gradually extensive and aggravated regional activation and functional alterations in the mPFC and PRH during the performance of OTOMT, which occurred prior to the onset of TOM deficits. Moreover, coherence analysis showed that the functional connectivity between the mPFC and PRH could predict the extent of future behavioral performance. Further analysis revealed that the aberrant mPFC-PRH interaction mainly attributed to the progressive deterioration of synaptic transmission, information flow and network coordination from mPFC to PRH, suggesting the mPFC dysfunction maybe the key area of origin underlying the early changes of TOM. These findings identify a pivotal role of the mPFC-PRH circuit in mediating the TOM deficits in the early stage of AD, which holds promising clinical translational value and offers potential early biological markers for predicting AD memory progression.
Collapse
Affiliation(s)
- Linan Zhuo
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China; Beijing Key Laboratory of Learning and Cognition, School of Psychology, Capital Normal University, Beijing 100048, China
| | - Keliang Pang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Aging and Longevity Institute & Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiajie Dai
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Bing Wu
- Beijing Key Laboratory of Learning and Cognition, School of Psychology, Capital Normal University, Beijing 100048, China
| | - Jiesi Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hang Xu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuming Yang
- Division of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510062, China
| | - Ziao Liu
- Beijing Key Laboratory of Learning and Cognition, School of Psychology, Capital Normal University, Beijing 100048, China
| | - Rongrong Niu
- Beijing Key Laboratory of Learning and Cognition, School of Psychology, Capital Normal University, Beijing 100048, China
| | - Ping Yu
- Beijing Key Laboratory of Learning and Cognition, School of Psychology, Capital Normal University, Beijing 100048, China.
| | - Weiwen Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
21
|
Damuka N, Irmen RE, Krizan I, Miller M, Gollapelli KK, Bhoopal B, Deep O, Bansode A, Lockhart SN, Orr ME, Jadiya P, Bashetti N, Kumar JVS, Mintz A, Whitlow CT, Craft S, Macauley SL, Solingapuram Sai KK. Exploring microtubule dynamics in Alzheimer's disease: Longitudinal assessment using [ 11C]MPC-6827 PET imaging in rodent models of Alzheimer's-related pathology. Alzheimers Dement 2024; 20:6082-6093. [PMID: 38967283 PMCID: PMC11497705 DOI: 10.1002/alz.14083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Microtubule (MT) stability is crucial for proper neuronal function. Understanding MT dysregulation is critical for connecting amyloid beta (Aβ) and tau-based degenerative events and early changes in presymptomatic Alzheimer's disease (AD). Herein we present positron emission tomography (PET) imaging properties of our MT-PET radiotracer, [11C]MPC-6827, in multiple established AD mouse models. METHODS Longitudinal PET, biodistribution, autoradiography, immunohistochemistry, and behavioral studies were conducted at multiple time points in APPswe/PSEN1dE9 (APP/PS1), P301S-PS19 (P301S), 5xFAD, and age-matched control mice. RESULTS Longitudinal [11C]MPC-6827 brain imaging showed significant increases in APP/PS1, P301S, and 5xFAD mice compared to controls. Longitudinal MT-PET correlated positively with biodistribution, autoradiography, and immunohistochemistry results and negatively with behavior data. DISCUSSION Our study demonstrated significant longitudinal [11C]MPC-6827 PET increases in multiple AD mouse models for the first time. Strong correlations between PET and biomarker data underscored the interplay of MT destabilization, amyloid, and tau pathology in AD. These results suggest [11C]MPC-6827 PET as a promising tool for monitoring MT dysregulation early in AD progression. HIGHLIGHTS Longitudinal positron emission tomography (PET) imaging studies using [11C]MPC-6827 in multiple established Alzheimer's disease (AD) mouse models revealed an early onset of microtubule dysregulation, with significant changes in brain radiotracer uptake evident from 2 to 4 months of age. Intra-group analysis showed a progressive increase in microtubule dysregulation with increasing AD burden, supported by significant correlations between PET imaging data and biodistribution, autoradiography, and molecular pathological markers. [11C]MPC-6827 PET imaging demonstrated its efficacy in detecting early microtubule alterations preceding observable behavioral changes in AD mouse models, suggesting its potential for early AD imaging. The inclusion of the 5xFAD mouse model further elucidated the impact of amyloid beta (Aβ) toxicity on inducing tau hyperphosphorylation-mediated microtubule dysregulation, highlighting the versatility of [11C]MPC-6827 in delineating various aspects of AD pathology. Our study provides immediate clarity on high uptake of the microtubule-based radiotracer in AD brains in a longitudinal setting, which directly informs clinical utility in Aβ/tau-based studies.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of RadiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Riley E. Irmen
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Ivan Krizan
- Department of RadiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Mack Miller
- Department of RadiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | | | - Ojasvi Deep
- Department of RadiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Avinash Bansode
- Department of RadiologyWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samuel N. Lockhart
- Department of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Miranda E. Orr
- Department of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Pooja Jadiya
- Department of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Nagaraju Bashetti
- Department of ChemistryKoneru Lakshmaiah Education FoundationVijayawadaAndhra PradeshIndia
| | - J. V. Shanmukha Kumar
- Department of ChemistryKoneru Lakshmaiah Education FoundationVijayawadaAndhra PradeshIndia
| | - Akiva Mintz
- Department of RadiologyColumbia University Medical CenterNew YorkNew YorkUSA
| | | | - Suzanne Craft
- Department of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | | |
Collapse
|
22
|
Gnörich J, Koehler M, Wind-Mark K, Klaus C, Zatcepin A, Palumbo G, Lalia M, Monasor LS, Beyer L, Eckenweber F, Scheifele M, Gildehaus FJ, von Ungern-Sternberg B, Barthel H, Sabri O, Bartenstein P, Herms J, Tahirovic S, Franzmeier N, Ziegler S, Brendel M. Towards multicenter β-amyloid PET imaging in mouse models: A triple scanner head-to-head comparison. Neuroimage 2024; 297:120748. [PMID: 39069223 DOI: 10.1016/j.neuroimage.2024.120748] [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: 03/29/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
AIM β-amyloid (Aβ) small animal PET facilitates quantification of fibrillar amyloidosis in Alzheimer's disease (AD) mouse models. Thus, the methodology is receiving growing interest as a monitoring tool in preclinical drug trials. In this regard, harmonization of data from different scanners at multiple sites would allow the establishment large collaborative cohorts and may facilitate efficacy comparison of different treatments. Therefore, we objected to determine the level of agreement of Aβ-PET quantification by a head-to-head comparison of three different state-of-the-art small animal PET scanners, which could help pave the way for future multicenter studies. METHODS Within a timeframe of 5 ± 2 weeks, transgenic APPPS1 (n = 9) and wild-type (WT) (n = 8) mice (age range: 13-16 months) were examined three times by Aβ-PET ([18F]florbetaben) using a Siemens Inveon DPET, a MedisonanoScan PET/MR, and a MedisonanoScan PET/CT with harmonized reconstruction protocols. Cortex-to-white-matter 30-60 min p.i. standardized uptake value ratios (SUVRCTX/WM) were calculated to compare binding differences, effect sizes (Cohen's d) and z-score values of APPPS1 relative to WT mice. Correlation coefficients (Pearson's r) were calculated for the agreement of individual SUVR between different scanners. Voxel-wise analysis was used to determine the agreement of spatial pathology patterns. For validation of PET imaging against the histological gold standard, individual SUVR values were subject to a correlation analysis with area occupancy of methoxy‑X04 staining. RESULTS All three small animal PET scanners yielded comparable group differences between APPPS1 and WT mice (∆PET=20.4 % ± 2.9 %, ∆PET/MR=18.4 % ± 4.5 %, ∆PET/CT=18.1 % ± 3.3 %). Voxel-wise analysis confirmed a high degree of congruency of the spatial pattern (Dice coefficient (DC)PETvs.PET/MR=83.0 %, DCPETvs.PET/CT=69.3 %, DCPET/MRvs.PET/CT=81.9 %). Differences in the group level variance of the three scanners resulted in divergent z-scores (zPET=11.5 ± 1.6; zPET/MR=5.3 ± 1.3; zPET/CT=3.4 ± 0.6) and effect sizes (dPET=8.5, dPET/MR=4.5, dPET/CT=4.1). However, correlations at the individual mouse level were still strong between scanners (rPETvs.PET/MR=0.96, rPETvs.PET/CT=0.91, rPET/MRvs.PET/CT=0.87; all p ≤ 0.0001). Methoxy-X04 staining exhibited a significant correlation across all three PET machines combined (r = 0.76, p < 0.0001) but also at individual level (PET: r = 0.81, p = 0.026; PET/MR: r = 0.89, p = 0.0074; PET/CT: r = 0.93, p = 0.0028). CONCLUSIONS Our comparison of standardized small animal Aβ-PET acquired by three different scanners substantiates the possibility of moving towards a multicentric approach in preclinical AD research. The alignment of image acquisition and analysis methods achieved good overall comparability between data sets. Nevertheless, differences in variance of sensitivity and specificity of different scanners may limit data interpretation at the individual mouse level and deserves methodological optimization.
Collapse
Affiliation(s)
- Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| | - Mara Koehler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Manvir Lalia
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Laura Sebastian Monasor
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Graduate School of Systemic Neuroscience, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany.
| |
Collapse
|
23
|
Margutti P, D’Ambrosio A, Zamboni S. Microbiota-Derived Extracellular Vesicle as Emerging Actors in Host Interactions. Int J Mol Sci 2024; 25:8722. [PMID: 39201409 PMCID: PMC11354844 DOI: 10.3390/ijms25168722] [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: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The human microbiota is an intricate micro-ecosystem comprising a diverse range of dynamic microbial populations mainly consisting of bacteria, whose interactions with hosts strongly affect several physiological and pathological processes. The gut microbiota is being increasingly recognized as a critical player in maintaining homeostasis, contributing to the main functions of the intestine and distal organs such as the brain. However, gut dysbiosis, characterized by composition and function alterations of microbiota with intestinal barrier dysfunction has been linked to the development and progression of several pathologies, including intestinal inflammatory diseases, systemic autoimmune diseases, such as rheumatic arthritis, and neurodegenerative diseases, such as Alzheimer's disease. Moreover, oral microbiota research has gained significant interest in recent years due to its potential impact on overall health. Emerging evidence on the role of microbiota-host interactions in health and disease has triggered a marked interest on the functional role of bacterial extracellular vesicles (BEVs) as mediators of inter-kingdom communication. Accumulating evidence reveals that BEVs mediate host interactions by transporting and delivering into host cells effector molecules that modulate host signaling pathways and cell processes, influencing health and disease. This review discusses the critical role of BEVs from the gut, lung, skin and oral cavity in the epithelium, immune system, and CNS interactions.
Collapse
Affiliation(s)
- Paola Margutti
- Department of Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.); (S.Z.)
| | | | | |
Collapse
|
24
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
25
|
Kramer M, Hoang TH, Yang H, Shchyglo O, Böge J, Neubacher U, Colitti-Klausnitzer J, Manahan-Vaughan D. Intracerebral inoculation of healthy non-transgenic rats with a single aliquot of oligomeric amyloid-β (1-42) profoundly and progressively alters brain function throughout life. Front Aging Neurosci 2024; 16:1397901. [PMID: 39156737 PMCID: PMC11327071 DOI: 10.3389/fnagi.2024.1397901] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
One of the puzzling aspects of sporadic Alzheimer's disease (AD) is how it commences. Changes in one key brain peptide, amyloid-beta (Aβ), accompany disease progression, but whether this comprises a trigger or a consequence of AD is still a topic of debate. It is clear however that the cerebral presence of oligomeric Aβ (1-42) is a key factor in early AD-pathogenesis. Furthermore, treatment of rodent brains with oligomeric Aβ (1-42) either in vitro or in vivo, acutely impairs hippocampal synaptic plasticity, creating a link between Aβ-pathology and learning impairments. Here, we show that a once-off inoculation of the brains of healthy adult rats with oligomeric Aβ (1-42) exerts debilitating effects on the long-term viability of the hippocampus, one of the primary targets of AD. Changes are progressive: months after treatment, synaptic plasticity, neuronal firing and spatial learning are impaired and expression of plasticity-related proteins are changed, in the absence of amyloid plaques. Early changes relate to activation of microglia, whereas later changes are associated with a reconstruction of astroglial morphology. These data suggest that a disruption of Aβ homeostasis may suffice to trigger an irreversible cascade, underlying progressive loss of hippocampal function, that parallels the early stages of AD.
Collapse
|
26
|
Fernandes S, Revanna J, Pratt J, Hayes N, Marchetto MC, Gage FH. Modeling Alzheimer's disease using human cell derived brain organoids and 3D models. Front Neurosci 2024; 18:1434945. [PMID: 39156632 PMCID: PMC11328153 DOI: 10.3389/fnins.2024.1434945] [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: 05/19/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024] Open
Abstract
Age-related neurodegenerative diseases, like Alzheimer's disease (AD), are challenging diseases for those affected with no cure and limited treatment options. Functional, human derived brain tissues that represent the diverse genetic background and cellular subtypes contributing to sporadic AD (sAD) are limited. Human stem cell derived brain organoids recapitulate some features of human brain cytoarchitecture and AD-like pathology, providing a tool for illuminating the relationship between AD pathology and neural cell dysregulation leading to cognitive decline. In this review, we explore current strategies for implementing brain organoids in the study of AD as well as the challenges associated with investigating age-related brain diseases using organoid models.
Collapse
Affiliation(s)
- Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Jasmin Revanna
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joshua Pratt
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biology, San Diego State University, San Diego, CA, United States
| | - Nicholas Hayes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, California State University, San Marcos, CA, United States
| | - Maria C. Marchetto
- Department of Anthropology, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, United States
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
27
|
Sánchez-Puebla L, de Hoz R, Salobrar-García E, Arias-Vázquez A, González-Jiménez M, Ramírez AI, Fernández-Albarral JA, Matamoros JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto Vaquero C, Cuartero MI, Moro MA, Salazar JJ, López-Cuenca I, Ramírez JM. Age-Related Retinal Layer Thickness Changes Measured by OCT in APPNL-F/NL-F Mice: Implications for Alzheimer's Disease. Int J Mol Sci 2024; 25:8221. [PMID: 39125789 PMCID: PMC11312090 DOI: 10.3390/ijms25158221] [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: 05/29/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
In Alzheimer's disease (AD), transgenic mouse models have established links between abnormalities in the retina and those in the brain. APPNL-F/NL-F is a murine, humanized AD model that replicates several pathological features observed in patients with AD. Research has focused on obtaining quantitative parameters from optical coherence tomography (OCT) in AD. The aim of this study was to analyze, in a transversal case-control study using manual retinal segmentation via SD-OCT, the changes occurring in the retinal layers of the APPNL/F-NF/L AD model in comparison to C57BL/6J mice (WT) at 6, 9, 12, 15, 17, and 20 months of age. The analysis focused on retinal thickness in RNFL-GCL, IPL, INL, OPL, and ONL based on the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Both APPNL-F/NL-F-model and WT animals exhibited thickness changes at the time points studied. While WT showed significant changes in INL, OPL, and ONL, the AD model showed changes in all retinal layers analyzed. The APPNL-F/NL-F displayed significant thickness variations in the analyzed layers except for the IPL compared to related WT. These thickness changes closely resembled those found in humans during preclinical stages, as well as during mild and moderate AD stages, making this AD model behave more similarly to the disease in humans.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Brain Science Institute, RIKEN, Laboratory for Proteolytic Neuroscience, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto Vaquero
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
28
|
Delvenne A, Vandendriessche C, Gobom J, Burgelman M, Dujardin P, De Nolf C, Tijms BM, Teunissen CE, Schindler SE, Verhey F, Ramakers I, Martinez-Lage P, Tainta M, Vandenberghe R, Schaeverbeke J, Engelborghs S, De Roeck E, Popp J, Peyratout G, Tsolaki M, Freund-Levi Y, Lovestone S, Streffer J, Bertram L, Blennow K, Zetterberg H, Visser PJ, Vandenbroucke RE, Vos SJB. Involvement of the choroid plexus in Alzheimer's disease pathophysiology: findings from mouse and human proteomic studies. Fluids Barriers CNS 2024; 21:58. [PMID: 39020361 PMCID: PMC11256635 DOI: 10.1186/s12987-024-00555-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/03/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Structural and functional changes of the choroid plexus (ChP) have been reported in Alzheimer's disease (AD). Nonetheless, the role of the ChP in the pathogenesis of AD remains largely unknown. We aim to unravel the relation between ChP functioning and core AD pathogenesis using a unique proteomic approach in mice and humans. METHODS We used an APP knock-in mouse model, APPNL-G-F, exhibiting amyloid pathology, to study the association between AD brain pathology and protein changes in mouse ChP tissue and CSF using liquid chromatography mass spectrometry. Mouse proteomes were investigated at the age of 7 weeks (n = 5) and 40 weeks (n = 5). Results were compared with previously published human AD CSF proteomic data (n = 496) to identify key proteins and pathways associated with ChP changes in AD. RESULTS ChP tissue proteome was dysregulated in APPNL-G-F mice relative to wild-type mice at both 7 and 40 weeks. At both ages, ChP tissue proteomic changes were associated with epithelial cells, mitochondria, protein modification, extracellular matrix and lipids. Nonetheless, some ChP tissue proteomic changes were different across the disease trajectory; pathways related to lysosomal function, endocytosis, protein formation, actin and complement were uniquely dysregulated at 7 weeks, while pathways associated with nervous system, immune system, protein degradation and vascular system were uniquely dysregulated at 40 weeks. CSF proteomics in both mice and humans showed similar ChP-related dysregulated pathways. CONCLUSIONS Together, our findings support the hypothesis of ChP dysfunction in AD. These ChP changes were related to amyloid pathology. Therefore, the ChP could become a novel promising therapeutic target for AD.
Collapse
Affiliation(s)
- Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Johan Gobom
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Marlies Burgelman
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Clint De Nolf
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (AUMC), Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, USA
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, USA
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Inez Ramakers
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | | | - Mikel Tainta
- Fundación CITA-Alzhéimer Fundazioa, San Sebastian, Spain
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Louvain, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | - Jolien Schaeverbeke
- Neurology Service, University Hospitals Leuven, Louvain, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel, Brussels, Belgium
- NEUR Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ellen De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Julius Popp
- Old Age Psychiatry, University Hospital Lausanne, Lausanne, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatry University Hospital Zürich, Zurich, Switzerland
| | | | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Makedonia, Thessaloniki, Greece
| | - Yvonne Freund-Levi
- Department of Neurobiology, Caring Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry in Region Örebro County and School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Old Age Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Simon Lovestone
- University of Oxford, Oxford, UK
- Johnson and Johnson Medical Ltd., Wokingham, UK
| | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- H. Lundbeck A/S, Valby, Denmark
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| |
Collapse
|
29
|
Caradonna E, Nemni R, Bifone A, Gandolfo P, Costantino L, Giordano L, Mormone E, Macula A, Cuomo M, Difruscolo R, Vanoli C, Vanoli E, Ferrara F. The Brain-Gut Axis, an Important Player in Alzheimer and Parkinson Disease: A Narrative Review. J Clin Med 2024; 13:4130. [PMID: 39064171 PMCID: PMC11278248 DOI: 10.3390/jcm13144130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are severe age-related disorders with complex and multifactorial causes. Recent research suggests a critical link between neurodegeneration and the gut microbiome, via the gut-brain communication pathway. This review examines the role of trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite, in the development of AD and PD, and investigates its interaction with microRNAs (miRNAs) along this bidirectional pathway. TMAO, which is produced from dietary metabolites like choline and carnitine, has been linked to increased neuroinflammation, protein misfolding, and cognitive decline. In AD, elevated TMAO levels are associated with amyloid-beta and tau pathologies, blood-brain barrier disruption, and neuronal death. TMAO can cross the blood-brain barrier and promote the aggregation of amyloid and tau proteins. Similarly, TMAO affects alpha-synuclein conformation and aggregation, a hallmark of PD. TMAO also activates pro-inflammatory pathways such as NF-kB signaling, exacerbating neuroinflammation further. Moreover, TMAO modulates the expression of various miRNAs that are involved in neurodegenerative processes. Thus, the gut microbiome-miRNA-brain axis represents a newly discovered mechanistic link between gut dysbiosis and neurodegeneration. MiRNAs regulate the key pathways involved in neuroinflammation, oxidative stress, and neuronal death, contributing to disease progression. As a direct consequence, specific miRNA signatures may serve as potential biomarkers for the early detection and monitoring of AD and PD progression. This review aims to elucidate the complex interrelationships between the gut microbiota, trimethylamine-N-oxide (TMAO), microRNAs (miRNAs), and the central nervous system, and the implications of these connections in neurodegenerative diseases. In this context, an overview of the current neuroradiology techniques available for studying neuroinflammation and of the animal models used to investigate these intricate pathologies will also be provided. In summary, a bulk of evidence supports the concept that modulating the gut-brain communication pathway through dietary changes, the manipulation of the microbiome, and/or miRNA-based therapies may offer novel approaches for implementing the treatment of debilitating neurological disorders.
Collapse
Affiliation(s)
- Eugenio Caradonna
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| | - Raffaello Nemni
- Unit of Neurology, Centro Diagnostico Italiano S.p.A., Milan Fondazione Crespi Spano, 20011 Milan, Italy;
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Angelo Bifone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Patrizia Gandolfo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Lucy Costantino
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Luca Giordano
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Elisabetta Mormone
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Anna Macula
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Colleretto Giacosa, 10010 Turin, Italy;
- Department of Physics, University of Torino, 10124 Torino, Italy
| | - Mariarosa Cuomo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | | | - Camilla Vanoli
- Department of Clinical Psychology, Antioch University Los Angeles, Culver City, CA 90230, USA
| | - Emilio Vanoli
- School of Nursing, Cardiovascular Diseases, University of Pavia, 27100 Pavia, Italy;
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| |
Collapse
|
30
|
Sánchez-Puebla L, López-Cuenca I, Salobrar-García E, González-Jiménez M, Arias-Vázquez A, Matamoros JA, Ramírez AI, Fernández-Albarral JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto-Vaquero C, Cuartero MI, Moro MA, Salazar JJ, de Hoz R, Ramírez JM. Retinal Vascular and Structural Changes in the Murine Alzheimer's APPNL-F/NL-F Model from 6 to 20 Months. Biomolecules 2024; 14:828. [PMID: 39062542 PMCID: PMC11274728 DOI: 10.3390/biom14070828] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) may manifest retinal changes preceding brain pathology. A transversal case-control study utilized spectral-domain OCT angiography (SD-OCTA) and Angio-Tool software 0.6a to assess retinal vascular structures and OCT for inner and outer retina thickness in the APPNL-F/NL-F AD model at 6, 9, 12, 15, 17, and 20 months old. Comparisons to age-matched wild type (WT) were performed. The analysis focused on the three vascular plexuses using AngiooTool and on retinal thickness, which was represented with the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Compared to WT, the APPNL-F/NL-F group exhibited both vascular and structural changes as early as 6 months persisting and evolving at 15, 17, and 20 months. Significant vascular alterations, principally in the superficial vascular complex (SVC), were observed. There was a significant decrease in the vessel area and the total vessel length in SVC, intermediate, and deep capillary plexus. The inner retina in the APPNL-F/NL-F group predominantly decreased in thickness while the outer retina showed increased thickness in most analyzed time points compared to the control group. There are early vascular and structural retinal changes that precede the cognitive changes, which appear at later stages. Therefore, the natural history of the APPNL-F/NL-F model may be more similar to human AD than other transgenic models.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto-Vaquero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
31
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PB, Perkins B, Soni D, Ingraham C, Lee‐Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. Alzheimers Dement 2024; 20:4970-4984. [PMID: 38687251 PMCID: PMC11247676 DOI: 10.1002/alz.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. HIGHLIGHTS A novel approach to validate genetic risk factors for late-onset AD (LOAD) is presented. LOAD risk variants were knocked in to conserved mouse loci. Variant effects were assayed by transcriptional analysis. Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease. This approach should generate more translationally relevant animal models.
Collapse
Affiliation(s)
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Asli Uyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | | | | | - Adrian L. Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | | | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| |
Collapse
|
32
|
Huang HX, Inglese P, Tang J, Yagoubi R, Correia GDS, Horneffer-van der Sluis VM, Camuzeaux S, Wu V, Kopanitsa MV, Willumsen N, Jackson JS, Barron AM, Saito T, Saido TC, Gentlemen S, Takats Z, Matthews PM. Mass spectrometry imaging highlights dynamic patterns of lipid co-expression with Aβ plaques in mouse and human brains. J Neurochem 2024; 168:1193-1214. [PMID: 38372586 DOI: 10.1111/jnc.16042] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/13/2023] [Accepted: 12/06/2023] [Indexed: 02/20/2024]
Abstract
Lipids play crucial roles in the susceptibility and brain cellular responses to Alzheimer's disease (AD) and are increasingly considered potential soluble biomarkers in cerebrospinal fluid (CSF) and plasma. To delineate the pathological correlations of distinct lipid species, we conducted a comprehensive characterization of both spatially localized and global differences in brain lipid composition in AppNL-G-F mice with spatial and bulk mass spectrometry lipidomic profiling, using human amyloid-expressing (h-Aβ) and WT mouse brains controls. We observed age-dependent increases in lysophospholipids, bis(monoacylglycerol) phosphates, and phosphatidylglycerols around Aβ plaques in AppNL-G-F mice. Immunohistology-based co-localization identified associations between focal pro-inflammatory lipids, glial activation, and autophagic flux disruption. Likewise, in human donors with varying Braak stages, similar studies of cortical sections revealed co-expression of lysophospholipids and ceramides around Aβ plaques in AD (Braak stage V/VI) but not in earlier Braak stage controls. Our findings in mice provide evidence of temporally and spatially heterogeneous differences in lipid composition as local and global Aβ-related pathologies evolve. Observing similar lipidomic changes associated with pathological Aβ plaques in human AD tissue provides a foundation for understanding differences in CSF lipids with reported clinical stage or disease severity.
Collapse
Affiliation(s)
- Helen Xuexia Huang
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Paolo Inglese
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
| | - Riad Yagoubi
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Gonçalo D S Correia
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Stephane Camuzeaux
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Vincen Wu
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Nanet Willumsen
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Johanna S Jackson
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Steve Gentlemen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Zoltan Takats
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Paul M Matthews
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
33
|
Rice M, Nuovo GJ, Sawant D, Mishra A, Tili E. Comparison of Neuroinflammation Induced by Hyperphosphorylated Tau Protein Versus Ab42 in Alzheimer's Disease. Mol Neurobiol 2024; 61:4589-4601. [PMID: 38105410 DOI: 10.1007/s12035-023-03822-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/11/2023] [Indexed: 12/19/2023]
Abstract
Both neurofibrillary tangles and senile plaques are associated with inflammation in Alzheimer's disease (AD). Their relative degree of induced neuroinflammation, however, is not well established. Mouse models of AD that expressed either human Aβ42 (n = 7) or human hyperphosphorylated tau protein alone (n = 3), wild type (n = 10), and human AD samples (n = 29 with 18 controls) were studied. The benefit of using mouse models that possess only human tau or amyloid-b is that it allows for the individual evaluation of how each protein affects neuroinflammation, something not possible in human tissue. Three indicators of neuroinflammation were examined: TLRs/RIG1 expression, the density of astrocytes and microglial cells, and well-established mediators of neuroinflammation (IL6, TNFα, IL1β, and CXCL10). There was a statistically significant increase in neuroinflammation with all three variables in the mouse models with human tau only as compared to human Aβ42 only or wild-type mice (each at p < 0.0001). Only the Aβ42 5xFAD mice (n = 4) showed statistically higher neuroinflammation versus wild type (p = 0.0030). The human AD tissues were segregated into Aβ42 only or hyperphosphorylated tau protein with Aβ42. The latter areas showed increased neuroinflammation with each of the three variables compared to the areas with only Aβ42. Of the TLRs and RIG-1, TLR8 was significantly elevated in both the mouse model and human AD and only in areas with the abnormal tau protein. It is concluded that although Aβ42 and hyperphosphorylated tau protein can each induce inflammation, the latter protein is associated with a much stronger neuroinflammatory response vis-a-vis a significantly greater activated microglial response.
Collapse
Affiliation(s)
| | | | | | | | - Esmerina Tili
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
34
|
Qu W, Lam M, McInvale JJ, Mares JA, Kwon S, Humala N, Mahajan A, Nguyen T, Jakubiak KA, Mun JY, Tedesco TG, Al-Dalahmah O, Hussaini SA, Sproul AA, Siegelin MD, De Jager PL, Canoll P, Menon V, Hargus G. Xenografted human iPSC-derived neurons with the familial Alzheimer's disease APP V717I mutation reveal dysregulated transcriptome signatures linked to synaptic function and implicate LINGO2 as a disease signaling mediator. Acta Neuropathol 2024; 147:107. [PMID: 38918213 PMCID: PMC11199265 DOI: 10.1007/s00401-024-02755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and disease mechanisms are still not fully understood. Here, we explored pathological changes in human induced pluripotent stem cell (iPSC)-derived neurons carrying the familial AD APPV717I mutation after cell injection into the mouse forebrain. APPV717I mutant iPSCs and isogenic controls were differentiated into neurons revealing enhanced Aβ42 production, elevated phospho-tau, and impaired neurite outgrowth in APPV717I neurons. Two months after transplantation, APPV717I and control neural cells showed robust engraftment but at 12 months post-injection, APPV717I grafts were smaller and demonstrated impaired neurite outgrowth compared to controls, while plaque and tangle pathology were not seen. Single-nucleus RNA-sequencing of micro-dissected grafts, performed 2 months after cell injection, identified significantly altered transcriptome signatures in APPV717I iPSC-derived neurons pointing towards dysregulated synaptic function and axon guidance. Interestingly, APPV717I neurons showed an increased expression of genes, many of which are also upregulated in postmortem neurons of AD patients including the transmembrane protein LINGO2. Downregulation of LINGO2 in cultured APPV717I neurons rescued neurite outgrowth deficits and reversed key AD-associated transcriptional changes related but not limited to synaptic function, apoptosis and cellular senescence. These results provide important insights into transcriptional dysregulation in xenografted APPV717I neurons linked to synaptic function, and they indicate that LINGO2 may represent a potential therapeutic target in AD.
Collapse
Affiliation(s)
- Wenhui Qu
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Matti Lam
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Neurological Institute, Columbia University, 710 West 168th Street, New York, NY, USA
| | - Julie J McInvale
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Jason A Mares
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Neurological Institute, Columbia University, 710 West 168th Street, New York, NY, USA
| | - Sam Kwon
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Nelson Humala
- Department of Neurosurgery, Columbia University, New York, NY, USA
| | - Aayushi Mahajan
- Department of Neurosurgery, Columbia University, New York, NY, USA
| | - Trang Nguyen
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Kelly A Jakubiak
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Jeong-Yeon Mun
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Thomas G Tedesco
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Osama Al-Dalahmah
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Syed A Hussaini
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Andrew A Sproul
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Philip L De Jager
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Neurological Institute, Columbia University, 710 West 168th Street, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Neurological Institute, Columbia University, 710 West 168th Street, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Presbyterian Hospital, Columbia University, 650W 168th Street, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
35
|
Tang J, Huang H, Muirhead RCJ, Zhou Y, Li J, DeFelice J, Kopanitsa MV, Serneels L, Davey K, Tilley BS, Gentleman S, Matthews PM. Associations of amyloid-β oligomers and plaques with neuropathology in the App NL-G-F mouse. Brain Commun 2024; 6:fcae218. [PMID: 39035420 PMCID: PMC11258573 DOI: 10.1093/braincomms/fcae218] [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: 08/29/2023] [Revised: 03/22/2024] [Accepted: 06/23/2024] [Indexed: 07/23/2024] Open
Abstract
Amyloid-β pathology and neurofibrillary tangles lead to glial activation and neurodegeneration in Alzheimer's disease. In this study, we investigated the relationships between the levels of amyloid-β oligomers, amyloid-β plaques, glial activation and markers related to neurodegeneration in the App NL-G-F triple mutation mouse line and in a knock-in line homozygous for the common human amyloid precursor protein (App hu mouse). The relationships between neuropathological features were characterized with immunohistochemistry and imaging mass cytometry. Markers assessing human amyloid-β proteins, microglial and astrocytic activation and neuronal and synaptic densities were used in mice between 2.5 and 12 months of age. We found that amyloid-β oligomers were abundant in the brains of App hu mice in the absence of classical amyloid-β plaques. These brains showed morphological changes consistent with astrocyte activation but no evidence of microglial activation or synaptic or neuronal pathology. In contrast, both high levels of amyloid-β oligomers and numerous plaques accumulated in App NL-G-F mice in association with substantial astrocytic and microglial activation. The increase in amyloid-β oligomers over time was more strongly correlated with astrocytic than with microglia activation. Spatial analyses suggested that activated microglia were more closely associated with amyloid-β oligomers than with amyloid-β plaques in App NL-G-F mice, which also showed age-dependent decreases in neuronal and synaptic density markers. A comparative study of the two models highlighted the dependence of glial and neuronal pathology on the nature and aggregation state of the amyloid-β peptide. Astrocyte activation and neuronal pathology appeared to be more strongly associated with amyloid-β oligomers than with amyloid-β plaques, although amyloid-β plaques were associated with microglia activation.
Collapse
Affiliation(s)
- Jiabin Tang
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Department of Anesthesiology, Weill Cornell Medicine, Cornell University, New York, NY 11106, USA
| | - Helen Huang
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Robert C J Muirhead
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Randall Centre for Cell and Molecular Biophysics, Kings College London, London SE5 9RX, UK
| | - Yue Zhou
- Department of Mechanical Engineering, Roberts Engineering Building, University College London, London WC1E 7JE, UK
| | - Junheng Li
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
| | - John DeFelice
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Lutgarde Serneels
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), 9052 Gent, Belgium
| | - Karen Davey
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- UK Dementia Research Institute, Kings College London, Denmark Hill Campus, London SE5 9RX, UK
| | - Bension S Tilley
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Steve Gentleman
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Paul M Matthews
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| |
Collapse
|
36
|
Kanoh T, Mizoguchi T, Tonoki A, Itoh M. Modeling of age-related neurological disease: utility of zebrafish. Front Aging Neurosci 2024; 16:1399098. [PMID: 38765773 PMCID: PMC11099255 DOI: 10.3389/fnagi.2024.1399098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Many age-related neurological diseases still lack effective treatments, making their understanding a critical and urgent issue in the globally aging society. To overcome this challenge, an animal model that accurately mimics these diseases is essential. To date, many mouse models have been developed to induce age-related neurological diseases through genetic manipulation or drug administration. These models help in understanding disease mechanisms and finding potential therapeutic targets. However, some age-related neurological diseases cannot be fully replicated in human pathology due to the different aspects between humans and mice. Although zebrafish has recently come into focus as a promising model for studying aging, there are few genetic zebrafish models of the age-related neurological disease. This review compares the aging phenotypes of humans, mice, and zebrafish, and provides an overview of age-related neurological diseases that can be mimicked in mouse models and those that cannot. We presented the possibility that reproducing human cerebral small vessel diseases during aging might be difficult in mice, and zebrafish has potential to be another animal model of such diseases due to their similarity of aging phenotype to humans.
Collapse
Affiliation(s)
- Tohgo Kanoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ayako Tonoki
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| |
Collapse
|
37
|
Homanics GE, Park JE, Bailey L, Schaeffer DJ, Schaeffer L, He J, Li S, Zhang T, Haber A, Spruce C, Greenwood A, Murai T, Schultz L, Mongeau L, Ha S, Oluoch J, Stein B, Choi SH, Huhe H, Thathiah A, Strick PL, Carter GW, Silva AC, Sukoff Rizzo SJ. Early molecular events of autosomal-dominant Alzheimer's disease in marmosets with PSEN1 mutations. Alzheimers Dement 2024; 20:3455-3471. [PMID: 38574388 PMCID: PMC11095452 DOI: 10.1002/alz.13806] [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: 11/15/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Fundamental questions remain about the key mechanisms that initiate Alzheimer's disease (AD) and the factors that promote its progression. Here we report the successful generation of the first genetically engineered marmosets that carry knock-in (KI) point mutations in the presenilin 1 (PSEN1) gene that can be studied from birth throughout lifespan. METHODS CRISPR/Cas9 was used to generate marmosets with C410Y or A426P point mutations in PSEN1. Founders and their germline offspring are comprehensively studied longitudinally using non-invasive measures including behavior, biomarkers, neuroimaging, and multiomics signatures. RESULTS Prior to adulthood, increases in plasma amyloid beta were observed in PSEN1 mutation carriers relative to non-carriers. Analysis of brain revealed alterations in several enzyme-substrate interactions within the gamma secretase complex prior to adulthood. DISCUSSION Marmosets carrying KI point mutations in PSEN1 provide the opportunity to study the earliest primate-specific mechanisms that contribute to the molecular and cellular root causes of AD onset and progression. HIGHLIGHTS We report the successful generation of genetically engineered marmosets harboring knock-in point mutations in the PSEN1 gene. PSEN1 marmosets and their germline offspring recapitulate the early emergence of AD-related biomarkers. Studies as early in life as possible in PSEN1 marmosets will enable the identification of primate-specific mechanisms that drive disease progression.
Collapse
Affiliation(s)
- Gregg E. Homanics
- Department of Anesthesiology & Perioperative MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jung Eun Park
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lauren Bailey
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - David J. Schaeffer
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lauren Schaeffer
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jie He
- Department of StatisticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Shuoran Li
- Department of StatisticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Tingting Zhang
- Department of StatisticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | | | | | | | - Takeshi Murai
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Laura Schultz
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lauren Mongeau
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Seung‐Kwon Ha
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Julia Oluoch
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Brianne Stein
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Sang Ho Choi
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Hasi Huhe
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Amantha Thathiah
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Peter L. Strick
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | | | - Afonso C. Silva
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Stacey J. Sukoff Rizzo
- Department of NeurobiologyUniversity of Pittsburgh Brain InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of MedicineUniversity of Pittsburgh Aging Institute, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
38
|
Choe MS, Yeo HC, Kim JS, Lee J, Lee HJ, Kim HR, Baek KM, Jung NY, Choi M, Lee MY. Simple modeling of familial Alzheimer's disease using human pluripotent stem cell-derived cerebral organoid technology. Stem Cell Res Ther 2024; 15:118. [PMID: 38659053 PMCID: PMC11040922 DOI: 10.1186/s13287-024-03732-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Cerebral organoids (COs) are the most advanced in vitro models that resemble the human brain. The use of COs as a model for Alzheimer's disease (AD), as well as other brain diseases, has recently gained attention. This study aimed to develop a human AD CO model using normal human pluripotent stem cells (hPSCs) that recapitulates the pathological phenotypes of AD and to determine the usefulness of this model for drug screening. METHODS We established AD hPSC lines from normal hPSCs by introducing genes that harbor familial AD mutations, and the COs were generated using these hPSC lines. The pathological features of AD, including extensive amyloid-β (Aβ) accumulation, tauopathy, and neurodegeneration, were analyzed using enzyme-linked immunosorbent assay, Amylo-Glo staining, thioflavin-S staining, immunohistochemistry, Bielschowsky's staining, and western blot analysis. RESULTS The AD COs exhibited extensive Aβ accumulation. The levels of paired helical filament tau and neurofibrillary tangle-like silver deposits were highly increased in the AD COs. The number of cells immunoreactive for cleaved caspase-3 was significantly increased in the AD COs. In addition, treatment of AD COs with BACE1 inhibitor IV, a β-secretase inhibitor, and compound E, a γ-secretase inhibitor, significantly attenuated the AD pathological features. CONCLUSION Our model effectively recapitulates AD pathology. Hence, it is a valuable platform for understanding the mechanisms underlying AD pathogenesis and can be used to test the efficacy of anti-AD drugs.
Collapse
Affiliation(s)
- Mu Seog Choe
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea
| | - Han Cheol Yeo
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea
| | - Joong Sun Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Chonnam National University, 61186, Gwangju, Republic of Korea
| | - Jean Lee
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 03080, Seoul, Republic of Korea
| | - Hae Jun Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), 01812, Seoul, Republic of Korea
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, 54896, Jeonju, Republic of Korea
| | - Kyung Min Baek
- Department of Cardiovascular and Neurologic Disease, College of Oriental Medicine, Daegu Haany University, 42158, Daegu, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, 50612, Yangsan, Republic of Korea
| | - Murim Choi
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 03080, Seoul, Republic of Korea.
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea.
| |
Collapse
|
39
|
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
|
40
|
Kuhn MK, Kang RY, Kim C, Tagay Y, Morris N, Tabdanov ED, Elcheva IA, Proctor EA. Dynamic neuroinflammatory profiles predict Alzheimer's disease pathology in microglia-containing cerebral organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.16.567220. [PMID: 38014053 PMCID: PMC10680718 DOI: 10.1101/2023.11.16.567220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Neuroinflammation and the underlying dysregulated immune responses of microglia actively contribute to the progression and, likely, the initiation of Alzheimer's disease (AD). Fine-tuned therapeutic modulation of immune dysfunction to ameliorate disease cannot be achieved without the characterization of diverse microglial states that initiate unique, and sometimes contradictory, immune responses that evolve over time in chronic inflammatory environments. Because of the functional differences between human and murine microglia, untangling distinct, disease-relevant reactive states and their corresponding effects on pathology or neuronal health may not be possible without the use of human cells. In order to profile shifting microglial states in early AD and identify microglia-specific drivers of disease, we differentiated human induced pluripotent stem cells (iPSCs) carrying a familial AD PSEN2 mutation or its isogenic control into cerebral organoids and quantified the changes in cytokine concentrations over time with Luminex XMAP technology. We used partial least squares (PLS) modeling to build cytokine signatures predictive of disease and age to identify key differential patterns of cytokine expression that inform the overall organoid immune milieu and quantified the corresponding changes in protein pathology. AD organoids exhibited an overall reduction in cytokine secretion after an initial amplified immune response. We demonstrate that reduced synapse density observed in the AD organoids is prevented with microglial depletion. Crucially, these differential effects of dysregulated immune signaling occurred without the accumulation of pathological proteins. In this study, we used microglia-containing AD organoids to quantitatively characterize an evolving immune milieu, made up of a diverse of collection of activation patterns and immune responses, to identify how a dynamic, overall neuroinflammatory state negatively impacts neuronal health and the cell-specific contribution of microglia.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Rachel Y Kang
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - ChaeMin Kim
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Yerbol Tagay
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Nathan Morris
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Erdem D Tabdanov
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | - Irina A Elcheva
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
41
|
Murai T, Bailey L, Schultz L, Mongeau L, DeSana A, Silva AC, Roberts AC, Sukoff Rizzo SJ. Improving preclinical to clinical translation of cognitive function for aging-related disorders: the utility of comprehensive touchscreen testing batteries in common marmosets. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2024; 24:325-348. [PMID: 38200282 PMCID: PMC11039501 DOI: 10.3758/s13415-023-01144-x] [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] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
Concerns about poor animal to human translation have come increasingly to the fore, in particular with regards to cognitive improvements in rodent models, which have failed to translate to meaningful clinical benefit in humans. This problem has been widely acknowledged, most recently in the field of Alzheimer's disease, although this issue pervades the spectrum of central nervous system (CNS) disorders, including neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Consequently, recent efforts have focused on improving preclinical to clinical translation by incorporating more clinically analogous outcome measures of cognition, such as touchscreen-based assays, which can be employed across species, and have great potential to minimize the translational gap. For aging-related research, it also is important to incorporate model systems that facilitate the study of the long prodromal phase in which cognitive decline begins to emerge and which is a major limitation of short-lived species, such as laboratory rodents. We posit that to improve translation of cognitive function and dysfunction, nonhuman primate models, which have conserved anatomical and functional organization of the primate brain, are necessary to move the field of translational research forward and to bridge the translational gaps. The present studies describe the establishment of a comprehensive battery of touchscreen-based tasks that capture a spectrum of domains sensitive to detecting aging-related cognitive decline, which will provide the greatest benefit through longitudinal evaluation throughout the prolonged lifespan of the marmoset.
Collapse
Affiliation(s)
- Takeshi Murai
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Bailey
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura Schultz
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Mongeau
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrew DeSana
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Afonso C Silva
- Department of Neurobiology, University of Pittsburgh School of Medicine, 514A Bridgeside Point 1, 100 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Stacey J Sukoff Rizzo
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Neurobiology, University of Pittsburgh School of Medicine, 514A Bridgeside Point 1, 100 Technology Drive, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
42
|
Jiang L, Roberts R, Wong M, Zhang L, Webber CJ, Libera J, Wang Z, Kilci A, Jenkins M, Ortiz AR, Dorrian L, Sun J, Sun G, Rashad S, Kornbrek C, Daley SA, Dedon PC, Nguyen B, Xia W, Saito T, Saido TC, Wolozin B. β-amyloid accumulation enhances microtubule associated protein tau pathology in an APP NL-G-F/MAPT P301S mouse model of Alzheimer's disease. Front Neurosci 2024; 18:1372297. [PMID: 38572146 PMCID: PMC10987964 DOI: 10.3389/fnins.2024.1372297] [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: 01/17/2024] [Accepted: 03/01/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction The study of the pathophysiology study of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. Methods The humanized APPNL-G-F knock-in mouse line was crossed to the PS19 MAPTP301S, over-expression mouse line to create the dual APPNL-G-F/PS19 MAPTP301S line. The resulting pathologies were characterized by immunochemical methods and PCR. Results We now report on a double transgenic APPNL-G-F/PS19 MAPTP301S mouse that at 6 months of age exhibits robust A plaque accumulation, intense MAPT pathology, strong inflammation and extensive neurodegeneration. The presence of A pathology potentiated the other major pathologies, including MAPT pathology, inflammation and neurodegeneration. MAPT pathology neither changed levels of amyloid precursor protein nor potentiated A accumulation. Interestingly, study of immunofluorescence in cleared brains indicates that microglial inflammation was generally stronger in the hippocampus, dentate gyrus and entorhinal cortex, which are regions with predominant MAPT pathology. The APPNL-G-F/MAPTP301S mouse model also showed strong accumulation of N6-methyladenosine (m6A), which was recently shown to be elevated in the AD brain. m6A primarily accumulated in neuronal soma, but also co-localized with a subset of astrocytes and microglia. The accumulation of m6A corresponded with increases in METTL3 and decreases in ALKBH5, which are enzymes that add or remove m6A from mRNA, respectively. Discussion Our understanding of the pathophysiology of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. The APPNL-G-F/MAPTP301S mouse recapitulates many features of AD pathology beginning at 6 months of aging, and thus represents a useful new mouse model for the field.
Collapse
Affiliation(s)
- Lulu Jiang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Rebecca Roberts
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Melissa Wong
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Lushuang Zhang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Chelsea Joy Webber
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Jenna Libera
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Zihan Wang
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Alper Kilci
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Matthew Jenkins
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Alejandro Rondón Ortiz
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Luke Dorrian
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Jingjing Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore, Singapore
| | - Guangxin Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sherif Rashad
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | | | - Sarah Anne Daley
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Peter C. Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Campus for Research Excellence and Technological Enterprise, Singapore, Singapore
| | - Brian Nguyen
- LifeCanvas Technologies, Cambridge, MA, United States
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Benjamin Wolozin
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| |
Collapse
|
43
|
Mohapatra O, Gopu M, Ashraf R, Easo George J, Patil S, Mukherjee R, Kumar S, Mampallil D. Spheroids formation in large drops suspended in superhydrophobic paper cones. BIOMICROFLUIDICS 2024; 18:024107. [PMID: 38606014 PMCID: PMC11006428 DOI: 10.1063/5.0197807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
The utilization of 3D cell culture for spheroid formation holds significant implications in cancer research, contributing to a fundamental understanding of the disease and aiding drug development. Conventional methods such as the hanging drop technique and other alternatives encounter limitations due to smaller drop volumes, leading to nutrient starvation and restricted culture duration. In this study, we present a straightforward approach to creating superhydrophobic paper cones capable of accommodating large volumes of culture media drops. These paper cones have sterility, autoclavability, and bacterial repellent properties. Leveraging these attributes, we successfully generate large spheroids of ovarian cancer cells and, as a proof of concept, conduct drug screening to assess the impact of carboplatin. Thus, our method enables the preparation of flexible superhydrophobic surfaces for laboratory applications in an expeditious manner, exemplified here through spheroid formation and drug screening demonstrations.
Collapse
Affiliation(s)
- Omkar Mohapatra
- Department of Physics, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Maheshwar Gopu
- Department of Physics, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Rahail Ashraf
- Department of Biology, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Jijo Easo George
- Department of Physics, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Saniya Patil
- Department of Biology, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Raju Mukherjee
- Department of Biology, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Sanjay Kumar
- Department of Biology, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| | - Dileep Mampallil
- Department of Physics, Indian Institute of Science Education and Research Tirupati, Mangalam P.O., 517507 Tirupati, AP, India
| |
Collapse
|
44
|
Kourti M, Metaxas A. A systematic review and meta-analysis of tau phosphorylation in mouse models of familial Alzheimer's disease. Neurobiol Dis 2024; 192:106427. [PMID: 38307366 DOI: 10.1016/j.nbd.2024.106427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Transgenic models of familial Alzheimer's disease (AD) serve as valuable tools for probing the molecular mechanisms associated with amyloid-beta (Aβ)-induced pathology. In this meta-analysis, we sought to evaluate levels of phosphorylated tau (p-tau) and explore potential age-related variations in tau hyperphosphorylation, within mouse models of AD. The PubMed and Scopus databases were searched for studies measuring soluble p-tau in 5xFAD, APPswe/PSEN1de9, J20 and APP23 mice. Data were extracted and analyzed using standardized procedures. For the 5xFAD model, the search yielded 36 studies eligible for meta-analysis. Levels of p-tau were higher in 5xFAD mice relative to control, a difference that was evident in both the carboxy-terminal (CT) and proline-rich (PR) domains of tau. Age negatively moderated the relationship between genotype and CT phosphorylated tau in studies using hybrid mice, female mice, and preparations from the neocortex. For the APPswe/PSEN1de9 model, the search yielded 27 studies. Analysis showed tau hyperphosphorylation in transgenic vs. control animals, evident in both the CT and PR regions of tau. Age positively moderated the relationship between genotype and PR domain phosphorylated tau in the neocortex of APPswe/PSEN1de9 mice. A meta-analysis was not performed for the J20 and APP23 models, due to the limited number of studies measuring p-tau levels in these mice (<10 studies). Although tau is hyperphosphorylated in both 5xFAD and APPswe/PSEN1de9 mice, the effects of ageing on p-tau are contingent upon the model being examined. These observations emphasize the importance of tailoring model selection to the appropriate disease stage when considering the relationship between Aβ and tau, and suggest that there are optimal intervention points for the administration of both anti-amyloid and anti-tau therapies.
Collapse
Affiliation(s)
- Malamati Kourti
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Centre, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus.
| | - Athanasios Metaxas
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
45
|
Pádua MS, Guil-Guerrero JL, Prates JAM, Lopes PA. Insights on the Use of Transgenic Mice Models in Alzheimer's Disease Research. Int J Mol Sci 2024; 25:2805. [PMID: 38474051 DOI: 10.3390/ijms25052805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, presents a significant global health challenge with no known cure to date. Central to our understanding of AD pathogenesis is the β-amyloid cascade hypothesis, which underlies drug research and discovery efforts. Despite extensive studies, no animal models of AD have completely validated this hypothesis. Effective AD models are essential for accurately replicating key pathological features of the disease, notably the formation of β-amyloid plaques and neurofibrillary tangles. These pathological markers are primarily driven by mutations in the amyloid precursor protein (APP) and presenilin 1 (PS1) genes in familial AD (FAD) and by tau protein mutations for the tangle pathology. Transgenic mice models have been instrumental in AD research, heavily relying on the overexpression of mutated APP genes to simulate disease conditions. However, these models do not entirely replicate the human condition of AD. This review aims to provide a comprehensive evaluation of the historical and ongoing research efforts in AD, particularly through the use of transgenic mice models. It is focused on the benefits gathered from these transgenic mice models in understanding β-amyloid toxicity and the broader biological underpinnings of AD. Additionally, the review critically assesses the application of these models in the preclinical testing of new therapeutic interventions, highlighting the gap between animal models and human clinical realities. This analysis underscores the need for refinement in AD research methodologies to bridge this gap and enhance the translational value of preclinical studies.
Collapse
Affiliation(s)
- Mafalda Soares Pádua
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| | - José L Guil-Guerrero
- Departamento de Tecnología de Alimentos, Universidad de Almería, 04120 Almería, Spain
| | - José A M Prates
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| | - Paula Alexandra Lopes
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisbon, Portugal
| |
Collapse
|
46
|
Ochi S, Yamada K, Saito T, Saido TC, Iinuma M, Azuma K, Kubo KY. Effects of early tooth loss on chronic stress and progression of neuropathogenesis of Alzheimer's disease in adult Alzheimer's model AppNL-G-F mice. Front Aging Neurosci 2024; 16:1361847. [PMID: 38469162 PMCID: PMC10925668 DOI: 10.3389/fnagi.2024.1361847] [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: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Alzheimer's disease (AD), the most common neurodegenerative disease, is characterized by accumulated amyloid-β (Aβ) plaques, aggregated phosphorylated tau protein, gliosis-associated neuroinflammation, synaptic dysfunction, and cognitive impairment. Many cohort studies indicate that tooth loss is a risk factor for AD. The detailed mechanisms underlying the association between AD and tooth loss, however, are not yet fully understood. Methods We explored the involvement of early tooth loss in the neuropathogenesis of the adult AppNL-G-F mouse AD model. The maxillary molars were extracted bilaterally in 1-month-old male mice soon after tooth eruption. Results Plasma corticosterone levels were increased and spatial learning memory was impaired in these mice at 6 months of age. The cerebral cortex and hippocampus of AD mice with extracted teeth showed an increased accumulation of Aβ plaques and phosphorylated tau proteins, and increased secretion of the proinflammatory cytokines, including interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α), accompanied by an increased number of microglia and astrocytes, and decreased synaptophysin expression. AD mice with extracted teeth also had a shorter lifespan than the control mice. Discussion These findings revealed that long-term tooth loss is a chronic stressor, activating the recruitment of microglia and astrocytes; exacerbating neuroinflammation, Aβ deposition, phosphorylated tau accumulation, and synaptic dysfunction; and leading to spatial learning and memory impairments in AD model mice.
Collapse
Affiliation(s)
- Suzuko Ochi
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Mizuho, Japan
| | - Kumiko Yamada
- Department of Health and Nutrition, Faculty of Health Science, Nagoya Women's University, Nagoya, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Mitsuo Iinuma
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Mizuho, Japan
| | - Kagaku Azuma
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kin-Ya Kubo
- Graduate School of Human Life Science, Nagoya Women's University, Nagoya, Japan
| |
Collapse
|
47
|
Kim YK, Jo D, Arjunan A, Ryu Y, Lim YH, Choi SY, Kim HK, Song J. Identification of IGF-1 Effects on White Adipose Tissue and Hippocampus in Alzheimer's Disease Mice via Transcriptomic and Cellular Analysis. Int J Mol Sci 2024; 25:2567. [PMID: 38473814 DOI: 10.3390/ijms25052567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) stands as the most prevalent neurodegenerative disorder, characterized by a multitude of pathological manifestations, prominently marked by the aggregation of amyloid beta. Recent investigations have revealed a compelling association between excessive adiposity and glial activation, further correlating with cognitive impairments. Additionally, alterations in levels of insulin-like growth factor 1 (IGF-1) have been reported in individuals with metabolic conditions accompanied by memory dysfunction. Hence, our research endeavors to comprehensively explore the impact of IGF-1 on the hippocampus and adipose tissue in the context of Alzheimer's disease. To address this, we have conducted an in-depth analysis utilizing APP/PS2 transgenic mice, recognized as a well-established mouse model for Alzheimer's disease. Upon administering IGF-1 injections to the APP/PS2 mice, we observed notable alterations in their behavioral patterns, prompting us to undertake a comprehensive transcriptomic analysis of both the hippocampal and adipose tissues. Our data unveiled significant modifications in the functional profiles of these tissues. Specifically, in the hippocampus, we identified changes associated with synaptic activity and neuroinflammation. Concurrently, the adipose tissue displayed shifts in processes related to fat browning and cell death signaling. In addition to these findings, our analysis enabled the identification of a collection of long non-coding RNAs and circular RNAs that exhibited significant changes in expression subsequent to the administration of IGF-1 injections. Furthermore, we endeavored to predict the potential roles of these identified RNA molecules within the context of our study. In summary, our study offers valuable transcriptome data for hippocampal and adipose tissues within an Alzheimer's disease model and posits a significant role for IGF-1 within both the hippocampus and adipose tissue.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Danbi Jo
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Yeongseo Ryu
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Yeong-Hwan Lim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Seo Yoon Choi
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Hee Kyung Kim
- Department of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| | - Juhyun Song
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanamdo, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanamdo, Republic of Korea
| |
Collapse
|
48
|
Daily KP, Badr A, Eltobgy M, Estfanous S, Whitham O, Tan MH, Carafice C, Krause K, McNamara A, Hamilton K, Houle S, Gupta S, Gupta GA, Madhu S, Fitzgerald J, Saadey AA, Laster B, Yan P, Webb A, Zhang X, Pietrzak M, Kokiko-Cochran ON, Ghoneim HE, Amer AO. DNA hypomethylation promotes the expression of CASPASE-4 which exacerbates inflammation and amyloid-β deposition in Alzheimer's disease. Alzheimers Res Ther 2024; 16:29. [PMID: 38326859 PMCID: PMC10851453 DOI: 10.1186/s13195-024-01390-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 01/09/2024] [Indexed: 02/09/2024]
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death in the USA. It is established that neuroinflammation contributes to the synaptic loss, neuronal death, and symptomatic decline of AD patients. Accumulating evidence suggests a critical role for microglia, innate immune phagocytes of the brain. For instance, microglia release pro-inflammatory products such as IL-1β which is highly implicated in AD pathobiology. The mechanisms underlying the transition of microglia to proinflammatory promoters of AD remain largely unknown. To address this gap, we performed reduced representation bisulfite sequencing (RRBS) to profile global DNA methylation changes in human AD brains compared to no disease controls. We identified differential DNA methylation of CASPASE-4 (CASP4), which when expressed promotes the generation of IL-1β and is predominantly expressed in immune cells. DNA upstream of the CASP4 transcription start site was hypomethylated in human AD brains, which was correlated with increased expression of CASP4. Furthermore, microglia from a mouse model of AD (5xFAD) express increased levels of CASP4 compared to wild-type (WT) mice. To study the role of CASP4 in AD, we developed a novel mouse model of AD lacking the mouse ortholog of CASP4 and CASP11, which is encoded by mouse Caspase-4 (5xFAD/Casp4-/-). The expression of CASP11 was associated with increased accumulation of pathologic protein aggregate amyloid-β (Aβ) and increased microglial production of IL-1β in 5xFAD mice. Utilizing RNA-sequencing, we determined that CASP11 promotes unique transcriptomic phenotypes in 5xFAD mouse brains, including alterations of neuroinflammatory and chemokine signaling pathways. Notably, in vitro, CASP11 promoted generation of IL-1β from macrophages in response to cytosolic Aβ through cleavage of downstream effector Gasdermin D (GSDMD). Therefore, here we unravel the role for CASP11 and GSDMD in the generation of IL-1β in response to Aβ and the progression of pathologic inflammation in AD. Overall, our results demonstrate that overexpression of CASP4 due to differential DNA methylation in AD microglia contributes to the progression of AD pathobiology. Thus, we identify CASP4 as a potential target for immunotherapies for the treatment and prevention of AD.
Collapse
Affiliation(s)
- Kylene P Daily
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Asmaa Badr
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Clinical Pathology Department, College of Medicine, Mansoura University, Mansoura, Egypt
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Owen Whitham
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Michelle H Tan
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Andrew McNamara
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Samuel Houle
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Spandan Gupta
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Gauruv A Gupta
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Shruthi Madhu
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Julie Fitzgerald
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Abbey A Saadey
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Brooke Laster
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Pearlly Yan
- Genomics Shared Resource, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Xiaoli Zhang
- Center for Biostatistics, Ohio State University, Columbus, OH, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | | | - Hazem E Ghoneim
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
- Pelotonia Institute for Immuno-Oncology, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Amal O Amer
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, The Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
49
|
Papazoglou A, Henseler C, Weickhardt S, Teipelke J, Papazoglou P, Daubner J, Schiffer T, Krings D, Broich K, Hescheler J, Sachinidis A, Ehninger D, Scholl C, Haenisch B, Weiergräber M. Sex- and region-specific cortical and hippocampal whole genome transcriptome profiles from control and APP/PS1 Alzheimer's disease mice. PLoS One 2024; 19:e0296959. [PMID: 38324617 PMCID: PMC10849391 DOI: 10.1371/journal.pone.0296959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024] Open
Abstract
A variety of Alzheimer's disease (AD) mouse models has been established and characterized within the last decades. To get an integrative view of the sophisticated etiopathogenesis of AD, whole genome transcriptome studies turned out to be indispensable. Here we carried out microarray data collection based on RNA extracted from the retrosplenial cortex and hippocampus of age-matched, eight months old male and female APP/PS1 AD mice and control animals to perform sex- and brain region specific analysis of transcriptome profiles. The results of our studies reveal novel, detailed insight into differentially expressed signature genes and related fold changes in the individual APP/PS1 subgroups. Gene ontology and Venn analysis unmasked that intersectional, upregulated genes were predominantly involved in, e.g., activation of microglial, astrocytic and neutrophilic cells, innate immune response/immune effector response, neuroinflammation, phagosome/proteasome activation, and synaptic transmission. The number of (intersectional) downregulated genes was substantially less in the different subgroups and related GO categories included, e.g., the synaptic vesicle docking/fusion machinery, synaptic transmission, rRNA processing, ubiquitination, proteasome degradation, histone modification and cellular senescence. Importantly, this is the first study to systematically unravel sex- and brain region-specific transcriptome fingerprints/signature genes in APP/PS1 mice. The latter will be of central relevance in future preclinical and clinical AD related studies, biomarker characterization and personalized medicinal approaches.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Sandra Weickhardt
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Jenni Teipelke
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Panagiota Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Teresa Schiffer
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Damian Krings
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Jürgen Hescheler
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Dan Ehninger
- Translational Biogerontology, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
| | - Catharina Scholl
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Bonn, Germany
- Center for Translational Medicine, Medical Faculty, University of Bonn, Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Bonn, Germany
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
50
|
Pagnon de la Vega M, Syvänen S, Giedraitis V, Hooley M, Konstantinidis E, Meier SR, Rokka J, Eriksson J, Aguilar X, Spires-Jones TL, Lannfelt L, Nilsson LNG, Erlandsson A, Hultqvist G, Ingelsson M, Sehlin D. Altered amyloid-β structure markedly reduces gliosis in the brain of mice harboring the Uppsala APP deletion. Acta Neuropathol Commun 2024; 12:22. [PMID: 38317196 PMCID: PMC10845526 DOI: 10.1186/s40478-024-01734-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: 10/17/2023] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
Deposition of amyloid beta (Aβ) into plaques is a major hallmark of Alzheimer's disease (AD). Different amyloid precursor protein (APP) mutations cause early-onset AD by altering the production or aggregation properties of Aβ. We recently identified the Uppsala APP mutation (APPUpp), which causes Aβ pathology by a triple mechanism: increased β-secretase and altered α-secretase APP cleavage, leading to increased formation of a unique Aβ conformer that rapidly aggregates and deposits in the brain. The aim of this study was to further explore the effects of APPUpp in a transgenic mouse model (tg-UppSwe), expressing human APP with the APPUpp mutation together with the APPSwe mutation. Aβ pathology was studied in tg-UppSwe brains at different ages, using ELISA and immunohistochemistry. In vivo PET imaging with three different PET radioligands was conducted in aged tg-UppSwe mice and two other mouse models; tg-ArcSwe and tg-Swe. Finally, glial responses to Aβ pathology were studied in cell culture models and mouse brain tissue, using ELISA and immunohistochemistry. Tg-UppSwe mice displayed increased β-secretase cleavage and suppressed α-secretase cleavage, resulting in AβUpp42 dominated diffuse plaque pathology appearing from the age of 5-6 months. The γ-secretase cleavage was not affected. Contrary to tg-ArcSwe and tg-Swe mice, tg-UppSwe mice were [11C]PiB-PET negative. Antibody-based PET with the 3D6 ligand visualized Aβ pathology in all models, whereas the Aβ protofibril selective mAb158 ligand did not give any signals in tg-UppSwe mice. Moreover, unlike the other two models, tg-UppSwe mice displayed a very faint glial response to the Aβ pathology. The tg-UppSwe mouse model thus recapitulates several pathological features of the Uppsala APP mutation carriers. The presumed unique structural features of AβUpp42 aggregates were found to affect their interaction with anti-Aβ antibodies and profoundly modify the Aβ-mediated glial response, which may be important aspects to consider for further development of AD therapies.
Collapse
Affiliation(s)
- María Pagnon de la Vega
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Monique Hooley
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Evangelos Konstantinidis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Silvio R Meier
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Johanna Rokka
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Division of Organic Pharmaceutical Chemistry, Uppsala University, Uppsala, Sweden
- PET Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Tara L Spires-Jones
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
- BioArctic AB, Stockholm, Sweden
| | - Lars N G Nilsson
- Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anna Erlandsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | | | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden.
| |
Collapse
|