201
|
d'Errico P, Meyer-Luehmann M. Mechanisms of Pathogenic Tau and Aβ Protein Spreading in Alzheimer's Disease. Front Aging Neurosci 2020; 12:265. [PMID: 33061903 PMCID: PMC7481386 DOI: 10.3389/fnagi.2020.00265] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is pathologically defined by extracellular accumulation of amyloid-β (Aβ) peptides generated by the cleavage of amyloid precursor protein (APP), strings of hyperphosphorylated Tau proteins accumulating inside neurons known as neurofibrillary tangles (NFTs) and neuronal loss. The association between the two hallmarks and cognitive decline has been known since the beginning of the 20th century when the first description of the disease was carried out by Alois Alzheimer. Today, more than 40 million people worldwide are affected by AD that represents the most common cause of dementia and there is still no effective treatment available to cure the disease. In general, the aggregation of Aβ is considered an essential trigger in AD pathogenesis that gives rise to NFTs, neuronal dysfunction and dementia. During the process leading to AD, tau and Aβ first misfold and form aggregates in one brain region, from where they spread to interconnected areas of the brain thereby inducing its gradual morphological and functional deterioration. In this mini-review article, we present an overview of the current literature on the spreading mechanisms of Aβ and tau pathology in AD since a more profound understanding is necessary to design therapeutic approaches aimed at preventing or halting disease progression.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
202
|
Tapp ZM, Kumar JE, Witcher KG, Atluri RR, Velasquez JA, O'Neil SM, Dziabis JE, Bray CE, Sheridan JF, Godbout JP, Kokiko-Cochran ON. Sleep Disruption Exacerbates and Prolongs the Inflammatory Response to Traumatic Brain Injury. J Neurotrauma 2020; 37:1829-1843. [PMID: 32164485 PMCID: PMC7404833 DOI: 10.1089/neu.2020.7010] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) alters stress responses, which may influence neuroinflammation and behavioral outcome. Sleep disruption (SD) is an understudied post-injury environmental stressor that directly engages stress-immune pathways. Thus, we predicted that maladaptive changes in the hypothalamic-pituitary-adrenal (HPA) axis after TBI compromise the neuroendocrine response to SD and exacerbate neuroinflammation. To test this, we induced lateral fluid percussion TBI or sham injury in female and male C57BL/6 mice aged 8-10 weeks that were then left undisturbed or exposed to 3 days of transient SD. At 3 days post-injury (DPI) plasma corticosterone (CORT) was reduced in TBI compared with sham mice, indicating altered HPA-mediated stress response to SD. This response was associated with approach-avoid conflict behavior and exaggerated cortical neuroinflammation. Post-injury SD specifically enhanced neutrophil trafficking to the injured brain in conjunction with dysregulated aquaporin-4 (AQP4) polarization. Delayed and persistent effects of post-injury SD were determined 4 days after SD concluded at 7 DPI. SD prolonged anxiety-like behavior regardless of injury and was associated with increased cortical Iba1 labeling in both sham and TBI mice. Strikingly, TBI SD mice displayed an increased number of CD45+ cells near the site of injury, enhanced cortical glial fibrillary acidic protein (GFAP) immunolabeling, and persistent expression of Trem2 and Tlr4 7 DPI compared with TBI mice. These results support the hypothesis that post-injury SD alters stress-immune pathways and inflammatory outcomes after TBI. These data provide new insight to the dynamic interplay between TBI, stress, and inflammation.
Collapse
Affiliation(s)
- Zoe M. Tapp
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Julia E. Kumar
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Kristina G. Witcher
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Ravitej R. Atluri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - John A. Velasquez
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Shane M. O'Neil
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Julia E. Dziabis
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Chelsea E. Bray
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - John F. Sheridan
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
- Neurological Institute, Institute for Behavioral Medicine Research (IBMR), The Ohio State University, Columbus, Ohio, USA
| | - Jonathan P. Godbout
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Neurological Institute, Institute for Behavioral Medicine Research (IBMR), The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio, USA
| | - Olga N. Kokiko-Cochran
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Neurological Institute, Institute for Behavioral Medicine Research (IBMR), The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
203
|
Angelopoulou E, Paudel YN, Shaikh MF, Piperi C. Fractalkine (CX3CL1) signaling and neuroinflammation in Parkinson’s disease: Potential clinical and therapeutic implications. Pharmacol Res 2020; 158:104930. [DOI: 10.1016/j.phrs.2020.104930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022]
|
204
|
Germann UA, Alam JJ. P38α MAPK Signaling-A Robust Therapeutic Target for Rab5-Mediated Neurodegenerative Disease. Int J Mol Sci 2020; 21:E5485. [PMID: 32751991 PMCID: PMC7432772 DOI: 10.3390/ijms21155485] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Multifactorial pathologies, involving one or more aggregated protein(s) and neuroinflammation are common in major neurodegenerative diseases, such as Alzheimer's disease and dementia with Lewy bodies. This complexity of multiple pathogenic drivers is one potential explanation for the lack of success or, at best, the partial therapeutic effects, respectively, with approaches that have targeted one specific driver, e.g., amyloid-beta, in Alzheimer's disease. Since the endosome-associated protein Rab5 appears to be a convergence point for many, if not all the most prominent pathogenic drivers, it has emerged as a major therapeutic target for neurodegenerative disease. Further, since the alpha isoform of p38 mitogen-activated protein kinase (p38α) is a major regulator of Rab5 activity and its effectors, a biology that is distinct from the classical nuclear targets of p38 signaling, brain-penetrant selective p38α kinase inhibitors provide the opportunity for significant therapeutic advances in neurogenerative disease through normalizing dysregulated Rab5 activity. In this review, we provide a brief summary of the role of Rab5 in the cell and its association with neurodegenerative disease pathogenesis. We then discuss the connection between Rab5 and p38α and summarize the evidence that through modulating Rab5 activity there are therapeutic opportunities in neurodegenerative diseases for p38α kinase inhibitors.
Collapse
|
205
|
González-Prieto M, Gutiérrez IL, García-Bueno B, Caso JR, Leza JC, Ortega-Hernández A, Gómez-Garre D, Madrigal JLM. Microglial CX3CR1 production increases in Alzheimer's disease and is regulated by noradrenaline. Glia 2020; 69:73-90. [PMID: 32662924 DOI: 10.1002/glia.23885] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
The loss of noradrenergic neurons and subsequent reduction of brain noradrenaline (NA) levels are associated with the progression of Alzheimer's disease (AD). This seems to be due mainly to the ability of NA to reduce the activation of microglial cells. We previously observed that NA induces the production of the chemokine Fractalkine/CX3CL1 in neurons. The activation of microglial CX3CR1, sole receptor for CX3CL1, reduces the activation of microglia, which is known to largely contribute to the neuronal damage characteristic of AD. Therefore, alterations of CX3CR1 production in microglia could translate into the enhancement or inhibition of CX3CL1 anti-inflammatory effects. In order to determine if microglial CX3CR1 production is altered in AD and if NA can control it, CX3CR1 expression and synthesis were analyzed in 5xFAD mice and human AD brain samples. In addition, the effects of NA and its reuptake inhibitor reboxetine were analyzed in microglial cultures and mice respectively. Our results indicate that in AD CX3CR1 production is increased in the brain cortex and that reboxetine administration further increases it and enhances microglial reactivity toward amyloid beta plaques. However, direct administration of NA to primary rat microglia or human HMC3 cells inhibits CX3CR1 production, suggesting that microglia responses to NA may be altered in the absence of CX3CL1-producing neurons or other nonmicroglial external factors.
Collapse
Affiliation(s)
- Marta González-Prieto
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Irene L Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| | - Adriana Ortega-Hernández
- Vascular Biology Laboratory and Flow Cytometric Unit, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.,Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Dulcenombre Gómez-Garre
- Vascular Biology Laboratory and Flow Cytometric Unit, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.,Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - José L M Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital, Madrid, Spain
| |
Collapse
|
206
|
Liu G, Fiock KL, Levites Y, Golde TE, Hefti MM, Lee G. Fyn depletion ameliorates tau P301L-induced neuropathology. Acta Neuropathol Commun 2020; 8:108. [PMID: 32665013 PMCID: PMC7362472 DOI: 10.1186/s40478-020-00979-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
The Src family non-receptor tyrosine kinase Fyn has been implicated in neurodegeneration of Alzheimer's disease through interaction with amyloid β (Aβ). However, the role of Fyn in the pathogenesis of primary tauopathies such as FTDP-17, where Aβ plaques are absent, is poorly understood. In the current study, we used AAV2/8 vectors to deliver tauP301L to the brains of WT and Fyn KO mice, generating somatic transgenic tauopathy models with the presence or absence of Fyn. Although both genotypes developed tau pathology, Fyn KO developed fewer neurofibrillary tangles on Bielschowsky and Thioflavin S stained sections and showed lower levels of phosphorylated tau. In addition, tauP301L-induced behavior abnormalities and depletion of synaptic proteins were not observed in the Fyn KO model. Our work provides evidence for Fyn being a critical protein in the disease pathogenesis of FTDP-17.
Collapse
Affiliation(s)
- Guanghao Liu
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Kimberly L. Fiock
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Yona Levites
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Todd E. Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Marco M. Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Gloria Lee
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 500 Newton Road, ML B191, Iowa City, IA 52242 USA
| |
Collapse
|
207
|
Liu J, Li H, Gong T, Chen W, Mao S, Kong Y, Yu J, Sun J. Anti-neuroinflammatory Effect of Short-Chain Fatty Acid Acetate against Alzheimer's Disease via Upregulating GPR41 and Inhibiting ERK/JNK/NF-κB. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7152-7161. [PMID: 32583667 DOI: 10.1021/acs.jafc.0c02807] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Alzheimer's disease (AD) is a high-incidence neurodegenerative disease in the elderly. Acetate (Ace) is a short-chain fatty acid (SCFA) with neuroprotective activity. The purpose of this study was to investigate the effects and its possible mechanisms of SCFA Ace on AD. A male APP/PS1 transgenic mouse was given intragastric administration Ace for 4 weeks. Cognitive function and microglia activation in mice were assessed. Furthermore, Ace pretreated amyloid-β (Aβ)-induced BV2 microglia, and the levels of CD11b, COX-2, and G-protein-coupled receptor 41 (GPR41) and phosphorylation of ERK, JNK, and NF-κB p65 were determined. Our results revealed that Ace significantly attenuated the cognitive impairment and decreased the CD11b level in the APP/PS1 mice. Moreover, Ace inhibited the phosphorylation of NF-κB p65, ERK, and JNK and decreased the levels of COX-2 and interleukin 1β in the Aβ-stimulated BV2 microglia. Finally, Ace increased the GPR41 level in the Aβ-stimulated BV2 cells. The finding indicated that Ace exerted antineuroinflammatory effects via the upregulation of GPR41 and suppression of the ERK/JNK/NF-κB pathway, which might provide an alternative therapy strategy of AD.
Collapse
Affiliation(s)
- Jiaming Liu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Haijun Li
- Department of Neurology, Taizhou Second People's Hospital, Taizhou, Zhejiang 317000, People's Republic of China
| | - Tianyu Gong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Wenyang Chen
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Shiyin Mao
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yu Kong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jiaheng Yu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jing Sun
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| |
Collapse
|
208
|
Zhu K, Pieber M, Han J, Blomgren K, Zhang XM, Harris RA, Lund H. Absence of microglia or presence of peripherally-derived macrophages does not affect tau pathology in young or old hTau mice. Glia 2020; 68:1466-1478. [PMID: 32039516 DOI: 10.1002/glia.23794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 01/02/2023]
Abstract
Microglia are implicated in the pathophysiology of several neurodegenerative disorders, including Alzheimer's disease. While the role of microglia and peripheral macrophages in regulating amyloid beta pathology has been well characterized, the impact of these distinct cell subsets on tau pathology remains poorly understood. We and others have recently demonstrated that monocytes can engraft the brain and give rise to long-lived parenchymal macrophages, even under nonpathological conditions. We undertook the current study to investigate the regulation of tau pathology by microglia and peripheral macrophages using hTau transgenic mice, which do not exhibit microglial activation/pathology or macrophage engraftment. To assess the direct impact of microglia on tau pathology we developed a protocol for long-term microglial depletion in Cx3cr1CreER R26DTA mice and crossed them with hTau mice. We then depleted microglia up to 3 months in both young and old mice, but no net change in forebrain soluble oligomeric tau or total or phosphorylated levels of aggregated tau was recorded. To investigate the consequence of peripherally-derived parenchymal macrophages on tau aggregation we partially repopulated the hTau microglial pool with peripheral macrophages, but this also did not affect levels of tau oligomers or insoluble aggregates. Our study questions the direct involvement of microglia or peripheral macrophages in the development of tau pathology in the hTau model.
Collapse
Affiliation(s)
- Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Melanie Pieber
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Harald Lund
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Anesthesiology, University of California San Diego, La Jolla, California
| |
Collapse
|
209
|
Wijeyekoon RS, Kronenberg-Versteeg D, Scott KM, Hayat S, Kuan WL, Evans JR, Breen DP, Cummins G, Jones JL, Clatworthy MR, Floto RA, Barker RA, Williams-Gray CH. Peripheral innate immune and bacterial signals relate to clinical heterogeneity in Parkinson's disease. Brain Behav Immun 2020; 87:473-488. [PMID: 32006615 PMCID: PMC7613010 DOI: 10.1016/j.bbi.2020.01.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 01/09/2023] Open
Abstract
The innate immune system is implicated in Parkinson's disease (PD), but peripheral in-vivo clinical evidence of the components and driving mechanisms involved and their relationship with clinical heterogeneity and progression to dementia remain poorly explored. We examined changes in peripheral innate immune-related markers in PD cases (n = 41) stratified according to risk of developing early dementia. 'Higher Risk'(HR) (n = 23) and 'Lower Risk' (LR) (n = 18) groups were defined according to neuropsychological predictors and MAPT H1/H2 genotype, and compared to age, gender and genotype-matched controls. Monocyte subsets and expression of key surface markers were measured using flow cytometry. Serum markers including alpha-synuclein, inflammasome-related caspase-1 and bacterial translocation-related endotoxin were measured using quantitative immuno-based assays. Specific markers were further investigated using monocyte assays and validated in plasma samples from a larger incident PD cohort (n = 95). We found that classical monocyte frequency was elevated in PD cases compared to controls, driven predominantly by the HR group, in whom Toll-Like Receptor (TLR)4+ monocytes and monocyte Triggering Receptor Expressed on Myeloid cells-2 (TREM2) expression were also increased. Monocyte Human Leukocyte Antigen (HLA)-DR expression correlated with clinical variables, with lower levels associated with worse cognitive/motor performance. Notably, monocyte changes were accompanied by elevated serum bacterial endotoxin, again predominantly in the HR group. Serum alpha-synuclein and inflammasome-related caspase-1 were decreased in PD cases compared to controls regardless of group, with decreased monocyte alpha-synuclein secretion in HR cases. Further, alpha-synuclein and caspase-1 correlated positively in serum and monocyte lysates, and in plasma from the larger cohort, though no associations were seen with baseline or 36-month longitudinal clinical data. Principal Components Analysis of all monocyte and significant serum markers indicated 3 major components. Component 1 (alpha-synuclein, caspase-1, TLR2+ monocytes) differentiated PD cases and controls in both groups, while Component 2 (endotoxin, monocyte TREM2, alpha-synuclein) did so predominantly in the HR group. Component 3 (classical monocytes, alpha-synuclein) also differentiated cases and controls overall in both groups. These findings demonstrate that systemic innate immune changes are present in PD and are greatest in those at higher risk of rapid progression to dementia. Markers associated with PD per-se (alpha-synuclein, caspase-1), differ from those related to cognitive progression and clinical heterogeneity (endotoxin, TREM2, TLR4, classical monocytes, HLA-DR), with mechanistic and therapeutic implications. Alpha-synuclein and caspase-1 are associated, suggesting inflammasome involvement common to all PD, while bacterial translocation associated changes may contribute towards progression to Parkinson's dementia. Additionally, HLA-DR-associated variations in antigen presentation/clearance may modulate existing clinical disease.
Collapse
Affiliation(s)
- Ruwani S. Wijeyekoon
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Corresponding Author;
| | | | - Kirsten M. Scott
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Shaista Hayat
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Wei-Li Kuan
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Jonathan R. Evans
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Nottingham University Hospital NHS Trust, Nottingham, UK
| | - David P. Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49, Little France Crescent, Edinburgh, EH16 4SB, UK,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Chancellor’s Building, 49, Little France Crescent, Edinburgh, EH16 4SB, UK,Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, 9, Little France Road, Edinburgh BioQuarter, Edinburgh, EH16 4UX, UK
| | - Gemma Cummins
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Joanne L. Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - R. Andres Floto
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Roger A. Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Caroline H. Williams-Gray
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| |
Collapse
|
210
|
Sinyor B, Mineo J, Ochner C. Alzheimer's Disease, Inflammation, and the Role of Antioxidants. J Alzheimers Dis Rep 2020; 4:175-183. [PMID: 32715278 PMCID: PMC7369138 DOI: 10.3233/adr-200171] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
The World Health Organization refers to Alzheimer's disease (AD) as a global health priority. As the average age of the world's population is increasing, so too is the rate of AD. There are an estimated 47 million people globally who have been diagnosed with AD dementia, and researchers have yet to figure out the root cause. All misfolded aggregate proteins that are involved in neurodegenerative disorders (amyloid-β, Huntington's tau, α-synuclein) induce oxidative stress. It is that oxidative stress that leads to inflammation and, in conjunction with amyloid protein and tau hyperphosphorylation, progresses to and exacerbates AD. The consumption of antioxidants and nutrients, specifically vitamin E, caffeine, and turmeric, may slow the progression of AD and can be found in a wide variety of dietary foods. This review explores the role of inflammation on AD, the antioxidants that can potentially combat it, and future directions of how the treatment of the disease can be better understood.
Collapse
Affiliation(s)
- Benjamin Sinyor
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| | - Jocelyn Mineo
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| | - Christopher Ochner
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| |
Collapse
|
211
|
Ho CY, Lin YT, Chen HH, Ho WY, Sun GC, Hsiao M, Lu PJ, Cheng PW, Tseng CJ. CX3CR1-microglia mediates neuroinflammation and blood pressure regulation in the nucleus tractus solitarii of fructose-induced hypertensive rats. J Neuroinflammation 2020; 17:185. [PMID: 32532282 PMCID: PMC7291459 DOI: 10.1186/s12974-020-01857-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inflammation is a common pathophysiological trait found in both hypertension and cardiac vascular disease. Recent evidence indicates that fractalkine (FKN) and its receptor CX3CR1 have been linked to inflammatory response in the brain of hypertensive animal models. Here, we investigated the role of CX3CR1-microglia in nitric oxide (NO) generation during chronic inflammation and systemic blood pressure recovery in the nucleus tractus solitarii (NTS). METHODS The hypertensive rat model was used to study the role of CX3CR1-microglia in NTS inflammation following hypertension induction by oral administration of 10% fructose water. The systolic blood pressure was measured by tail-cuff method of non-invasive blood pressure. The CX3CR1 inhibitor AZD8797 was administered intracerebroventricularly (ICV) in the fructose-induced hypertensive rat. Using immunoblotting, we studied the nitric oxide synthase signaling pathway, NO concentration, and the levels of FKN and CX3CR1, and pro-inflammatory cytokines were analyzed by immunohistochemistry staining. RESULTS The level of pro-inflammatory cytokines IL-1β, IL-6, TNF-α, FKN, and CX3CR1 were elevated two weeks after fructose feeding. AZD8797 inhibited CX3CR1-microglia, which improved the regulation of systemic blood pressure and NO generation in the NTS. We also found that IL-1β, IL-6, and TNF-α levels were recovered by AZD8797 addition. CONCLUSION We conclude that CX3CR1-microglia represses the nNOS signaling pathway and promotes chronic inflammation in fructose-induced hypertension. Collectively, our results reveal the role of chemokines such as IL-1β, IL-6, and TNF-α in NTS neuroinflammation with the involvement of FKN and CX3CR1.
Collapse
Affiliation(s)
- Chiu-Yi Ho
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Yu-Te Lin
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Section of Neurology, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan
- Center for Geriatrics and Gerontology, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan
- Shu-Zen Junior College of Medicine and Management, Kaohsiung, 82144, Taiwan
| | - Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan
| | - Wen-Yu Ho
- Division of General Internal Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, National Cheng-Kung University, Tainan, 70101, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
| | - Ching-Jiunn Tseng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81300, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
212
|
Kleineidam L, Chouraki V, Próchnicki T, van der Lee SJ, Madrid-Márquez L, Wagner-Thelen H, Karaca I, Weinhold L, Wolfsgruber S, Boland A, Martino Adami PV, Lewczuk P, Popp J, Brosseron F, Jansen IE, Hulsman M, Kornhuber J, Peters O, Berr C, Heun R, Frölich L, Tzourio C, Dartigues JF, Hüll M, Espinosa A, Hernández I, de Rojas I, Orellana A, Valero S, Stringa N, van Schoor NM, Huisman M, Scheltens P, Rüther E, Deleuze JF, Wiltfang J, Tarraga L, Schmid M, Scherer M, Riedel-Heller S, Heneka MT, Amouyel P, Jessen F, Boada M, Maier W, Schneider A, González-Pérez A, van der Flier WM, Wagner M, Lambert JC, Holstege H, Sáez ME, Latz E, Ruiz A, Ramirez A. PLCG2 protective variant p.P522R modulates tau pathology and disease progression in patients with mild cognitive impairment. Acta Neuropathol 2020; 139:1025-1044. [PMID: 32166339 PMCID: PMC7244617 DOI: 10.1007/s00401-020-02138-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
A rare coding variant (rs72824905, p.P522R) conferring protection against Alzheimer's disease (AD) was identified in the gene encoding the enzyme phospholipase-C-γ2 (PLCG2) that is highly expressed in microglia. To explore the protective nature of this variant, we employed latent process linear mixed models to examine the association of p.P522R with longitudinal cognitive decline in 3595 MCI patients, and in 10,097 individuals from population-based studies. Furthermore, association with CSF levels of pTau181, total tau, and Aβ1-42 was assessed in 1261 MCI patients. We found that MCI patients who carried the p.P522R variant showed a slower rate of cognitive decline compared to non-carriers and that this effect was mediated by lower pTau181 levels in CSF. The effect size of the association of p.P522R with the cognitive decline and pTau181 was similar to that of APOE-ε4, the strongest genetic risk factor for AD. Interestingly, the protective effect of p.P522R was more pronounced in MCI patients with low Aβ1-42 levels suggesting a role of PLCG2 in the response to amyloid pathology. In line with this hypothesis, we observed no protective effect of the PLCG2 variant on the cognitive decline in population-based studies probably due to the lower prevalence of amyloid positivity in these samples compared to MCI patients. Concerning the potential biological underpinnings, we identified a network of co-expressed proteins connecting PLCG2 to APOE and TREM2 using unsupervised co-regulatory network analysis. The network was highly enriched for the complement cascade and genes differentially expressed in disease-associated microglia. Our data show that p.P522R in PLCG2 reduces AD disease progression by mitigating tau pathology in the presence of amyloid pathology and, as a consequence, maintains cognitive function. Targeting the enzyme PLCG2 might provide a new therapeutic approach for treating AD.
Collapse
Affiliation(s)
- Luca Kleineidam
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Vincent Chouraki
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France
- Epidemiology and Public Health Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Tomasz Próchnicki
- Institute of Innate Immunity, University Hospitals Bonn, Bonn, Germany
| | - Sven J van der Lee
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Holger Wagner-Thelen
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Ilker Karaca
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Leonie Weinhold
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany
| | - Steffen Wolfsgruber
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Évry, France
| | - Pamela V Martino Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland
- Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| | - Julius Popp
- Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland
- Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland
| | - Frederic Brosseron
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Iris E Jansen
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marc Hulsman
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Peters
- Department of Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Claudine Berr
- INSERM, University Montpellier, Neuropsychiatry: Epidemiological and Clinical Research, Montpellier, France
| | - Reinhard Heun
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127, Bonn, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Christophe Tzourio
- Inserm, Bordeaux Population Health Research Center, UMR1219, University of Bordeaux, Bordeaux, France
| | - Jean-François Dartigues
- Inserm, Bordeaux Population Health Research Center, UMR1219, University of Bordeaux, Bordeaux, France
| | - Michael Hüll
- Department of Psychiatry and Psychotherapy, Center for Psychiatry, Clinic for Geriatric Psychiatry and Psychotherapy Emmendingen, University of Freiburg, Freiburg, Germany
| | - Ana Espinosa
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
| | - Sergi Valero
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Najada Stringa
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martijn Huisman
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eckart Rüther
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Jean-Francois Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Évry, France
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Lluis Tarraga
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany
| | - Martin Scherer
- Department of Primary Medical Care, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steffi Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Merce Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Wolfgang Maier
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Anja Schneider
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Michael Wagner
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Henne Holstege
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mª Eugenia Sáez
- Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute of Innate Immunity, University Hospitals Bonn, Bonn, Germany
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Centre for Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, Trondheim, Norway
| | - Agustin Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfredo Ramirez
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
213
|
Joly-Amado A, Hunter J, Quadri Z, Zamudio F, Rocha-Rangel PV, Chan D, Kesarwani A, Nash K, Lee DC, Morgan D, Gordon MN, Selenica MLB. CCL2 Overexpression in the Brain Promotes Glial Activation and Accelerates Tau Pathology in a Mouse Model of Tauopathy. Front Immunol 2020; 11:997. [PMID: 32508844 PMCID: PMC7251073 DOI: 10.3389/fimmu.2020.00997] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Innate immune activation is a major contributor to Alzheimer's Disease (AD) pathophysiology, although the mechanisms involved are poorly understood. Chemokine C-C motif ligand (CCL) 2 is produced by neurons and glial cells and is upregulated in the AD brain. Transgene expression of CCL2 in mouse models of amyloidosis produces microglia-induced amyloid β oligomerization, a strong indication of the role of these activation pathways in the amyloidogenic processes of AD. We have previously shown that CCL2 polarizes microglia in wild type mice. However, how CCL2 signaling contributes to tau pathogenesis remains unknown. To address this question, CCL2 was delivered via recombinant adeno-associated virus serotype 9 into both cortex and hippocampus of a mouse model with tau pathology (rTg4510). We report that CCL2 overexpression aggravated tau pathology in rTg4510 as shown by the increase in Gallyas stained neurofibrillary tangles as well as phosphorylated tau-positive inclusions. In addition, biochemical analysis showed a reduction in the levels of detergent-soluble tau species followed by increase in the insoluble fraction, indicating a shift toward larger tau aggregates. Indeed, increased levels of high molecular weight species of phosphorylated tau were found in the mice injected with CCL2. We also report that worsening of tau pathology following CCL2 overexpression was accompanied by a distinct inflammatory response. We report an increase in leukocyte common antigen (CD45) and Cluster of differentiation 68 (CD68) expression in the brain of rTg4510 mice without altering the expression levels of a cell-surface protein Transmembrane Protein 119 (Tmem119) and ionized calcium-binding adaptor molecule 1 (Iba-1) in resident microglia. Furthermore, the analysis of cytokines in brain extract showed a significant increase in interleukin (IL)-6 and CCL3, while CCL5 levels were decreased in CCL2 mice. No changes were observed in IL-1α, IL-1β, TNF-α. IL-4, Vascular endothelial growth factor-VEGF, IL-13 and CCL11. Taken together our data report for the first time that overexpression of CCL2 promotes the increase of pathogenic tau species and is associated with glial neuroinflammatory changes that are deleterious. We propose that these events may contribute to the pathogenesis of Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Aurelie Joly-Amado
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jordan Hunter
- Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Zainuddin Quadri
- Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Frank Zamudio
- Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Patricia V Rocha-Rangel
- Michigan State University, Department of Translational Neuroscience, Grand Rapids, MI, United States
| | - Deanna Chan
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Anisha Kesarwani
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Kevin Nash
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Daniel C Lee
- Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Dave Morgan
- Michigan State University, Department of Translational Neuroscience, Grand Rapids, MI, United States
| | - Marcia N Gordon
- Michigan State University, Department of Translational Neuroscience, Grand Rapids, MI, United States
| | - Maj-Linda B Selenica
- Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States.,Sanders-Brown Center on Aging, Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
214
|
Winter AN, Subbarayan MS, Grimmig B, Weesner JA, Moss L, Peters M, Weeber E, Nash K, Bickford PC. Two forms of CX3CL1 display differential activity and rescue cognitive deficits in CX3CL1 knockout mice. J Neuroinflammation 2020; 17:157. [PMID: 32410624 PMCID: PMC7227354 DOI: 10.1186/s12974-020-01828-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
Background Fractalkine (CX3CL1; FKN) is a chemokine expressed by neurons that mediates communication between neurons and microglia. By regulating microglial activity, CX3CL1 can mitigate the damaging effects of chronic microglial inflammation within the brain, a state that plays a major role in aging and neurodegeneration. CX3CL1 is present in two forms, a full-length membrane-bound form and a soluble cleaved form (sFKN), generated by a disintegrin and metalloproteinase (ADAM) 10 or 17. Levels of sFKN decrease with aging, which could lead to enhanced inflammation, deficits in synaptic remodeling, and subsequent declines in cognition. Recently, the idea that these two forms of CX3CL1 may display differential activities within the CNS has garnered increased attention, but remains unresolved. Methods Here, we assessed the consequences of CX3CL1 knockout (CX3CL1-/-) on cognitive behavior as well as the functional rescue with the two different forms of CX3CL1 in mice. CX3CL1-/- mice were treated with adeno-associated virus (AAV) expressing either green fluorescent protein (GFP), sFKN, or an obligate membrane-bound form of CX3CL1 (mFKN) and then subjected to behavioral testing to assess cognition and motor function. Following behavioral analysis, brains were collected and analyzed for markers of neurogenesis, or prepared for electrophysiology to measure long-term potentiation (LTP) in hippocampal slices. Results CX3CL1−/− mice showed significant deficits in cognitive tasks for long-term memory and spatial learning and memory in addition to demonstrating enhanced basal motor performance. These alterations correlated with deficits in both hippocampal neurogenesis and LTP. Treatment of CX3CL1−/− mice with AAV-sFKN partially corrected changes in both cognitive and motor function and restored neurogenesis and LTP to levels similar to wild-type animals. Treatment with AAV-mFKN partially restored spatial learning and memory in CX3CL1−/− mice, but did not rescue long-term memory, or neurogenesis. Conclusions These results are the first to demonstrate that CX3CL1 knockout causes significant cognitive deficits that can be rescued by treatment with sFKN and only partially rescued with mFKN. This suggests that treatments that restore signaling of soluble forms of CX3CL1 may be a viable therapeutic option for aging and disease.
Collapse
Affiliation(s)
- Aimee N Winter
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Meena S Subbarayan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL, 33620, USA.,Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL, 33620, USA.,Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Jason A Weesner
- Integrated Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Lauren Moss
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Melinda Peters
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Edwin Weeber
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Kevin Nash
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - Paula C Bickford
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, USF Morsani College of Medicine, Tampa, FL, 33620, USA. .,Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA. .,Research Service, James A. Haley Veterans Affairs Hospital, Tampa, FL, 33620, USA.
| |
Collapse
|
215
|
Mattsson-Carlgren N, Andersson E, Janelidze S, Ossenkoppele R, Insel P, Strandberg O, Zetterberg H, Rosen HJ, Rabinovici G, Chai X, Blennow K, Dage JL, Stomrud E, Smith R, Palmqvist S, Hansson O. Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive Tau PET in Alzheimer's disease. SCIENCE ADVANCES 2020; 6:eaaz2387. [PMID: 32426454 PMCID: PMC7159908 DOI: 10.1126/sciadv.aaz2387] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/22/2020] [Indexed: 05/09/2023]
Abstract
The links between β-amyloid (Aβ) and tau in Alzheimer's disease are unclear. Cognitively unimpaired persons with signs of Aβ pathology had increased cerebrospinal fluid (CSF) phosphorylated tau (P-tau181 and P-tau217) and total-tau (T-tau), which increased over time, despite no detection of insoluble tau aggregates [normal Tau positron emission tomography (PET)]. CSF P-tau and T-tau started to increase before the threshold for Amyloid PET positivity, while Tau PET started to increase after Amyloid PET positivity. Effects of Amyloid PET on Tau PET were mediated by CSF P-tau, and high CSF P-tau predicted increased Tau PET rates. Individuals with MAPT mutations and signs of tau deposition (but without Aβ pathology) had normal CSF P-tau levels. In 5xFAD mice, CSF tau increased when Aβ aggregation started. These results show that Aβ pathology may induce changes in soluble tau release and phosphorylation, which is followed by tau aggregation several years later in humans.
Collapse
Affiliation(s)
- Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Emelie Andersson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- VU University Medical Center, Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Philip Insel
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
| | - Howard J. Rosen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Gil Rabinovici
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Xiyun Chai
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
216
|
Walterfang M, Di Biase MA, Cropley VL, Scott AM, O'Keefe G, Velakoulis D, Pathmaraj K, Ackermann U, Pantelis C. Imaging of neuroinflammation in adult Niemann-Pick type C disease: A cross-sectional study. Neurology 2020; 94:e1716-e1725. [PMID: 32209649 DOI: 10.1212/wnl.0000000000009287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 11/05/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that neuroinflammation is a key process in adult Niemann-Pick type C (NPC) disease, we undertook PET scanning utilizing a ligand binding activated microglia on 9 patients and 9 age- and sex-matched controls. METHOD We scanned all participants with the PET radioligand 11C-(R)-PK-11195 and undertook structural MRI to measure gray matter volume and white matter fractional anisotropy (FA). RESULTS We found increased binding of 11C-(R)-PK-11195 in total white matter compared to controls (p < 0.01), but not in gray matter regions, and this did not correlate with illness severity or duration. Gray matter was reduced in the thalamus (p < 0.0001) in patients, who also showed widespread reductions in FA across the brain compared to controls (p < 0.001). A significant correlation between 11C-(R)-PK11195 binding and FA was shown (p = 0.002), driven by the NPC patient group. CONCLUSIONS Our findings suggest that neuroinflammation-particularly in white matter-may underpin some structural and degenerative changes in patients with NPC.
Collapse
Affiliation(s)
- Mark Walterfang
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Maria A Di Biase
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Vanessa L Cropley
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Andrew M Scott
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Graeme O'Keefe
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Dennis Velakoulis
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Kunthi Pathmaraj
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Uwe Ackermann
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia
| | - Christos Pantelis
- From the Neuropsychiatry Unit (M.W., D.V.), Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre (M.W., M.A.D., V.L.C., D.V., C.P.), The University of Melbourne & North Western Mental Health; The Florey Institute of Neuroscience and Mental Health (M.W., C.P.), Department of Psychiatry (M.W., M.A.D., V.L.C., D.V., C.P.), and Centre for Neural Engineering, Department of Electrical and Electronic Engineering (C.P.), The University of Melbourne; Department of Molecular Imaging and Therapy (A.M.S., G.O., K.P., U.A.), Austin Health and The University of Melbourne, Heidelberg; Olivia Newton John Cancer Centre and La Trobe University (A.M.S., G.O., U.A.), Melbourne; and Cooperative Centre for Mental Health Research (C.P.), Carlton, Australia.
| |
Collapse
|
217
|
Binder JL, Chander P, Deretic V, Weick JP, Bhaskar K. Optical induction of autophagy via Transcription factor EB (TFEB) reduces pathological tau in neurons. PLoS One 2020; 15:e0230026. [PMID: 32208437 PMCID: PMC7092971 DOI: 10.1371/journal.pone.0230026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/19/2020] [Indexed: 01/25/2023] Open
Abstract
Pathological accumulation of microtubule associated protein tau in neurons is a major neuropathological hallmark of Alzheimer's disease (AD) and related tauopathies. Several attempts have been made to promote clearance of pathological tau (p-Tau) from neurons. Transcription factor EB (TFEB) has shown to clear p-Tau from neurons via autophagy. However, sustained TFEB activation and autophagy can create burden on cellular bioenergetics and can be deleterious. Here, we modified previously described two-plasmid systems of Light Activated Protein (LAP) from bacterial transcription factor-EL222 and Light Responsive Element (LRE) to encode TFEB. Upon blue-light (465 nm) illumination, the conformation changes in LAP induced LRE-driven expression of TFEB, its nuclear entry, TFEB-mediated expression of autophagy-lysosomal genes and clearance of p-Tau from neuronal cells and AD patient-derived human iPSC-neurons. Turning the blue-light off reversed the expression of TFEB-target genes and attenuated p-Tau clearance. Together, these results suggest that optically regulated TFEB expression unlocks the potential of opto-therapeutics to treat AD and other dementias.
Collapse
Affiliation(s)
- Jessica L. Binder
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
- Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence (CoBRE), University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| |
Collapse
|
218
|
Malamon JS, Kriete A. Erosion of Gene Co-expression Networks Reveal Deregulation of Immune System Processes in Late-Onset Alzheimer's Disease. Front Neurosci 2020; 14:228. [PMID: 32265636 PMCID: PMC7099620 DOI: 10.3389/fnins.2020.00228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
We have applied a novel and integrative analysis framework for next-generation sequencing (NGS) data to 503 human subjects provided by the Religious Orders Study and Memory and Aging Project (ROSMAP) to examine changes in transcriptomic organization and common variants in association with late-onset Alzheimer's disease (LOAD). Our framework identified seven reproducible, co-regulated modules after quality control (QC), clinical segregation, preservation filtering, and functional ontology analysis. These modules were specifically enriched in several innate and adaptive immune system processes, the synaptic vesicle cycle, and Hippo signaling. Topological and functional erosion of these modules due to shedding of genes and loss of in-module connectivity was diagnostic of disease progression. Perturbation analysis revealed that only 1% of eQTLs overlapped genes participating in these co-regulated modules. Common variants nevertheless identified components of the immune systems like human leukocyte antigen (HLA) complex and microtubule-associated protein tau (MAPT) regions in association with LOAD. Our results implicate microglial function, adaptive immune response, and the structural degeneration of neurons as contributors to the transcriptional deregulation observed along with common genetic variants in the progression of LOAD.
Collapse
Affiliation(s)
- John Stephen Malamon
- Bossone Research Center, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Andres Kriete
- Bossone Research Center, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
219
|
Anwar S, Rivest S. Alzheimer's disease: microglia targets and their modulation to promote amyloid phagocytosis and mitigate neuroinflammation. Expert Opin Ther Targets 2020; 24:331-344. [PMID: 32129117 DOI: 10.1080/14728222.2020.1738391] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Introduction: Despite the revolutionary progress in neurodegenerative disease research, there is no cure for Alzheimer's disease (AD). This is a chronic progressive neurodegenerative disease affecting aged people and is associated with chronic neuroinflammation and amyloid-beta (Aβ) deposition in the brain parenchyma. Microglia, the resident myeloid cells in the central nervous system, are critically involved in the pathogenesis of AD and have emerged as a potential therapeutic target for treating or preventing AD. The failure of microglia to keep up with persistent amyloid-beta development along with secretion of inflammatory cytokines is detrimental to neurons and favors Aβ accumulation.Areas covered: This review illuminates the latest research that is focused on molecules and their intracellular targets that promote microglial phagocytosis and /or its polarization to an anti-inflammatory state.Expert opinion: A robust inflammatory response of microglia is not necessary to improve their efficiency of Aβ clearance. The challenge is to master inflammatory/anti-inflammatory phenotypes depending on the stage of AD and to maintain efficient responses to remove Aβ. Therefore, promoting microglia phagocytosis without a persistent excessive inflammatory response could be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Shehata Anwar
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada.,Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
220
|
Barron MR, Gartlon J, Dawson LA, Atkinson PJ, Pardon MC. Increasing Tau 4R Tau Levels Exacerbates Hippocampal Tau Hyperphosphorylation in the hTau Model of Tauopathy but Also Tau Dephosphorylation Following Acute Systemic Inflammation. Front Immunol 2020; 11:293. [PMID: 32194553 PMCID: PMC7066213 DOI: 10.3389/fimmu.2020.00293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Inflammation is considered a mechanistic driver of Alzheimer's disease, thought to increase tau phosphorylation, the first step to the formation of neurofibrillary tangles (NFTs). To further understand how inflammation impacts the development of tau pathology, we used (hTau) mice, which express all six, non-mutated, human tau isoforms, but with an altered ratio of tau isoforms favoring 3R tau due to the concomitant loss of murine tau (mTau) that is predominantly 4R. Such an imbalance pattern has been related to susceptibility to NFTs formation, but whether or not this also affects susceptibility to systemic inflammation and related changes in tau phosphorylation is not known. To reduce the predominance of 3R tau by increasing 4R tau availability, we bred hTau mice on a heterozygous mTau background and compared the impact of systemic inflammation induced by lipopolysaccharide (LPS) in hTau mice hetero- or homozygous mTau knockout. Three-month-old male wild-type (Wt), mTau+/-, mTau-/-, hTau/mTau+/-, and hTau/mTau-/- mice were administered 100, 250, or 330 μg/kg of LPS or its vehicle phosphate buffer saline (PBS) [intravenously (i.v.), n = 8-9/group]. Sickness behavior, reflected by behavioral suppression in the spontaneous alternation task, hippocampal tau phosphorylation, measured by western immunoblotting, and circulating cytokine levels were quantified 4 h after LPS administration. The persistence of the LPS effects (250 μg/kg) on these measures, and food burrowing behavior, was assessed at 24 h post-inoculation in Wt, mTau+/-, and hTau/mTau+/- mice (n = 9-10/group). In the absence of immune stimulation, increasing 4R tau levels in hTau/mTau+/- exacerbated pS202 and pS396/404 tau phosphorylation, without altering total tau levels or worsening early behavioral perturbations characteristic of hTau/mTau-/- mice. We also show for the first time that modulating 4R tau levels in hTau mice affects the response to systemic inflammation. Behavior was suppressed in all genotypes 4 h following LPS administration, but hTau/mTau+/- exhibited more severe sickness behavior at the 100 μg/kg dose and a milder behavioral and cytokine response than hTau/mTau-/- mice at the 330 μg/kg dose. All LPS doses decreased tau phosphorylation at both epitopes in hTau/mTau+/- mice, but pS202 levels were selectively reduced at the 100 μg/kg dose in hTau/mTau-/- mice. Behavioral suppression and decreased tau phosphorylation persisted at 24 h following LPS administration in hTau/mTau+/- mice.
Collapse
Affiliation(s)
- Matthew R Barron
- School of Life Sciences, Division of Physiology, Pharmacology and Neuroscience, Medical School, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Jane Gartlon
- EMEA Knowledge Centre, Eisai Ltd., Hatfield, United Kingdom
| | | | | | - Marie-Christine Pardon
- School of Life Sciences, Division of Physiology, Pharmacology and Neuroscience, Medical School, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
221
|
Bevan-Jones WR, Cope TE, Jones PS, Kaalund SS, Passamonti L, Allinson K, Green O, Hong YT, Fryer TD, Arnold R, Coles JP, Aigbirhio FI, Larner AJ, Patterson K, O’Brien JT, Rowe JB. Neuroinflammation and protein aggregation co-localize across the frontotemporal dementia spectrum. Brain 2020; 143:1010-1026. [PMID: 32179883 PMCID: PMC7089669 DOI: 10.1093/brain/awaa033] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 12/04/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
The clinical syndromes of frontotemporal dementia are clinically and neuropathologically heterogeneous, but processes such as neuroinflammation may be common across the disease spectrum. We investigated how neuroinflammation relates to the localization of tau and TDP-43 pathology, and to the heterogeneity of clinical disease. We used PET in vivo with (i) 11C-PK-11195, a marker of activated microglia and a proxy index of neuroinflammation; and (ii) 18F-AV-1451, a radioligand with increased binding to pathologically affected regions in tauopathies and TDP-43-related disease, and which is used as a surrogate marker of non-amyloid-β protein aggregation. We assessed 31 patients with frontotemporal dementia (10 with behavioural variant, 11 with the semantic variant and 10 with the non-fluent variant), 28 of whom underwent both 18F-AV-1451 and 11C-PK-11195 PET, and matched control subjects (14 for 18F-AV-1451 and 15 for 11C-PK-11195). We used a univariate region of interest analysis, a paired correlation analysis of the regional relationship between binding distributions of the two ligands, a principal component analysis of the spatial distributions of binding, and a multivariate analysis of the distribution of binding that explicitly controls for individual differences in ligand affinity for TDP-43 and different tau isoforms. We found significant group-wise differences in 11C-PK-11195 binding between each patient group and controls in frontotemporal regions, in both a regions-of-interest analysis and in the comparison of principal spatial components of binding. 18F-AV-1451 binding was increased in semantic variant primary progressive aphasia compared to controls in the temporal regions, and both semantic variant primary progressive aphasia and behavioural variant frontotemporal dementia differed from controls in the expression of principal spatial components of binding, across temporal and frontotemporal cortex, respectively. There was a strong positive correlation between 11C-PK-11195 and 18F-AV-1451 uptake in all disease groups, across widespread cortical regions. We confirmed this association with post-mortem quantification in 12 brains, demonstrating strong associations between the regional densities of microglia and neuropathology in FTLD-TDP (A), FTLD-TDP (C), and FTLD-Pick's. This was driven by amoeboid (activated) microglia, with no change in the density of ramified (sessile) microglia. The multivariate distribution of 11C-PK-11195 binding related better to clinical heterogeneity than did 18F-AV-1451: distinct spatial modes of neuroinflammation were associated with different frontotemporal dementia syndromes and supported accurate classification of participants. These in vivo findings indicate a close association between neuroinflammation and protein aggregation in frontotemporal dementia. The inflammatory component may be important in shaping the clinical and neuropathological patterns of the diverse clinical syndromes of frontotemporal dementia.
Collapse
Affiliation(s)
| | - Thomas E Cope
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sanne S Kaalund
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Luca Passamonti
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), via Fratelli Cervi, Milano, Italy
| | - Kieren Allinson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridge, UK
| | - Oliver Green
- Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), via Fratelli Cervi, Milano, Italy
| | - Young T Hong
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Robert Arnold
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | | | | | - Karalyn Patterson
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
222
|
Pilipović I, Stojić-Vukanić Z, Prijić I, Jasnić N, Leposavić G. Propranolol diminished severity of rat EAE by enhancing immunoregulatory/protective properties of spinal cord microglia. Neurobiol Dis 2020; 134:104665. [DOI: 10.1016/j.nbd.2019.104665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/08/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
|
223
|
Fan Q, He W, Gayen M, Benoit MR, Luo X, Hu X, Yan R. Activated CX3CL1/Smad2 Signals Prevent Neuronal Loss and Alzheimer's Tau Pathology-Mediated Cognitive Dysfunction. J Neurosci 2020; 40:1133-1144. [PMID: 31822518 PMCID: PMC6989010 DOI: 10.1523/jneurosci.1333-19.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 11/24/2022] Open
Abstract
Neurofibrillary tangles likely cause neurodegeneration in Alzheimer's disease (AD). We demonstrate that the CX3CL1 C-terminal domain can upregulate neurogenesis, which may ameliorate neurodegeneration. Here we generated transgenic (Tg-CX3CL1) mice by overexpressing CX3CL1 in neurons. Tg-CX3CL1 mice exhibit enhanced neurogenesis in both subgranular and subventricular zones. This enhanced neurogenesis correlates well with elevated expression of TGF-β2 and TGF-β3, and activation of their downstream signaling molecule Smad2. Intriguingly, the enhanced adult neurogenesis was mitigated when Smad2 expression was deleted in neurons, supporting a role for the CX3CL1-TGF-β2/3-Smad2 pathway in the control of adult neurogenesis. When Tg-CX3CL1 mice were crossed with Alzheimer's PS19 mice, which overexpress a tau P301S mutation and exhibit age-dependent neurofibrillary tangles and neurodegeneration, overexpressed CX3CL1 in both male and female mice was sufficient to rescue the neurodegeneration, increase survival time, and improve cognitive function. Hence, we provide in vivo evidence that CX3CL1 is a strong activator of adult neurogenesis, and that it reduces neuronal loss and improves cognitive function in AD.SIGNIFICANCE STATEMENT This study will be the first to demonstrate that enhanced neurogenesis by overexpressed CX3CL1 is mitigated by disruption of Smad2 signaling and is independent of its interaction with CX3CR1. Overexpression of CX3CL1 lengthens the life span of PS19 tau mice by enhancing adult neurogenesis while having minimal effect on tau pathology. Enhancing neuronal CX3CL1, mainly the C-terminal fragment, is a therapeutic strategy for blocking or reversing neuronal loss in Alzheimer's disease or related neurodegenerative disease patients.
Collapse
Affiliation(s)
- Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, and
| | - Wanxia He
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut 06032
| | - Manoshi Gayen
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut 06032
| | - Marc Robert Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut 06032
| | - Xiaoyang Luo
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, and
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut 06032
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, and
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut 06032
| |
Collapse
|
224
|
Lahiani-Cohen I, Touloumi O, Lagoudaki R, Grigoriadis N, Rosenmann H. Exposure to 3-Nitropropionic Acid Mitochondrial Toxin Induces Tau Pathology in Tangle-Mouse Model and in Wild Type-Mice. Front Cell Dev Biol 2020; 7:321. [PMID: 32010684 PMCID: PMC6971403 DOI: 10.3389/fcell.2019.00321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/21/2019] [Indexed: 01/24/2023] Open
Abstract
Oxidative stress, particularly of mitochondrial origin, plays an important role in the pathogenesis of neurodegenerative disorders, including Alzheimer’s disease (AD) and other tauopathies. Controversies regarding the responses of tau phosphorylation state to various stimuli causing oxidative stress have been reported. Here we investigated the effect of 3-nitropropionic acid (3NP), a mitochondrial toxin which induces oxidative stress, on the tangle-pathology in our previously generated double mutant (E257T/P301S, DM) -Tau-tg mice and in WT-mice. We detected an increase in tangle pathology in the hippocampus and cortex of the DM-Tau-tg mice following exposure of the mice to the toxin, as well as generation of tangles in WT-mice. This increase was accompanied with alterations in the level of the glycogen synthase kinase 3β (GSK3β), the kinase which phosphorylates the tau protein, and in the phosphorylation state of this kinase. A response of microglial cells was noticed. These results point to the involvement of mitochondrial dysfunction in the development of the tangle-pathology and may suggest that interfering with mitochondrial dysfunction may have an anti-tangle therapeutic potential.
Collapse
Affiliation(s)
- Inbal Lahiani-Cohen
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Olga Touloumi
- B' Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Roza Lagoudaki
- B' Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Hanna Rosenmann
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
225
|
Abstract
The symptoms of Alzheimer disease reflect a loss of neural circuit integrity in the brain, but neurons do not work in isolation. Emerging evidence suggests that the intricate balance of interactions between neurons, astrocytes, microglia and vascular cells required for healthy brain function becomes perturbed during the disease, with early changes likely protecting neural circuits from damage, followed later by harmful effects when the balance cannot be restored. Moving beyond a neuronal focus to understand the complex cellular interactions in Alzheimer disease and how these change throughout the course of the disease may provide important insight into developing effective therapeutics.
Collapse
|
226
|
Medina CS, Uselman TW, Barto DR, Cháves F, Jacobs RE, Bearer EL. Decoupling the Effects of the Amyloid Precursor Protein From Amyloid-β Plaques on Axonal Transport Dynamics in the Living Brain. Front Cell Neurosci 2019; 13:501. [PMID: 31849608 PMCID: PMC6901799 DOI: 10.3389/fncel.2019.00501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/22/2019] [Indexed: 11/16/2022] Open
Abstract
Amyloid precursor protein (APP) is the precursor to Aβ plaques. The cytoplasmic domain of APP mediates attachment of vesicles to molecular motors for axonal transport. In APP-KO mice, transport of Mn2+ is decreased. In old transgenic mice expressing mutated human (APPSwInd) linked to Familial Alzheimer's Disease, with both expression of APPSwInd and plaques, the rate and destination of Mn2+ axonal transport is altered, as detected by time-lapse manganese-enhanced magnetic resonance imaging (MEMRI) of the brain in living mice. To determine the relative contribution of expression of APPSwInd versus plaque on transport dynamics, we developed a Tet-off system to decouple expression of APPSwInd from plaque, and then studied hippocampal to forebrain transport by MEMRI. Three groups of mice were compared to wild-type (WT): Mice with plaque and APPSwInd expression; mice with plaque but suppression of APPSwInd expression; and mice with APPSwInd suppressed from mating until 2 weeks before imaging with no plaque. MR images were captured before at successive time points after stereotactic injection of Mn2+ (3-5 nL) into CA3 of the hippocampus. Mice were returned to their home cage between imaging sessions so that transport would occur in the awake freely moving animal. Images of multiple mice from the three groups (suppressed or expressed) together with C57/B6J WT were aligned and processed with our automated computational pipeline, and voxel-wise statistical parametric mapping (SPM) performed. At the conclusion of MR imaging, brains were harvested for biochemistry or histopathology. Paired T-tests within-group between time points (p = 0.01 FDR corrected) support the impression that both plaque alone and APPSwInd expression alone alter transport rates and destination of Mn2+ accumulation. Expression of APPSwInd in the absence of plaque or detectable Aβ also resulted in transport defects as well as pathology of hippocampus and medial septum, suggesting two sources of pathology occur in familial Alzheimer's disease, from toxic mutant protein as well as plaque. Alternatively mice with plaque without APPSwInd expression resemble the human condition of sporadic Alzheimer's, and had better transport. Thus, these mice with APPSwInd expression suppressed after plaque formation will be most useful in preclinical trials.
Collapse
Affiliation(s)
- Christopher S. Medina
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Taylor W. Uselman
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Daniel R. Barto
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Frances Cháves
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Russell E. Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- California Institute of Technology, Pasadena, CA, United States
| | - Elaine L. Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
227
|
Gyoneva S, Hosur R, Gosselin D, Zhang B, Ouyang Z, Cotleur AC, Peterson M, Allaire N, Challa R, Cullen P, Roberts C, Miao K, Reynolds TL, Glass CK, Burkly L, Ransohoff RM. Cx3cr1-deficient microglia exhibit a premature aging transcriptome. Life Sci Alliance 2019; 2:2/6/e201900453. [PMID: 31792059 PMCID: PMC6892408 DOI: 10.26508/lsa.201900453] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 12/13/2022] Open
Abstract
Gyoneva et al use RNA-seq to show that Cx3cr1-deficient microglia in young mice display a gene expression profile similar to microglia in aged mice, suggesting premature microglial aging. CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.
Collapse
Affiliation(s)
| | | | - David Gosselin
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Baohong Zhang
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Zhengyu Ouyang
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | | | | | - Norm Allaire
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Ravi Challa
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Patrick Cullen
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Chris Roberts
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Kelly Miao
- Acute Neurology, Biogen, Cambridge, MA, USA
| | | | - Christopher K Glass
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA.,School of Medicine, University of California San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
228
|
Hickman SE, Allison EK, Coleman U, Kingery-Gallagher ND, El Khoury J. Heterozygous CX3CR1 Deficiency in Microglia Restores Neuronal β-Amyloid Clearance Pathways and Slows Progression of Alzheimer's Like-Disease in PS1-APP Mice. Front Immunol 2019; 10:2780. [PMID: 31849963 PMCID: PMC6900980 DOI: 10.3389/fimmu.2019.02780] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/13/2019] [Indexed: 02/03/2023] Open
Abstract
CX3CR1 is a chemokine receptor expressed on microglia that binds Fractalkine (CX3CL1) and regulates microglial recruitment to sites of neuroinflammation. Full deletion of CX3CR1 in mouse models of Alzheimer's disease have opposing effects on amyloid-β and tau pathologies raising concerns about the benefits of targeting CX3CR1 for treatment of this disease. Since most therapies achieve only partial blockade of their targets, we investigated the effects of partial CX3CR1 deficiency on the development and progression of amyloid-β deposition in the PS1-APP Alzheimer's mouse model. We generated PS1-APP mice heterozygous for CX3CR1 (PS1-APP-CX3CR1+/−) and analyzed these mice for Alzheimer's-like pathology. We found that partial CX3CR1 deficiency was associated with a significant reduction in Aβ levels and in senile-like plaque load in the brain as compared with age-matched PS1-APP mice. Reduced Aβ level in the brain was associated with improved cognitive function. Levels of the neuronal-expressed Aβ-degrading enzymes insulysin and matrix metalloproteinase 9, which are reduced in the brains of regular PS1-APP mice, were significantly higher in PS1-APP-CX3CR1+/− mice. Our data indicate that lowering CX3CR1 levels or partially inhibiting its activity in the brain may be a therapeutic strategy to increase neuronal Aβ clearance, reduce Aβ levels and delay progression of Alzheimer's-Like disease. Our findings also suggest a novel pathway where microglial CX3CR1 can regulates gene expression in neurons.
Collapse
Affiliation(s)
- Suzanne E Hickman
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Elizabeth K Allison
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Uwanda Coleman
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Nathan D Kingery-Gallagher
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Joseph El Khoury
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
229
|
Mancuso R, Van Den Daele J, Fattorelli N, Wolfs L, Balusu S, Burton O, Liston A, Sierksma A, Fourne Y, Poovathingal S, Arranz-Mendiguren A, Sala Frigerio C, Claes C, Serneels L, Theys T, Perry VH, Verfaillie C, Fiers M, De Strooper B. Stem-cell-derived human microglia transplanted in mouse brain to study human disease. Nat Neurosci 2019; 22:2111-2116. [PMID: 31659342 PMCID: PMC7616913 DOI: 10.1038/s41593-019-0525-x] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 09/26/2019] [Indexed: 11/09/2022]
Abstract
Although genetics highlights the role of microglia in Alzheimer's disease, one-third of putative Alzheimer's disease risk genes lack adequate mouse orthologs. Here we successfully engraft human microglia derived from embryonic stem cells in the mouse brain. The cells recapitulate transcriptionally human primary microglia ex vivo and show expression of human-specific Alzheimer's disease risk genes. Oligomeric amyloid-β induces a divergent response in human versus mouse microglia. This model can be used to study the role of microglia in neurological diseases.
Collapse
Affiliation(s)
- Renzo Mancuso
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Johanna Van Den Daele
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Nicola Fattorelli
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Leen Wolfs
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Sriram Balusu
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Oliver Burton
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Adrian Liston
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Annerieke Sierksma
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Yannick Fourne
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Suresh Poovathingal
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Amaia Arranz-Mendiguren
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Carlo Sala Frigerio
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Christel Claes
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Lutgarde Serneels
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tom Theys
- Department of Neurosciences, Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - V Hugh Perry
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Mark Fiers
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- UK Dementia Research Institute at UCL, University College London, London, UK.
| |
Collapse
|
230
|
Benoit MR, Gayen M, Yan R. CX3CL1 intracellular domain and adult neurogenesis. Aging (Albany NY) 2019; 11:10783-10785. [PMID: 31785144 PMCID: PMC6932901 DOI: 10.18632/aging.102504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 12/03/2022]
Affiliation(s)
- Marc R Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06032, USA
| | - Manoshi Gayen
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06032, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06032, USA
| |
Collapse
|
231
|
Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat Rev Neurosci 2019; 21:21-35. [PMID: 31780819 DOI: 10.1038/s41583-019-0240-3] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
The global epidemic of Alzheimer disease (AD) is worsening, and no approved treatment can revert or arrest progression of this disease. AD pathology is characterized by the accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain. Genetic data, as well as autopsy and neuroimaging studies in patients with AD, indicate that Aβ plaque deposition precedes cortical tau pathology. Because Aβ accumulation has been considered the initial insult that drives both the accumulation of tau pathology and tau-mediated neurodegeneration in AD, the development of AD therapeutics has focused mostly on removing Aβ from the brain. However, striking preclinical evidence from AD mouse models and patient-derived human induced pluripotent stem cell models indicates that tau pathology can progress independently of Aβ accumulation and arises downstream of genetic risk factors for AD and aberrant metabolic pathways. This Review outlines novel insights from preclinical research that implicate apolipoprotein E, the endocytic system, cholesterol metabolism and microglial activation as Aβ-independent regulators of tau pathology. These factors are discussed in the context of emerging findings from clinical pathology, functional neuroimaging and other approaches in humans. Finally, we discuss the implications of these new insights for current Aβ-targeted strategies and highlight the emergence of novel therapeutic strategies that target processes upstream of both Aβ and tau.
Collapse
|
232
|
Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, Schwartz S, Albasset S, McManus RM, Tejera D, Griep A, Santarelli F, Brosseron F, Opitz S, Stunden J, Merten M, Kayed R, Golenbock DT, Blum D, Latz E, Buée L, Heneka MT. NLRP3 inflammasome activation drives tau pathology. Nature 2019; 575:669-673. [PMID: 31748742 DOI: 10.1038/s41586-019-1769-z] [Citation(s) in RCA: 921] [Impact Index Per Article: 153.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is characterized by the accumulation of amyloid-beta in plaques, aggregation of hyperphosphorylated tau in neurofibrillary tangles and neuroinflammation, together resulting in neurodegeneration and cognitive decline1. The NLRP3 inflammasome assembles inside of microglia on activation, leading to increased cleavage and activity of caspase-1 and downstream interleukin-1β release2. Although the NLRP3 inflammasome has been shown to be essential for the development and progression of amyloid-beta pathology in mice3, the precise effect on tau pathology remains unknown. Here we show that loss of NLRP3 inflammasome function reduced tau hyperphosphorylation and aggregation by regulating tau kinases and phosphatases. Tau activated the NLRP3 inflammasome and intracerebral injection of fibrillar amyloid-beta-containing brain homogenates induced tau pathology in an NLRP3-dependent manner. These data identify an important role of microglia and NLRP3 inflammasome activation in the pathogenesis of tauopathies and support the amyloid-cascade hypothesis in Alzheimer's disease, demonstrating that neurofibrillary tangles develop downstream of amyloid-beta-induced microglial activation.
Collapse
Affiliation(s)
- Christina Ising
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Carmen Venegas
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - Shuangshuang Zhang
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Hannah Scheiblich
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Ana Vieira-Saecker
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stephanie Schwartz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Shadi Albasset
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Róisín M McManus
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dario Tejera
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | | | - Sabine Opitz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Maximilian Merten
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases and Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Douglas T Golenbock
- Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - David Blum
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, "Alzheimer & Tauopathies", Labex DISTALZ, Lille, France
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany.,Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, "Alzheimer & Tauopathies", Labex DISTALZ, Lille, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany. .,Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
233
|
Zhang SZ, Wang QQ, Yang QQ, Gu HY, Yin YQ, Li YD, Hou JC, Chen R, Sun QQ, Sun YF, Hu G, Zhou JW. NG2 glia regulate brain innate immunity via TGF-β2/TGFBR2 axis. BMC Med 2019; 17:204. [PMID: 31727112 PMCID: PMC6857135 DOI: 10.1186/s12916-019-1439-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Brain innate immunity is vital for maintaining normal brain functions. Immune homeostatic imbalances play pivotal roles in the pathogenesis of neurological diseases including Parkinson's disease (PD). However, the molecular and cellular mechanisms underlying the regulation of brain innate immunity and their significance in PD pathogenesis are still largely unknown. METHODS Cre-inducible diphtheria toxin receptor (iDTR) and diphtheria toxin-mediated cell ablation was performed to investigate the impact of neuron-glial antigen 2 (NG2) glia on the brain innate immunity. RNA sequencing analysis was carried out to identify differentially expressed genes in mouse brain with ablated NG2 glia and lipopolysaccharide (LPS) challenge. Neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice were used to evaluate neuroinflammatory response in the presence or absence of NG2 glia. The survival of dopaminergic neurons or glial cell activation was evaluated by immunohistochemistry. Co-cultures of NG2 glia and microglia were used to examine the influence of NG2 glia to microglial activation. RESULTS We show that NG2 glia are required for the maintenance of immune homeostasis in the brain via transforming growth factor-β2 (TGF-β2)-TGF-β type II receptor (TGFBR2)-CX3C chemokine receptor 1 (CX3CR1) signaling, which suppresses the activation of microglia. We demonstrate that mice with ablated NG2 glia display a profound downregulation of the expression of microglia-specific signature genes and remarkable inflammatory response in the brain following exposure to endotoxin lipopolysaccharides. Gain- or loss-of-function studies show that NG2 glia-derived TGF-β2 and its receptor TGFBR2 in microglia are key regulators of the CX3CR1-modulated immune response. Furthermore, deficiency of NG2 glia contributes to neuroinflammation and nigral dopaminergic neuron loss in MPTP-induced mouse PD model. CONCLUSIONS These findings suggest that NG2 glia play a critical role in modulation of neuroinflammation and provide a compelling rationale for the development of new therapeutics for neurological disorders.
Collapse
Affiliation(s)
- Shu-Zhen Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Qin-Qin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.,Neurobiology Key Laboratory, Jining Medical University, Jining, 272067, Shandong, China
| | - Qiao-Qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Huan-Yu Gu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yan-Qing Yin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yan-Dong Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Jin-Can Hou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Rong Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Qing Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying-Feng Sun
- Center for Brain Disorders Research, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100053, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jia-Wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China. .,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China. .,Co-innovation Center of Neuroregeneration, School of Medicine, Nantong University, Nantong, 226001, Jiangsu, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| |
Collapse
|
234
|
Hassan-Abdi R, Brenet A, Bennis M, Yanicostas C, Soussi-Yanicostas N. Neurons Expressing Pathological Tau Protein Trigger Dramatic Changes in Microglial Morphology and Dynamics. Front Neurosci 2019; 13:1199. [PMID: 31787873 PMCID: PMC6855094 DOI: 10.3389/fnins.2019.01199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/22/2019] [Indexed: 01/12/2023] Open
Abstract
Microglial cells, the resident macrophages of the brain, are important players in the pathological process of numerous neurodegenerative disorders, including tauopathies, a heterogeneous class of diseases characterized by intraneuronal Tau aggregates. However, microglia response in Tau pathologies remains poorly understood. Here, we exploit a genetic zebrafish model of tauopathy, combined with live microglia imaging, to investigate the behavior of microglia in vivo in the disease context. Results show that while microglia were almost immobile and displayed long and highly dynamic branches in a wild-type context, in presence of diseased neurons, cells became highly mobile and displayed morphological changes, with highly mobile cell bodies together with fewer and shorter processes. We also imaged, for the first time to our knowledge, the phagocytosis of apoptotic tauopathic neurons by microglia in vivo and observed that microglia engulfed about as twice materials as in controls. Finally, genetic ablation of microglia in zebrafish tauopathy model significantly increased Tau hyperphosphorylation, suggesting that microglia provide neuroprotection to diseased neurons. Our findings demonstrate for the first time the dynamics of microglia in contact with tauopathic neurons in vivo and open perspectives for the real-time study of microglia in many neuronal diseases.
Collapse
Affiliation(s)
- Rahma Hassan-Abdi
- INSERM, UMR1141, Hôpital Robert Debré, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Alexandre Brenet
- INSERM, UMR1141, Hôpital Robert Debré, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Constantin Yanicostas
- INSERM, UMR1141, Hôpital Robert Debré, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Nadia Soussi-Yanicostas
- INSERM, UMR1141, Hôpital Robert Debré, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
235
|
Cao S, Fisher DW, Yu T, Dong H. The link between chronic pain and Alzheimer's disease. J Neuroinflammation 2019; 16:204. [PMID: 31694670 PMCID: PMC6836339 DOI: 10.1186/s12974-019-1608-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic pain often occurs in the elderly, particularly in the patients with neurodegenerative disorders such as Alzheimer's disease (AD). Although studies indicate that chronic pain correlates with cognitive decline, it is unclear whether chronic pain accelerates AD pathogenesis. In this review, we provide evidence that supports a link between chronic pain and AD and discuss potential mechanisms underlying this connection based on currently available literature from human and animal studies. Specifically, we describe two intertwined processes, locus coeruleus noradrenergic system dysfunction and neuroinflammation resulting from microglial pro-inflammatory activation in brain areas mediating the affective component of pain and cognition that have been found to influence both chronic pain and AD. These represent a pathological overlap that likely leads chronic pain to accelerate AD pathogenesis. Further, we discuss potential therapeutic interventions targeting noradrenergic dysfunction and microglial activation that may improve patient outcomes for those with chronic pain and AD.
Collapse
Affiliation(s)
- Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 56300, Guizhou, China
- Guizhou Key Lab of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 56300, Guizhou, China
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Daniel W Fisher
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Tain Yu
- Guizhou Key Lab of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 56300, Guizhou, China
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA.
| |
Collapse
|
236
|
Golde TE. Harnessing Immunoproteostasis to Treat Neurodegenerative Disorders. Neuron 2019; 101:1003-1015. [PMID: 30897353 DOI: 10.1016/j.neuron.2019.02.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
Abstract
Immunoproteostasis is a term used to reflect interactions between the immune system and the proteinopathies that are presumptive "triggers" of many neurodegenerative disorders. The study of immunoproteostasis is bolstered by several observations. Mutations or rare variants in genes expressed in microglial cells, known to regulate immune functions, or both can cause, or alter risk for, various neurodegenerative disorders. Additionally, genetic association studies identify numerous loci harboring genes that encode proteins of known immune function that alter risk of developing Alzheimer's disease (AD) and other neurodegenerative proteinopathies. Further, preclinical studies reveal beneficial effects and liabilities of manipulating immune pathways in various neurodegenerative disease models. Although there are concerns that manipulation of the immune system may cause more harm than good, there is considerable interest in developing immune modulatory therapies for neurodegenerative disorders. Herein, I highlight the promise and challenges of harnessing immunoproteostasis to treat neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Todd E Golde
- McKnight Brain Institute, Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience and Neurology, University of Florida, Gainesville, FL 32607, USA.
| |
Collapse
|
237
|
Bugay V, Bozdemir E, Vigil FA, Chun SH, Holstein DM, Elliott WR, Sprague CJ, Cavazos JE, Zamora DO, Rule G, Shapiro MS, Lechleiter JD, Brenner R. A Mouse Model of Repetitive Blast Traumatic Brain Injury Reveals Post-Trauma Seizures and Increased Neuronal Excitability. J Neurotrauma 2019; 37:248-261. [PMID: 31025597 DOI: 10.1089/neu.2018.6333] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Repetitive blast traumatic brain injury (TBI) affects numerous soldiers on the battlefield. Mild TBI has been shown to have long-lasting effects with repeated injury. We have investigated effects on neuronal excitability after repetitive, mild TBI in a mouse model of blast-induced brain injury. We exposed mice to mild blast trauma of an average peak overpressure of 14.6 psi, repeated across three consecutive days. While a single exposure did not reveal trauma as indicated by the glial fibrillary acidic protein indicator, three repetitive blasts did show significant increases. As well, mice had an increased indicator of inflammation (Iba-1) and increased tau, tau phosphorylation, and altered cytokine levels in the spleen. Video-electroencephalographic monitoring 48 h after the final blast exposure demonstrated seizures in 50% (12/24) of the mice, most of which were non-convulsive seizures. Long-term monitoring revealed that spontaneous seizures developed in at least 46% (6/13) of the mice. Patch clamp recording of dentate gyrus hippocampus neurons 48 h post-blast TBI demonstrated a shortened latency to the first spike and hyperpolarization of action potential threshold. We also found that evoked excitatory postsynaptic current amplitudes were significantly increased. These findings indicate that mild, repetitive blast exposures cause increases in neuronal excitability and seizures and eventual epilepsy development in some animals. The non-convulsive nature of the seizures suggests that subclinical seizures may occur in individuals experiencing even mild blast events, if repeated.
Collapse
Affiliation(s)
- Vladislav Bugay
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Fabio A Vigil
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - William R Elliott
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Cassie J Sprague
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Jose E Cavazos
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Neurology, University of Texas Health San Antonio, San Antonio, Texas
| | - David O Zamora
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | | | - Mark S Shapiro
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert Brenner
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
238
|
Torvell M, Hampton DW, Connick P, MacLullich AMJ, Cunningham C, Chandran S. A single systemic inflammatory insult causes acute motor deficits and accelerates disease progression in a mouse model of human tauopathy. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:579-591. [PMID: 31650014 PMCID: PMC6804509 DOI: 10.1016/j.trci.2019.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Introduction Neuroinflammation, which contributes to neurodegeneration, is a consistent hallmark of dementia. Emerging evidence suggests that systemic inflammation also contributes to disease progression. Methods The ability of systemically administered lipopolysaccharide (LPS - 500 μg/kg) to effect acute and chronic behavioural changes in C57BL/6 and P301S tauopathy mice was assessed. Markers of pathology were assessed in the brain and spinal cord. Results P301S mice display regional microgliosis. Systemic LPS treatment induced exaggerated acute sickness behaviour and motor dysfunction in P301S mice compared with wild-type controls and advanced the onset and accelerated chronic decline. LPS treatment was associated with increased tau pathology 24 hours after LPS injection and spinal cord microgliosis at the end stage. Discussion This is the first demonstration that a single systemic inflammatory episode causes exaggerated acute functional impairments and accelerates the long-term trajectory of functional decline associated with neurodegeneration in a mouse model of human tauopathy. The findings have relevance to management of human dementias. P301S microgliosis is regional; activation occurs in the spinal cord but not in the cortex. Systemic LPS injection caused acute neurological deficits in P301S mice. This was associated with increased tau pathology 24 hours after LPS injection. This was independent of microglial priming as measured by IL-1β hyperexpression. LPS injection advanced the onset of chronic decline in P301S mice.
Collapse
Affiliation(s)
- Megan Torvell
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Cardiff, Cardiff, UK
| | - David W Hampton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Peter Connick
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian, UK
| | - Alasdair M J MacLullich
- Edinburgh Delirium Research Group, Geriatric Medicine, University of Edinburgh, Edinburgh, UK
| | - Colm Cunningham
- Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK.,The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian, UK
| |
Collapse
|
239
|
Španić E, Langer Horvat L, Hof PR, Šimić G. Role of Microglial Cells in Alzheimer's Disease Tau Propagation. Front Aging Neurosci 2019; 11:271. [PMID: 31636558 PMCID: PMC6787141 DOI: 10.3389/fnagi.2019.00271] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022] Open
Abstract
Uncontrolled immune response in the brain contributes to the progression of all neurodegenerative disease, including Alzheimer's disease (AD). Recent investigations have documented the prion-like features of tau protein and the involvement of microglial changes with tau pathology. While it is still unclear what sequence of events is causal, it is likely that tau seeding potential and microglial contribution to tau propagation act together, and are essential for the development and progression of degenerative changes. Based on available evidence, targeting tau seeds and controlling some signaling pathways in a complex inflammation process could represent a possible new therapeutic approach for treating neurodegenerative diseases. Recent findings propose novel diagnostic assays and markers that may be used together with standard methods to complete and improve the diagnosis and classification of these diseases. In conclusion, a novel perspective on microglia-tau relations reveals new issues to investigate and imposes different approaches for developing therapeutic strategies for AD.
Collapse
Affiliation(s)
- Ena Španić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lea Langer Horvat
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
240
|
Li Z, Cao X, Ma H, Cui Y, Li X, Wang N, Zhou Y. Surgical Trauma Exacerbates Cognitive Deficits and Neuroinflammation in Aged Rats: The Role of CX3CL1-CX3CR1 Signaling. J Neuropathol Exp Neurol 2019; 77:736-746. [PMID: 29939299 DOI: 10.1093/jnen/nly051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Age is the most prominent risk factor for the development of postoperative cognitive dysfunction. The present study investigated the role of CX3CL1-CX3CR1 signaling in age-related differences in surgery-induced cognitive deficits and neuroinflammation. Adult and aged male Sprague-Dawley rats were subjected to partial hepatectomy or partial hepatectomy with intracerebroventricular infusion of CX3CL1. On postoperative days 3, 7, and 14, the rats were subjected to an open field test and the Morris water maze test. Hippocampal interleukin-1β, CX3CL1, CX3CR1, brain derived neurotrophic factor (BDNF), ionized calcium-binding adapter molecule 1 (Iba-1), and Arginase-1 (Arg1) levels were measured. Age exacerbated cognitive impairment and increased neuroinflammation following surgery. Surgery-induced decreases in CX3CL1 and CX3CR1 proteins were accompanied by increased microglial activation, as indicated by increased Iba-1 expression. Corresponding decline in Arg1 and BDNF levels were observed. Treatment with CX3CL1 decreased proinflammatory cytokines expression, increased BDNF and Arg1 levels in the brain, and enhanced behavioral recovery. The surgery-induced decreases in CX3CL1 and CX3CR1 expression exacerbated postoperative cognitive deficits and exaggerated neuroinflammatory responses in this rodent model. Treatment with CX3CL1 attenuated these effects, at least partly by inhibiting microglial activation, decreasing the associated production of proinflammatory cytokines, and enhancing BDNF expression.
Collapse
Affiliation(s)
- Zhe Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuezhao Cao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hong Ma
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yong Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoqian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yongjian Zhou
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
241
|
Mancuso R, Fryatt G, Cleal M, Obst J, Pipi E, Monzón-Sandoval J, Ribe E, Winchester L, Webber C, Nevado A, Jacobs T, Austin N, Theunis C, Grauwen K, Daniela Ruiz E, Mudher A, Vicente-Rodriguez M, Parker CA, Simmons C, Cash D, Richardson J. CSF1R inhibitor JNJ-40346527 attenuates microglial proliferation and neurodegeneration in P301S mice. Brain 2019; 142:3243-3264. [PMID: 31504240 PMCID: PMC6794948 DOI: 10.1093/brain/awz241] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 11/12/2022] Open
Abstract
Neuroinflammation and microglial activation are significant processes in Alzheimer's disease pathology. Recent genome-wide association studies have highlighted multiple immune-related genes in association with Alzheimer's disease, and experimental data have demonstrated microglial proliferation as a significant component of the neuropathology. In this study, we tested the efficacy of the selective CSF1R inhibitor JNJ-40346527 (JNJ-527) in the P301S mouse tauopathy model. We first demonstrated the anti-proliferative effects of JNJ-527 on microglia in the ME7 prion model, and its impact on the inflammatory profile, and provided potential CNS biomarkers for clinical investigation with the compound, including pharmacokinetic/pharmacodynamics and efficacy assessment by TSPO autoradiography and CSF proteomics. Then, we showed for the first time that blockade of microglial proliferation and modification of microglial phenotype leads to an attenuation of tau-induced neurodegeneration and results in functional improvement in P301S mice. Overall, this work strongly supports the potential for inhibition of CSF1R as a target for the treatment of Alzheimer's disease and other tau-mediated neurodegenerative diseases.
Collapse
Affiliation(s)
- Renzo Mancuso
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Gemma Fryatt
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Madeleine Cleal
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Juliane Obst
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Elena Pipi
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jimena Monzón-Sandoval
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Elena Ribe
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Laura Winchester
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Caleb Webber
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Alejo Nevado
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Tom Jacobs
- Janssen Research and Development, Turnhoutseweg 30, box 270, 2340 Beerse 1, Belgium
| | - Nigel Austin
- Janssen Research and Development, Turnhoutseweg 30, box 270, 2340 Beerse 1, Belgium
| | - Clara Theunis
- Janssen Neuroscience Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Karolien Grauwen
- Janssen Neuroscience Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Eva Daniela Ruiz
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Amrit Mudher
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Marta Vicente-Rodriguez
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Christine A Parker
- Experimental Medicine Imaging, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Camilla Simmons
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Jill Richardson
- Neurosciences Therapeutic Area, GlaxoSmithKline R&D, Stevenage, UK
| |
Collapse
|
242
|
Abstract
Sporadic Alzheimer's disease is the most common neurodegenerative disorder and represents a very important public healthcare problem with a devastating economic burden for industrialized countries. Recent knowledge acquired from experimental, epidemiological, radiological and genome-wide association studies (GWAS) underline the role of the innate immune system in the pathophysiology of this disease. This article reviews and discusses the function of the cerebral innate immune system, the newly discovered genes associated with the disease development and the experimental evidence around the role of microglia in the onset and progression of Alzheimer's disease. The discovery of different microglia phenotypes associated with the pathology as well as new molecular players will enable the development of new preventive and therapeutic strategies by modulating neuroinflammation in neurodegenerative diseases.
Collapse
|
243
|
Abstract
The endotoxin hypothesis of neurodegeneration is the hypothesis that endotoxin causes or contributes to neurodegeneration. Endotoxin is a lipopolysaccharide (LPS), constituting much of the outer membrane of gram-negative bacteria, present at high concentrations in gut, gums and skin and in other tissue during bacterial infection. Blood plasma levels of endotoxin are normally low, but are elevated during infections, gut inflammation, gum disease and neurodegenerative disease. Adding endotoxin at such levels to blood of healthy humans induces systemic inflammation and brain microglial activation. Adding high levels of endotoxin to the blood or body of rodents induces microglial activation, priming and/or tolerance, memory deficits and loss of brain synapses and neurons. Endotoxin promotes amyloid β and tau aggregation and neuropathology, suggesting the possibility that endotoxin synergises with different aggregable proteins to give different neurodegenerative diseases. Blood and brain endotoxin levels are elevated in Alzheimer's disease, which is accelerated by systemic infections, including gum disease. Endotoxin binds directly to APOE, and the APOE4 variant both sensitises to endotoxin and predisposes to Alzheimer's disease. Intestinal permeability increases early in Parkinson's disease, and injection of endotoxin into mice induces α-synuclein production and aggregation, as well as loss of dopaminergic neurons in the substantia nigra. The gut microbiome changes in Parkinson's disease, and changing the endotoxin-producing bacterial species can affect the disease in patients and mouse models. Blood endotoxin is elevated in amyotrophic lateral sclerosis, and endotoxin promotes TDP-43 aggregation and neuropathology. Peripheral diseases that elevate blood endotoxin, such as sepsis, AIDS and liver failure, also result in neurodegeneration. Endotoxin directly and indirectly activates microglia that damage neurons via nitric oxide, oxidants and cytokines, and by phagocytosis of synapses and neurons. The endotoxin hypothesis is unproven, but if correct, then neurodegeneration may be reduced by decreasing endotoxin levels or endotoxin-induced neuroinflammation.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK.
| |
Collapse
|
244
|
Dunn-Meynell AA, Dowling P, Marchese M, Rodriguez E, Blumberg B, Choi YB, Gaindh D, Lu W. In vivo Bioluminescence Imaging Used to Monitor Disease Activity and Therapeutic Response in a Mouse Model of Tauopathy. Front Aging Neurosci 2019; 11:252. [PMID: 31572168 PMCID: PMC6751306 DOI: 10.3389/fnagi.2019.00252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/26/2019] [Indexed: 01/03/2023] Open
Abstract
Many studies of tauopathy use transgenic mice that overexpress the P301S mutant form of tau. Neuronal damage in these mice is associated with astrogliosis and induction of glial fibrillary acidic protein (GFAP) expression. GFAP-luc transgenic mice express firefly luciferase under the GFAP promoter, allowing bioluminescence to be measured non-invasively as a surrogate biomarker for astrogliosis. We bred double transgenic mice possessing both P301S and GFAP-luc cassettes and compared them to control mice bearing only the GFAP-luc transgene. We used serial bioluminescent images to define the onset and the time course of astrogliosis in these mice and this was correlated with the development of clinical deficit. Mice containing both GFAP-luc and P301S transgenes showed increased luminescence indicative of astroglial activation in the brain and spinal cord. Starting at 5 months old, the onset of clinical deterioration in these mice corresponded closely to the initial rise in the luminescent signal. Post mortem analysis showed the elevated luminescence was correlated with hyperphosphorylated tau deposition in the hippocampus of double transgenic mice. We used this method to determine the therapeutic effect of JM4 peptide [a small peptide immunomodulatory agent derived from human erythropoietin (EPO)] on double transgenic mice. JM4 treatment significantly decreased the intensity of luminescence, neurological deficit and hyperphosphorylated tau in mice with both the P301S and GFAP-luc transgenes. These findings indicate that bioluminescence imaging (BLI) is a powerful tool for quantifying GFAP expression in living P301S mice and can be used as a noninvasive biomarker of tau-induced neurodegeneration in preclinical therapeutic trials.
Collapse
Affiliation(s)
- Ambrose A. Dunn-Meynell
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Peter Dowling
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Michelle Marchese
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Esther Rodriguez
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Benjamin Blumberg
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Yun-Beom Choi
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Deeya Gaindh
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Wei Lu
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| |
Collapse
|
245
|
Moore Z, Taylor JM, Crack PJ. The involvement of microglia in Alzheimer's disease: a new dog in the fight. Br J Pharmacol 2019; 176:3533-3543. [PMID: 30445661 PMCID: PMC6715787 DOI: 10.1111/bph.14546] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/17/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
First described clinically in 1906, Alzheimer's disease (AD) is the most common neurodegenerative disease and form of dementia worldwide. Despite its prevalence, only five therapies are currently approved for AD, all dealing with the symptoms rather than the underlying causes of the disease. A multitude of experimental evidence has suggested that the once thought inconsequential process of neuroinflammation does, in fact, contribute to the AD pathogenesis. One such CNS cell type critical to this process are microglia. Plastic in nature with varied roles, microglia are emerging as key contributors to AD pathology. This review will focus on the role of microglia in the neuroinflammatory response in AD, highlighting recent studies implicating aberrant changes in microglial function in disease progression. Of critical note is that with these advances, a reconceptualization of the framework in which we view microglia is required. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Zachery Moore
- Neuropharmacology Laboratory, Department of Pharmacology and TherapeuticsUniversity of MelbourneMelbourneVICAustralia
| | - Juliet M Taylor
- Neuropharmacology Laboratory, Department of Pharmacology and TherapeuticsUniversity of MelbourneMelbourneVICAustralia
| | - Peter J Crack
- Neuropharmacology Laboratory, Department of Pharmacology and TherapeuticsUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
246
|
Mayer AR, Dodd AB, Vermillion MS, Stephenson DD, Chaudry IH, Bragin DE, Gigliotti AP, Dodd RJ, Wasserott BC, Shukla P, Kinsler R, Alonzo SM. A systematic review of large animal models of combined traumatic brain injury and hemorrhagic shock. Neurosci Biobehav Rev 2019; 104:160-177. [PMID: 31255665 PMCID: PMC7307133 DOI: 10.1016/j.neubiorev.2019.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023]
Abstract
Traumatic brain injury (TBI) and severe blood loss (SBL) frequently co-occur in human trauma, resulting in high levels of mortality and morbidity. Importantly, each of the individual post-injury cascades is characterized by complex and potentially opposing pathophysiological responses, complicating optimal resuscitation and therapeutic approaches. Large animal models of poly-neurotrauma closely mimic human physiology, but a systematic literature review of published models has been lacking. The current review suggests a relative paucity of large animal poly-neurotrauma studies (N = 52), with meta-statistics revealing trends for animal species (exclusively swine), characteristics (use of single biological sex, use of juveniles) and TBI models. Although most studies have targeted blood loss volumes of 35-45%, the associated mortality rates are much lower relative to Class III/IV human trauma. This discrepancy may result from potentially mitigating experimental factors (e.g., mechanical ventilation prior to or during injury, pausing/resuming blood loss based on physiological parameters, administration of small volume fluid resuscitation) that are rarely associated with human trauma, highlighting the need for additional work in this area.
Collapse
Affiliation(s)
- Andrew R Mayer
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States; Neurology Department, University of New Mexico School of Medicine, Albuquerque, NM 87131, United States; Psychiatry Department, University of New Mexico School of Medicine, Albuquerque, NM 87131, United States; Psychology Department, University of New Mexico, Albuquerque, NM 87131, United States.
| | - Andrew B Dodd
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Meghan S Vermillion
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - David D Stephenson
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States
| | - Denis E Bragin
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM 87131, United States
| | - Andrew P Gigliotti
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Rebecca J Dodd
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Benjamin C Wasserott
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Priyank Shukla
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| | - Rachel Kinsler
- Department of the Army Civilian, U.S. Army Aeromedical Research Laboratory, Fort Rucker, AL 36362-0577, United States
| | - Sheila M Alonzo
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, 1011 Yale Blvd. NE, Albuquerque, NM 87106, United States
| |
Collapse
|
247
|
Abstract
Animal models are indispensable tools for Alzheimer disease (AD) research. Over the course of more than two decades, an increasing number of complementary rodent models has been generated. These models have facilitated testing hypotheses about the aetiology and progression of AD, dissecting the associated pathomechanisms and validating therapeutic interventions, thereby providing guidance for the design of human clinical trials. However, the lack of success in translating rodent data into therapeutic outcomes may challenge the validity of the current models. This Review critically evaluates the genetic and non-genetic strategies used in AD modelling, discussing their strengths and limitations, as well as new opportunities for the development of better models for the disease.
Collapse
|
248
|
Shi Y, Holtzman DM. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 2019; 18:759-772. [PMID: 30140051 DOI: 10.1038/s41577-018-0051-1] [Citation(s) in RCA: 400] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer disease is more than a pure proteopathy. Chronic neuroinflammation stands out during the pathogenesis of the disease and in turn modulates disease progression. The central nervous system (CNS) is separated from the blood circulation by the blood-brain barrier. In Alzheimer disease, neuroinflammation heavily relies on innate immune responses that are primarily mediated by CNS-resident microglia. APOE (which encodes apolipoprotein E) is the strongest genetic risk factor for Alzheimer disease, and APOE was recently shown to affect the disease in part through its immunomodulatory function. This function of APOE is likely linked to triggering receptor expressed on myeloid cells 2 (TREM2), which is expressed by microglia in the CNS. Here, we review the rapidly growing literature on the role of disease-associated microglia, TREM2 and APOE in the pathogenesis of Alzheimer disease and present an integrated view of innate immune function in Alzheimer disease.
Collapse
Affiliation(s)
- Yang Shi
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
249
|
Vogels T, Murgoci AN, Hromádka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun 2019; 7:109. [PMID: 31277708 PMCID: PMC6612163 DOI: 10.1186/s40478-019-0754-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Tauopathies are a heterogenous class of diseases characterized by cellular accumulation of aggregated tau and include diseases such as Alzheimer’s disease (AD), progressive supranuclear palsy and chronic traumatic encephalopathy. Tau pathology is strongly linked to neurodegeneration and clinical symptoms in tauopathy patients. Furthermore, synapse loss is an early pathological event in tauopathies and is the strongest correlate of cognitive decline. Tau pathology is additionally associated with chronic neuroinflammatory processes, such as reactive microglia, astrocytes, and increased levels of pro-inflammatory molecules (e.g. complement proteins, cytokines). Recent studies show that as the principal immune cells of the brain, microglia play a particularly important role in the initiation and progression of tau pathology and associated neurodegeneration. Furthermore, AD risk genes such as Triggering receptor expressed on myeloid cells 2 (TREM2) and Apolipoprotein E (APOE) are enriched in the innate immune system and modulate the neuroinflammatory response of microglia to tau pathology. Microglia can play an active role in synaptic dysfunction by abnormally phagocytosing synaptic compartments of neurons with tau pathology. Furthermore, microglia are involved in synaptic spreading of tau – a process which is thought to underlie the progressive nature of tau pathology propagation through the brain. Spreading of pathological tau is also the predominant target for tau-based immunotherapy. Active tau vaccines, therapeutic tau antibodies and other approaches targeting the immune system are actively explored as treatment options for AD and other tauopathies. This review describes the role of microglia in the pathobiology of tauopathies and the mechanism of action of potential therapeutics targeting the immune system in tauopathies.
Collapse
|
250
|
Šimić G, Španić E, Langer Horvat L, Hof PR. Blood-brain barrier and innate immunity in the pathogenesis of Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:99-145. [PMID: 31699331 DOI: 10.1016/bs.pmbts.2019.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathogenesis of Alzheimer's disease (AD) is only partly understood. This is the probable reason why significant efforts to treat or prevent AD have been unsuccessful. In fact, as of April 2019, there have been 2094 studies registered for AD on the clinicaltrials.gov U.S. National Library of Science web page, of which only a few are still ongoing. In AD, abnormal accumulation of amyloid and tau proteins in the brain are thought to begin 10-20 years before the onset of overt symptoms, suggesting that interventions designed to prevent pathological amyloid and tau accumulation may be more effective than attempting to reverse a pathology once it is established. However, to be successful, such early interventions need to be selectively administered to individuals who will likely develop the disease long before the symptoms occur. Therefore, it is critical to identify early biomarkers that are strongly predictive of AD. Currently, patients are diagnosed on the basis of a variety of clinical scales, neuropsychological tests, imaging and laboratory modalities, but definitive diagnosis can be made only by postmortem assessment of underlying neuropathology. People suffering from AD thus may be misdiagnosed clinically with other primary causes of dementia, and vice versa, thereby also reducing the power of clinical trials. The amyloid cascade hypothesis fits well for the familial cases of AD with known mutations, but is not sufficient to explain sporadic, late-onset AD (LOAD) that accounts for over 95% of all cases. Since the earliest descriptions of AD there have been neuropathological features described other than amyloid plaques (AP) and neurofibrillary tangles (NFT), most notably gliosis and neuroinflammation. However, it is only recently that genetic and experimental studies have implicated microglial dysfunction as a causal factor for AD, as opposed to a merely biological response of its accumulation around AP. Additionally, many studies have suggested the importance of changes in blood-brain barrier (BBB) permeability in the pathogenesis of AD. Here we suggest how these less investigated aspects of the disease that have gained increased attention in recent years may contribute mechanistically to the development of lesions and symptoms of AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Ena Španić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|