101
|
Chaney A, Williams SR, Boutin H. In vivo molecular imaging of neuroinflammation in Alzheimer's disease. J Neurochem 2018; 149:438-451. [PMID: 30339715 PMCID: PMC6563454 DOI: 10.1111/jnc.14615] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
It has become increasingly evident that neuroinflammation plays a critical role in the pathophysiology of Alzheimer's disease (AD) and other neurodegenerative disorders. Increased glial cell activation is consistently reported in both rodent models of AD and in AD patients. Moreover, recent genome wide association studies have revealed multiple genes associated with inflammation and immunity are significantly associated with an increased risk of AD development (e.g. TREM2). Non‐invasive in vivo detection and tracking of neuroinflammation is necessary to enhance our understanding of the contribution of neuroinflammation to the initiation and progression of AD. Importantly, accurate methods of quantifying neuroinflammation may aid early diagnosis and serve as an output for therapeutic monitoring and disease management. This review details current in vivo imaging biomarkers of neuroinflammation being explored and summarizes both pre‐clinical and clinical results from molecular imaging studies investigating the role of neuroinflammation in AD, with a focus on positron emission tomography and magnetic resonance spectroscopy (MRS). ![]()
Collapse
Affiliation(s)
- Aisling Chaney
- School of Health Sciences, Division of Informatics, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK.,Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Steve R Williams
- School of Health Sciences, Division of Informatics, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Herve Boutin
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.,School of Biological Sciences, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
102
|
Song Q, Li Y, Cao Z, Liu H, Tian C, Yang Z, Qiang X, Tan Z, Deng Y. Discovery of novel 2,5-dihydroxyterephthalamide derivatives as multifunctional agents for the treatment of Alzheimer's disease. Bioorg Med Chem 2018; 26:6115-6127. [PMID: 30470598 DOI: 10.1016/j.bmc.2018.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 10/27/2022]
Abstract
A series of 2,5-dihydroxyterephthalamide derivatives were designed, synthesized and evaluated as multifunctional agents for the treatment of Alzheimer's disease. In vitro assays demonstrated that most of the derivatives exhibited good multifunctional activities. Among them, compound 9d showed the best inhibitory activity against both RatAChE and EeAChE (IC50 = 0.56 μM and 5.12 μM, respectively). Moreover, 9d exhibited excellent inhibitory effects on self-induced Aβ1-42 aggregation (IC50 = 3.05 μM) and Cu2+-induced Aβ1-42 aggregation (71.7% at 25.0 μM), and displayed significant disaggregation ability to self- and Cu2+-induced Aβ1-42 aggregation fibrils (75.2% and 77.2% at 25.0 μM, respectively). Furthermore, 9d also showed biometal chelating abilities, antioxidant activity, anti-neuroinflammatory activities and appropriate BBB permeability. These multifunctional properties highlight 9d as promising candidate for further studies directed to the development of novel drugs against AD.
Collapse
Affiliation(s)
- Qing Song
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Yan Li
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zhongcheng Cao
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Hongyan Liu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Chaoquan Tian
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Ziyi Yang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xiaoming Qiang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zhenghuai Tan
- Institute of Traditional Chinese Medicine Pharmacology and Toxicology, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, PR China
| | - Yong Deng
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
103
|
Knezevic D, Verhoeff NPL, Hafizi S, Strafella AP, Graff-Guerrero A, Rajji T, Pollock BG, Houle S, Rusjan PM, Mizrahi R. Imaging microglial activation and amyloid burden in amnestic mild cognitive impairment. J Cereb Blood Flow Metab 2018; 38:1885-1895. [PMID: 29135331 PMCID: PMC6259323 DOI: 10.1177/0271678x17741395] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amnestic mild cognitive impairment (aMCI) is defined as a transitional state between normal aging and Alzheimer's disease (AD). Given the replicated finding of increased microglial activation in AD, we sought to investigate whether microglial activation is also elevated in aMCI and whether it is related to amyloid beta (Aβ) burden in-vivo . Eleven aMCI participants and 14 healthy volunteers completed positron emission tomography (PET) scans with [18F]-FEPPA and [11C]-PIB. Given the known sensitivity in affinity of second-generation TSPO radioligands, participants were genotyped for the TSPO polymorphism and only high-affinity binders were included. Dynamic [18F]-FEPPA PET images were analyzed using the 2-tissue compartment model with arterial plasma input function. Additionally, a supplementary method, the standardized uptake value ratio (SUVR), was explored. [11C]-PIB PET images were analyzed using the Logan graphical method. aMCI participants had significantly higher [11C]-PIB binding in the cortical regions. No significant differences in [18F]-FEPPA binding were observed between aMCI participants and healthy volunteers. In the aMCI group, [18F]-FEPPA and [11C]-PIB bindings were correlated in the hippocampus. There were no correlations between our PET measures and cognition. Our findings demonstrate that while Aβ burden is evident in the aMCI stage, microglial activation may not be present.
Collapse
Affiliation(s)
- Dunja Knezevic
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Nicolaas Paul Lg Verhoeff
- 1 University of Toronto, Toronto, Ontario, Canada.,3 Baycrest Health Sciences, Toronto, Ontario, Canada
| | - Sina Hafizi
- 1 University of Toronto, Toronto, Ontario, Canada
| | - Antonio P Strafella
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,4 Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ariel Graff-Guerrero
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Tarek Rajji
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Bruce G Pollock
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sylvain Houle
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Pablo M Rusjan
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Romina Mizrahi
- 1 University of Toronto, Toronto, Ontario, Canada.,2 Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
104
|
Villa A, Klein B, Janssen B, Pedragosa J, Pepe G, Zinnhardt B, Vugts DJ, Gelosa P, Sironi L, Beaino W, Damont A, Dollé F, Jego B, Winkeler A, Ory D, Solin O, Vercouillie J, Funke U, Laner-Plamberger S, Blomster LV, Christophersen P, Vegeto E, Aigner L, Jacobs A, Planas AM, Maggi A, Windhorst AD. Identification of new molecular targets for PET imaging of the microglial anti-inflammatory activation state. Am J Cancer Res 2018; 8:5400-5418. [PMID: 30555554 PMCID: PMC6276082 DOI: 10.7150/thno.25572] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Microglia are potential targets for therapeutic intervention in neurological and neurodegenerative diseases affecting the central nervous system. In order to assess the efficacy of therapies aimed to reduce the tissue damaging activities of microglia and/or to promote the protective potential of these cells, suitable pre-clinical and clinical tools for the in vivo analysis of microglia activities and dynamics are required. The aim of this work was to identify new translational markers of the anti-inflammatory / protective state of microglia for the development of novel PET tracers. Methods: New translational markers of the anti-inflammatory/protective activation state of microglia were selected by bioinformatic approaches and were in vitro and ex vivo validated by qPCR and immunohistochemistry in rodent and human samples. Once a viable marker was identified, a novel PET tracer was developed. This tracer was subsequently confirmed by autoradiography experiments in murine and human brain tissues. Results: Here we provide evidence that P2RY12 expression increases in murine and human microglia following exposure to anti-inflammatory stimuli, and that its expression is modulated in the reparative phase of experimental and clinical stroke. We then synthesized a novel carbon-11 labeled tracer targeting P2RY12, showing increased binding in brain sections of mice treated with IL4, and low binding to brain sections of a murine stroke model and of a stroke patient. Conclusion: This study provides new translational targets for PET tracers for the anti-inflammatory/protective activation state of microglia and shows the potential of a rationale-based approach. It therefore paves the way for the development of novel non-invasive methodologies aimed to monitor the success of therapeutic approaches in various neurological diseases.
Collapse
|
105
|
Shen Z, Bao X, Wang R. Clinical PET Imaging of Microglial Activation: Implications for Microglial Therapeutics in Alzheimer's Disease. Front Aging Neurosci 2018; 10:314. [PMID: 30349474 PMCID: PMC6186779 DOI: 10.3389/fnagi.2018.00314] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/19/2018] [Indexed: 12/19/2022] Open
Abstract
In addition to extracellular β-amyloid plaques and intracellular neurofibrillary tangles, neuroinflammation has been identified as a key pathological characteristic of Alzheimer's disease (AD). Once activated, neuroinflammatory cells called microglia acquire different activation phenotypes. At the early stage of AD, activated microglia are mainly dominated by the neuroprotective and anti-inflammatory M2 phenotype. Conversely, in the later stage of AD, the excessive activation of microglia is considered detrimental and pro-inflammatory, turning into the M1 phenotype. Therapeutic strategies targeting the modulation of microglia may regulate their specific phenotype. Fortunately, with the rapid development of in vivo imaging methodologies, visualization of microglial activation has been well-explored. In this review, we summarize the critical role of activated microglia during the pathogenesis of AD and current studies concerning imaging of microglial activation in AD patients. We explore the possibilities for identifying activated microglial phenotypes with imaging techniques and highlight promising therapies that regulate the microglial phenotype in AD mice.
Collapse
Affiliation(s)
- Zhiwei Shen
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
106
|
Sejimo S, Hossain MS, Akashi K. Scallop-derived plasmalogens attenuate the activation of PKCδ associated with the brain inflammation. Biochem Biophys Res Commun 2018; 503:837-842. [DOI: 10.1016/j.bbrc.2018.06.084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 06/16/2018] [Indexed: 12/20/2022]
|
107
|
Edison P, Donat CK, Sastre M. In vivo Imaging of Glial Activation in Alzheimer's Disease. Front Neurol 2018; 9:625. [PMID: 30131755 PMCID: PMC6090997 DOI: 10.3389/fneur.2018.00625] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by memory loss and decline of cognitive function, associated with progressive neurodegeneration. While neuropathological processes like amyloid plaques and tau neurofibrillary tangles have been linked to neuronal death in AD, the precise role of glial activation on disease progression is still debated. It was suggested that neuroinflammation could occur well ahead of amyloid deposition and may be responsible for clearing amyloid, having a neuroprotective effect; however, later in the disease, glial activation could become deleterious, contributing to neuronal toxicity. Recent genetic and preclinical studies suggest that the different activation states of microglia and astrocytes are complex, not as polarized as previously thought, and that the heterogeneity in their phenotype can switch during disease progression. In the last few years, novel imaging techniques e.g., new radiotracers for assessing glia activation using positron emission tomography and advanced magnetic resonance imaging technologies have emerged, allowing the correlation of neuro-inflammatory markers with cognitive decline, brain function and brain pathology in vivo. Here we review all new imaging technology in AD patients and animal models that has the potential to serve for early diagnosis of the disease, to monitor disease progression and to test the efficacy and the most effective time window for potential anti-inflammatory treatments.
Collapse
Affiliation(s)
- Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Cornelius K Donat
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
108
|
Edison P, Brooks DJ. Role of Neuroinflammation in the Trajectory of Alzheimer’s Disease and in vivo Quantification Using PET. J Alzheimers Dis 2018; 64:S339-S351. [DOI: 10.3233/jad-179929] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Paul Edison
- Neurology Imaging Unit, Department of Medicine, Imperial College London, London, UK
| | - David J. Brooks
- Department of Nuclear Medicine, Aarhus University, Denmark
- Institute of Neuroscience, University of Newcastle upon Tyne, UK
| |
Collapse
|
109
|
Neurobiological links between depression and AD: The role of TGF-β1 signaling as a new pharmacological target. Pharmacol Res 2018; 130:374-384. [DOI: 10.1016/j.phrs.2018.02.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/03/2018] [Accepted: 02/07/2018] [Indexed: 12/19/2022]
|
110
|
Hamelin L, Lagarde J, Dorothée G, Potier MC, Corlier F, Kuhnast B, Caillé F, Dubois B, Fillon L, Chupin M, Bottlaender M, Sarazin M. Distinct dynamic profiles of microglial activation are associated with progression of Alzheimer's disease. Brain 2018; 141:1855-1870. [DOI: 10.1093/brain/awy079] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/02/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Lorraine Hamelin
- Unit of Neurology of Memory and Language, Université Paris Descartes, Sorbonne Paris Cité, Centre Hospitalier Sainte Anne, Paris, France
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Julien Lagarde
- Unit of Neurology of Memory and Language, Université Paris Descartes, Sorbonne Paris Cité, Centre Hospitalier Sainte Anne, Paris, France
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Guillaume Dorothée
- INSERM, UMRS 938, CdR Saint-Antoine, Laboratory Immune System, Neuroinflammation and Neurodegenerative Diseases, Hôpital St-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 938, CdR Saint-Antoine, Hôpital Saint-Antoine, Paris, France
| | - Marie Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Fabian Corlier
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, USA
| | - Bertrand Kuhnast
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Fabien Caillé
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Bruno Dubois
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du cerveau et la moelle (ICM), Hôpital Pitié-Salpêtrière, Boulevard de l’Hôpital, F-75013, Paris, France
- Dementia Research Center (IM2A), AP-HP, Hôpital Pitié-Salpêtrière, F-75013, Paris, France
| | - Ludovic Fillon
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du cerveau et la moelle (ICM), Hôpital Pitié-Salpêtrière, Boulevard de l’Hôpital, F-75013, Paris, France
- CATI, Centre d’Acquisition et de Traitement des Images, Paris, France
| | - Marie Chupin
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du cerveau et la moelle (ICM), Hôpital Pitié-Salpêtrière, Boulevard de l’Hôpital, F-75013, Paris, France
- CATI, Centre d’Acquisition et de Traitement des Images, Paris, France
| | - Michel Bottlaender
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
- UNIACT, Neurospin, CEA, Gif-sur-Yvette, F-91191, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Université Paris Descartes, Sorbonne Paris Cité, Centre Hospitalier Sainte Anne, Paris, France
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| |
Collapse
|
111
|
Parametric mapping using spectral analysis for 11C-PBR28 PET reveals neuroinflammation in mild cognitive impairment subjects. Eur J Nucl Med Mol Imaging 2018. [PMID: 29523926 PMCID: PMC5993844 DOI: 10.1007/s00259-018-3984-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE Neuroinflammation and microglial activation play an important role in amnestic mild cognitive impairment (MCI) and Alzheimer's disease. In this study, we investigated the spatial distribution of neuroinflammation in MCI subjects, using spectral analysis (SA) to generate parametric maps and quantify 11C-PBR28 PET, and compared these with compartmental and other kinetic models of quantification. METHODS Thirteen MCI and nine healthy controls were enrolled in this study. Subjects underwent 11C-PBR28 PET scans with arterial cannulation. Spectral analysis with an arterial plasma input function was used to generate 11C-PBR28 parametric maps. These maps were then compared with regional 11C-PBR28 VT (volume of distribution) using a two-tissue compartment model and Logan graphic analysis. Amyloid load was also assessed with 18F-Flutemetamol PET. RESULTS With SA, three component peaks were identified in addition to blood volume. The 11C-PBR28 impulse response function (IRF) at 90 min produced the lowest coefficient of variation. Single-subject analysis using this IRF demonstrated microglial activation in five out of seven amyloid-positive MCI subjects. IRF parametric maps of 11C-PBR28 uptake revealed a group-wise significant increase in neuroinflammation in amyloid-positive MCI subjects versus HC in multiple cortical association areas, and particularly in the temporal lobe. Interestingly, compartmental analysis detected group-wise increase in 11C-PBR28 binding in the thalamus of amyloid-positive MCI subjects, while Logan parametric maps did not perform well. CONCLUSIONS This study demonstrates for the first time that spectral analysis can be used to generate parametric maps of 11C-PBR28 uptake, and is able to detect microglial activation in amyloid-positive MCI subjects. IRF parametric maps of 11C-PBR28 uptake allow voxel-wise single-subject analysis and could be used to evaluate microglial activation in individual subjects.
Collapse
|
112
|
Rissanen E, Tuisku J, Vahlberg T, Sucksdorff M, Paavilainen T, Parkkola R, Rokka J, Gerhard A, Hinz R, Talbot PS, Rinne JO, Airas L. Microglial activation, white matter tract damage, and disability in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2018. [PMID: 29520366 PMCID: PMC5840890 DOI: 10.1212/nxi.0000000000000443] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective To investigate the relationship of in vivo microglial activation to clinical and MRI parameters in MS. Methods Patients with secondary progressive MS (n = 10) or relapsing-remitting MS (n = 10) and age-matched healthy controls (n = 17) were studied. Microglial activation was measured using PET and radioligand [11C](R)-PK11195. Clinical assessment and structural and quantitative MRI including diffusion tensor imaging (DTI) were performed for comparison. Results [11C](R)-PK11195 binding was significantly higher in the normal-appearing white matter (NAWM) of patients with secondary progressive vs relapsing MS and healthy controls, in the thalami of patients with secondary progressive MS vs controls, and in the perilesional area among the progressive compared with relapsing patients. Higher binding in the NAWM was associated with higher clinical disability and reduced white matter (WM) structural integrity, as shown by lower fractional anisotropy, higher mean diffusivity, and increased WM lesion load. Increasing age contributed to higher microglial activation in the NAWM among patients with MS but not in healthy controls. Conclusions PET can be used to quantitate microglial activation, which associates with MS progression. This study demonstrates that increased microglial activity in the NAWM correlates closely with impaired WM structural integrity and thus offers one rational pathologic correlate to diffusion tensor imaging (DTI) parameters.
Collapse
Affiliation(s)
- Eero Rissanen
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Jouni Tuisku
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Tero Vahlberg
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Marcus Sucksdorff
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Teemu Paavilainen
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Riitta Parkkola
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Johanna Rokka
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Alexander Gerhard
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Rainer Hinz
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Peter S Talbot
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Juha O Rinne
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| | - Laura Airas
- Turku PET Centre (E.R., J.T., M.S., J.R., J.O.R.), Division of Clinical Neurosciences (E.R., M.S., J.O.R., L.A.), Department of Biostatistics (T.V.), and Medical Imaging Centre of Southwest Finland (T.P., R.P.), Turku University Hospital and University of Turku, Finland; Division of Neuroscience and Experimental Psychology (A.G.), University of Manchester, United Kingdom; Department of Nuclear Medicine and Geriatric Medicine (A.G.), University Hospital Essen, Germany; and Wolfson Molecular Imaging Centre (R.H., P.S.T.), University of Manchester, United Kingdom
| |
Collapse
|
113
|
Knezevic D, Mizrahi R. Molecular imaging of neuroinflammation in Alzheimer's disease and mild cognitive impairment. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:123-131. [PMID: 28533150 DOI: 10.1016/j.pnpbp.2017.05.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 02/28/2017] [Accepted: 05/09/2017] [Indexed: 11/26/2022]
Abstract
Neuroinflammatory changes have been demonstrated to be an important feature of Alzheimer's disease (AD); however, the exact role of neuroinflammation and its progression during disease is still not well understood. One of the main drivers of the neuroinflammatory process are microglial cells. Positron Emission Tomography allows for the quantification of microglial activation by labelling the Translocator Protein 18kDa (TSPO), which becomes overexpressed upon activation of microglial cells. Several radioligands have been designed to target TSPO and have been studied in-vivo in AD populations. While most studies have shown important increases in TSPO binding in AD populations compared to healthy volunteers, whether the neuroinflammatory progress occurs early on or later during disease is still unclear. In order to investigate the early changes in neuroinflammation, studies have sought to investigate microglial activation in patients with mild cognitive impairment (MCI), which is defined as a transitional stage between normal aging and dementia. In this prodromal population, conflicting results have been reported with some studies reporting increased binding in MCI, while others demonstrate no differences from controls. Here we review the TSPO PET studies in AD and MCI populations and discuss the important methodological considerations of imaging microglial activation.
Collapse
Affiliation(s)
- Dunja Knezevic
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Romina Mizrahi
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| |
Collapse
|
114
|
Donat CK, Mirzaei N, Tang SP, Edison P, Sastre M. Imaging of Microglial Activation in Alzheimer's Disease by [ 11C]PBR28 PET. Methods Mol Biol 2018; 1750:323-339. [PMID: 29512083 DOI: 10.1007/978-1-4939-7704-8_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Deficits in neuronal function and synaptic plasticity in Alzheimer's disease (AD) are believed to be linked to microglial activation. A hallmark of reactive microglia is the upregulation of mitochondrial translocator protein (TSPO) expression. Positron emission tomography (PET) is a nuclear imaging technique that measures the distribution of trace doses of radiolabeled compounds in the body over time. PET imaging using the 2nd generation TSPO tracer [11C]PBR28 provides an opportunity for accurate visualization and quantification of changes in microglial density in transgenic mouse models of Alzheimer's disease (AD). Here, we describe the methodology for the in vivo use of [11C]PBR28 in AD patients and the 5XFAD transgenic mouse model of AD and compare the results against healthy individuals and wild-type controls. To confirm the results, autoradiography with [3H]PBR28 and immunochemistry was carried out in the same mouse brains. Our data shows that [11C]PBR28 is suitable as a tool for in vivo monitoring of microglial activation and may be useful to assess treatment response in future studies.
Collapse
Affiliation(s)
- Cornelius K Donat
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Nazanin Mirzaei
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Paul Edison
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Magdalena Sastre
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
115
|
Neuroinflammatory responses in Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:771-779. [PMID: 29273951 DOI: 10.1007/s00702-017-1831-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/17/2017] [Indexed: 12/21/2022]
Abstract
Neuroinflammatory responses in Alzheimer's disease (AD) are complex and not fully understood. They involve various cellular and molecular players and associate interaction between the central nervous system (CNS) and the periphery. Amyloid peptides within the senile plaques and abnormally phosphorylated tau in neurofibrillary tangles are able to initiate inflammatory responses, in brain of AD patients and in mouse models of this disease. The outcome of these responses on the pathophysiology of AD depends on several factors and can be either beneficial or detrimental. Thus, understanding the role of neuroinflammation in AD could help to develop safer and more efficient therapeutic strategies. This review discusses recent knowledge on microglia responses toward amyloid and tau pathology in AD, focusing on the role of Toll-like receptors and NOD-like receptor protein 3 (NLRP3) inflammasome activation in microglial cells.
Collapse
|
116
|
Chaney A, Bauer M, Bochicchio D, Smigova A, Kassiou M, Davies KE, Williams SR, Boutin H. Longitudinal investigation of neuroinflammation and metabolite profiles in the APP swe ×PS1 Δe9 transgenic mouse model of Alzheimer's disease. J Neurochem 2017; 144:318-335. [PMID: 29124761 PMCID: PMC5846890 DOI: 10.1111/jnc.14251] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 10/03/2017] [Accepted: 10/29/2017] [Indexed: 12/11/2022]
Abstract
There is increasing evidence linking neuroinflammation to many neurological disorders including Alzheimer's disease (AD); however, its exact contribution to disease manifestation and/or progression is poorly understood. Therefore, there is a need to investigate neuroinflammation in both health and disease. Here, we investigate cognitive decline, neuroinflammatory and other pathophysiological changes in the APPswe×PS1Δe9 transgenic mouse model of AD. Transgenic (TG) mice were compared to C57BL/6 wild type (WT) mice at 6, 12 and 18 months of age. Neuroinflammation was investigated by [18F]DPA‐714 positron emission tomography and myo‐inositol levels using 1H magnetic resonance spectroscopy (MRS) in vivo. Neuronal and cellular dysfunction was investigated by looking at N‐acetylaspartate (NAA), choline‐containing compounds, taurine and glutamate also using MRS. Cognitive decline was first observed at 12 m of age in the TG mice as assessed by working memory tests . A significant increase in [18F]DPA‐714 uptake was seen in the hippocampus and cortex of 18 m‐old TG mice when compared to age‐matched WT mice and 6 m‐old TG mice. No overall effect of gene was seen on metabolite levels; however, a significant reduction in NAA was observed in 18 m‐old TG mice when compared to WT. In addition, age resulted in a decrease in glutamate and an increase in choline levels. Therefore, we can conclude that increased neuroinflammation and cognitive decline are observed in TG animals, whereas NAA alterations occurring with age are exacerbated in the TG mice. These results support the role of neuroinflammation and metabolite alteration in AD and in ageing. ![]()
Collapse
Affiliation(s)
- Aisling Chaney
- Centre for Imaging Science, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK.,Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | - Daniela Bochicchio
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Alison Smigova
- Centre for Imaging Science, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK.,Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | | | - Karen E Davies
- Centre for Imaging Science, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Steve R Williams
- Centre for Imaging Science, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Herve Boutin
- Centre for Imaging Science, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK.,Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
117
|
Kreisl WC, Henter ID, Innis RB. Imaging Translocator Protein as a Biomarker of Neuroinflammation in Dementia. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:163-185. [PMID: 29413519 PMCID: PMC6190574 DOI: 10.1016/bs.apha.2017.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuroinflammation has long been considered a potential contributor to neurodegenerative disorders that result in dementia. Accumulation of abnormal protein aggregates in Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies is associated with the activation of microglia and astrocytes into proinflammatory states, and chronic low-level activation of glial cells likely contributes to the pathological changes observed in these and other neurodegenerative diseases. The 18kDa translocator protein (TSPO) is a key biomarker for measuring inflammation in the brain via positron emission tomography (PET). Increased TSPO density has been observed in brain tissue from patients with neurodegenerative diseases and colocalizes to activated microglia and reactive astrocytes. Several radioligands have been developed to measure TSPO density in vivo with PET, and these have been used in clinical studies of different dementia syndromes. However, TSPO radioligands have limitations, including low specific-to-nonspecific signal and differential affinity to a polymorphism on the TSPO gene, which must be taken into consideration in designing and interpreting human PET studies. Nonetheless, most PET studies have shown that increased TSPO binding is associated with various dementias, suggesting that TSPO has potential as a biomarker to further explore the role of neuroinflammation in dementia pathogenesis and may prove useful in monitoring disease progression.
Collapse
Affiliation(s)
- William C Kreisl
- Taub Institute, Columbia University Medical Center, New York, NY, United States.
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| |
Collapse
|
118
|
Let's make microglia great again in neurodegenerative disorders. J Neural Transm (Vienna) 2017; 125:751-770. [PMID: 29027011 DOI: 10.1007/s00702-017-1792-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
All of the common neurodegenerative disorders-Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prion diseases-are characterized by accumulation of misfolded proteins that trigger activation of microglia; brain-resident mononuclear phagocytes. This chronic form of neuroinflammation is earmarked by increased release of myriad cytokines and chemokines in patient brains and biofluids. Microglial phagocytosis is compromised early in the disease process, obfuscating clearance of abnormal proteins. This review identifies immune pathologies shared by the major neurodegenerative disorders. The overarching concept is that aberrant innate immune pathways can be targeted for return to homeostasis in hopes of coaxing microglia into clearing neurotoxic misfolded proteins.
Collapse
|
119
|
Wu Z, Ni J, Liu Y, Teeling JL, Takayama F, Collcutt A, Ibbett P, Nakanishi H. Cathepsin B plays a critical role in inducing Alzheimer's disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice. Brain Behav Immun 2017; 65:350-361. [PMID: 28610747 DOI: 10.1016/j.bbi.2017.06.002] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/14/2022] Open
Abstract
A number of clinical and experimental studies have revealed a strong association between periodontitis and accelerated cognitive decline in Alzheimer's disease (AD); however, the mechanism of the association is unknown. In the present study, we tested the hypothesis that cathepsin (Cat) B plays a critical role in the initiation of neuroinflammation and neural dysfunction following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis (PgLPS) in mice (1mg/kg, daily, intraperitoneally). Young (2months old) and middle-aged (12months old) wild-type (WT; C57BL/6N) or CatB-deficient (CatB-/-) mice were exposed to PgLPS daily for 5 consecutive weeks. The learning and memory function were assessed using the passive avoidance test, and the expression of amyloid precursor protein (APP), CatB, TLR2 and IL-1β was analyzed in brain tissues by immunohistochemistry and Western blotting. We found that chronic systemic exposure to PgLPS for five consecutive weeks induced learning and memory deficits with the intracellular accumulation of Aβ in neurons in the middle-aged WT mice, but not in young WT or middle-aged CatB-/- mice. PgLPS significantly increased the expression of CatB in both microglia and neurons in middle-aged WT mice, while increased expression of mature IL-1β and TLR2 was restricted to microglia in the hippocampus of middle-aged WT mice, but not in that of the middle-aged CatB-/- ones. In in vitro studies, PgLPS (1µg/ml) stimulation upregulated the mean mRNA expression of IL-1β, TLR2 and downregulated the protein levels of IκBα in the cultured MG6 microglia as well as in the primary microglia from WT mice, which were significantly inhibited by the CatB-specific inhibitor CA-074Me as well as by the primary microglia from CatB-/- mice. Furthermore, the mean mRNA expression of APP and CatB were significantly increased in the primary cultured hippocampal neurons after treatment with conditioned medium from PgLPS-treated WT primary microglia, but not after treatment with conditioned medium neutralized with anti-IL-1beta, and not after treatment with conditioned medium from PgLPS-treated CatB-/- primary microglia or with PgLPS directly. Taken together, these findings indicate that chronic systemic exposure to PgLPS induces AD-like phenotypes, including microglia-mediated neuroinflammation, intracellular Aβ accumulation in neurons and impairment of the learning and memory functions in the middle-aged mice in a CatB-dependent manner. We propose that CatB may be a therapeutic target for preventing periodontitis-associated cognitive decline in AD.
Collapse
Affiliation(s)
- Zhou Wu
- Department of Aging Science and Pharmacology, Kyushu University, Japan; OBT Research Center, Faculty of Dental Science, Kyushu University, Japan.
| | - Junjun Ni
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Yicong Liu
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Jessica L Teeling
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Fumiko Takayama
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Alex Collcutt
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Paul Ibbett
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Hiroshi Nakanishi
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| |
Collapse
|
120
|
Lagarde J, Sarazin M, Bottlaender M. In vivo PET imaging of neuroinflammation in Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:847-867. [PMID: 28516240 DOI: 10.1007/s00702-017-1731-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that neuroinflammation contributes to the pathophysiology of many neurodegenerative diseases, especially Alzheimer's disease (AD). Molecular imaging by PET may be a useful tool to assess neuroinflammation in vivo, thus helping to decipher the complex role of inflammatory processes in the pathophysiology of neurodegenerative diseases and providing a potential means of monitoring the effect of new therapeutic approaches. For this objective, the main target of PET studies is the 18 kDa translocator protein (TSPO), as it is overexpressed by activated microglia. In the present review, we describe the most widely used PET tracers targeting the TSPO, the methodological issues in tracer quantification and summarize the results obtained by TSPO PET imaging in AD, as well as in neurodegenerative disorders associated with AD, in psychiatric disorders and ageing. We also briefly describe alternative PET targets and imaging modalities to study neuroinflammation. Lastly, we question the meaning of PET imaging data in the context of a highly complex and multifaceted role of neuroinflammation in neurodegenerative diseases. This overview leads to the conclusion that PET imaging of neuroinflammation is a promising way of deciphering the enigma of the pathophysiology of AD and of monitoring the effect of new therapies.
Collapse
Affiliation(s)
- Julien Lagarde
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Michel Bottlaender
- UNIACT, NeuroSpin, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91191, Gif-sur-Yvette, France. .,Laboratoire Imagerie Moléculaire in Vivo, UMR 1023, Service Hospitalier Frédéric Joliot, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91400, Orsay, France.
| |
Collapse
|
121
|
Cerami C, Iaccarino L, Perani D. Molecular Imaging of Neuroinflammation in Neurodegenerative Dementias: The Role of In Vivo PET Imaging. Int J Mol Sci 2017; 18:ijms18050993. [PMID: 28475165 PMCID: PMC5454906 DOI: 10.3390/ijms18050993] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 11/16/2022] Open
Abstract
Neurodegeneration elicits neuroinflammatory responses to kill pathogens, clear debris and support tissue repair. Neuroinflammation is a dynamic biological response characterized by the recruitment of innate and adaptive immune system cells in the site of tissue damage. Resident microglia and infiltrating immune cells partake in the restoration of central nervous system homeostasis. Nevertheless, their activation may shift to chronic and aggressive responses, which jeopardize neuron survival and may contribute to the disease process itself. Positron Emission Tomography (PET) molecular imaging represents a unique tool contributing to in vivo investigating of neuroinflammatory processes in patients. In the present review, we first provide an overview on the molecular basis of neuroinflammation in neurodegenerative diseases with emphasis on microglia activation, astrocytosis and the molecular targets for PET imaging. Then, we review the state-of-the-art of in vivo PET imaging for neuroinflammation in dementia conditions associated with different proteinopathies, such as Alzheimer’s disease, frontotemporal lobar degeneration and Parkinsonian spectrum.
Collapse
Affiliation(s)
- Chiara Cerami
- Clinical Neuroscience Department, San Raffaele Turro Hospital, Milan 20121-20162, Italy.
- Division of Neuroscience, San Raffaele Scientific Institute, Milan 20121-20162, Italy.
| | - Leonardo Iaccarino
- Division of Neuroscience, San Raffaele Scientific Institute, Milan 20121-20162, Italy.
- Faculty of Psychology and Molecular Medicine Doctoral Course, Vita-Salute San Raffaele University, Milan 20121-20162, Italy.
| | - Daniela Perani
- Division of Neuroscience, San Raffaele Scientific Institute, Milan 20121-20162, Italy.
- Faculty of Psychology and Molecular Medicine Doctoral Course, Vita-Salute San Raffaele University, Milan 20121-20162, Italy.
- Nuclear Medicine Unit, San Raffaele Hospital, Milan 20121-20162, Italy.
| |
Collapse
|
122
|
Bao W, Jia H, Finnema S, Cai Z, Carson RE, Huang YH. PET Imaging for Early Detection of Alzheimer's Disease: From Pathologic to Physiologic Biomarkers. PET Clin 2017; 12:329-350. [PMID: 28576171 DOI: 10.1016/j.cpet.2017.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article describes the application of various PET imaging agents in the investigation and diagnosis of Alzheimer's disease (AD), including radiotracers for pathologic biomarkers of AD such as β-amyloid deposits and tau protein aggregates, and the neuroinflammation biomarker 18 kDa translocator protein, as well as physiologic biomarkers, such as cholinergic receptors, glucose metabolism, and the synaptic density biomarker synaptic vesicle glycoprotein 2A. Potential of these biomarkers for early AD diagnosis is also assessed.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, No. 518, East Wuzhong Road, Xuhui District, Shanghai 200235, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Beijing 10075, China
| | - Sjoerd Finnema
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA.
| |
Collapse
|
123
|
Iaccarino L, Moresco RM, Presotto L, Bugiani O, Iannaccone S, Giaccone G, Tagliavini F, Perani D. An In Vivo 11C-(R)-PK11195 PET and In Vitro Pathology Study of Microglia Activation in Creutzfeldt-Jakob Disease. Mol Neurobiol 2017; 55:2856-2868. [PMID: 28455699 DOI: 10.1007/s12035-017-0522-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/06/2017] [Indexed: 01/08/2023]
Abstract
Microgliosis is part of the immunobiology of Creutzfeldt-Jakob disease (CJD). This is the first report using 11C-(R)-PK11195 PET imaging in vivo to measure 18 kDa translocator protein (TSPO) expression, indexing microglia activation, in symptomatic CJD patients, followed by a postmortem neuropathology comparison. One genetic CJD (gCJD) patient, two sporadic CJD (sCJD) patients, one variant CJD (vCJD) patient (mean ± SD age, 47.50 ± 15.95 years), and nine healthy controls (mean ± SD age, 44.00 ± 11.10 years) were included in the study. TSPO binding potentials were estimated using clustering and parametric analyses of reference regions. Statistical comparisons were run at the regional and at the voxel-wise levels. Postmortem evaluation measured scrapie prion protein (PrPSc) immunoreactivity, neuronal loss, spongiosis, astrogliosis, and microgliosis. 11C-(R)-PK11195-PET showed a significant TSPO overexpression at the cortical level in the two sCJD patients, as well as thalamic and cerebellar involvement; very limited parieto-occipital activation in the gCJD case; and significant increases at the subcortical level in the thalamus, basal ganglia, and midbrain and in the cerebellum in the vCJD brain. Along with misfolded prion deposits, neuropathology in all patients revealed neuronal loss, spongiosis and astrogliosis, and a diffuse cerebral and cerebellar microgliosis which was particularly dense in thalamic and basal ganglia structures in the vCJD brain. These findings confirm significant microgliosis in CJD, which was variably modulated in vivo and more diffuse at postmortem evaluation. Thus, TSPO overexpression in microglia activation, topography, and extent can vary in CJD subtypes, as shown in vivo, possibly related to the response to fast apoptotic processes, but reaches a large amount at the final disease course.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy.,IBFM-CNR, Via F.lli Cervi 93, Segrate, 20090, Milan, Italy.,Department of Health Sciences, University of Milan Bicocca, Piazza dell'Ateneo Nuovo, 1, 20126, Milan, Italy
| | - Luca Presotto
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Orso Bugiani
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Sandro Iannaccone
- Neurological Rehabilitation Unit, Clinical Neurosciences Department, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgio Giaccone
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Fabrizio Tagliavini
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy. .,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy. .,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
124
|
Dupont AC, Largeau B, Santiago Ribeiro MJ, Guilloteau D, Tronel C, Arlicot N. Translocator Protein-18 kDa (TSPO) Positron Emission Tomography (PET) Imaging and Its Clinical Impact in Neurodegenerative Diseases. Int J Mol Sci 2017; 18:ijms18040785. [PMID: 28387722 PMCID: PMC5412369 DOI: 10.3390/ijms18040785] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
In vivo exploration of activated microglia in neurodegenerative diseases is achievable by Positron Emission Tomography (PET) imaging, using dedicated radiopharmaceuticals targeting the translocator protein-18 kDa (TSPO). In this review, we emphasized the major advances made over the last 20 years, thanks to TSPO PET imaging, to define the pathophysiological implication of microglia activation and neuroinflammation in neurodegenerative diseases, including Parkinson’s disease, Huntington’s disease, dementia, amyotrophic lateral sclerosis, multiple sclerosis, and also in psychiatric disorders. The extent and upregulation of TSPO as a molecular biomarker of activated microglia in the human brain is now widely documented in these pathologies, but its significance, and especially its protective or deleterious action regarding the disease’s stage, remains under debate. Thus, we exposed new and plausible suggestions to enhance the contribution of TSPO PET imaging for biomedical research by exploring microglia’s role and interactions with other cells in brain parenchyma. Multiplex approaches, associating TSPO PET radiopharmaceuticals with other biomarkers (PET imaging of cellular metabolism, neurotransmission or abnormal protein aggregates, but also other imaging modalities, and peripheral cytokine levels measurement and/or metabolomics analysis) was considered. Finally, the actual clinical impact of TSPO PET imaging as a routine biomarker of neuroinflammation was put into perspective regarding the current development of diagnostic and therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne-Claire Dupont
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | | | - Maria Joao Santiago Ribeiro
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Denis Guilloteau
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Claire Tronel
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Nicolas Arlicot
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| |
Collapse
|
125
|
Baldacci F, Lista S, Cavedo E, Bonuccelli U, Hampel H. Diagnostic function of the neuroinflammatory biomarker YKL-40 in Alzheimer’s disease and other neurodegenerative diseases. Expert Rev Proteomics 2017; 14:285-299. [DOI: 10.1080/14789450.2017.1304217] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Simone Lista
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Enrica Cavedo
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
- IRCCS Istituto Centro San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| |
Collapse
|
126
|
Schain M, Kreisl WC. Neuroinflammation in Neurodegenerative Disorders—a Review. Curr Neurol Neurosci Rep 2017; 17:25. [DOI: 10.1007/s11910-017-0733-2] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
127
|
|
128
|
Iwahara N, Hisahara S, Kawamata J, Matsumura A, Yokokawa K, Saito T, Fujikura M, Manabe T, Suzuki H, Matsushita T, Suzuki S, Shimohama S. Role of Suppressor of Cytokine Signaling 3 (SOCS3) in Altering Activated Microglia Phenotype in APPswe/PS1dE9 Mice. J Alzheimers Dis 2017; 55:1235-1247. [PMID: 27814300 DOI: 10.3233/jad-160887] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In response to changes of the central nervous system environment, microglia are capable of acquiring diverse phenotypes for cytotoxic or immune regulation and resolution of injury. Alzheimer's disease (AD) pathology also induces several microglial activations, resulting in production of pro-inflammatory cytokines and reactive oxygen species or clearance of amyloid-β (Aβ) through phagocytosis. We previously demonstrated that microglial activation and increase in oxidative stress started from the middle age in APPswe/PS1dE9 mice, and hypothesized that M1 activation occurs in middle-aged AD mice by Aβ stimulation. In the present study, we analyzed in vivo expressions of pro-inflammatory cytokines (M1 microglial markers), M2 microglial markers, and suppressor of cytokine signaling (SOCS) family, and examined the microglial phenotypic profile in APPswe/PS1dE9 mice. Then we compared the in vitro gene expression patterns of Aβ- and lipopolysaccharide (LPS)-stimulated primary-cultured microglia. Microglia in APPswe/PS1dE9 mice exhibited an M1-like phenotype, expressing tumor necrosis factor α (TNFα) but not interleukin 6 (IL6). Aβ-stimulated primary-cultured microglia also expressed TNFα but not IL6, whereas LPS-stimulated primary-cultured microglia expressed both pro-inflammatory cytokines. Furthermore, both microglia in APPswe/PS1dE9 mice and Aβ-stimulated primary-cultured microglia expressed SOCS3. Reduction of SOCS3 expression in Aβ-challenged primary-cultured microglia resulted in upregulation of IL6 expression. Our findings indicate that SOCS3 suppresses complete polarization to M1 phenotype through blocking IL6 production, and Aβ-challenged primary-cultured microglia replicate the in vivo gene expression pattern of microglia in APPswe/PS1dE9 mice. Aβ may induce the M1-like phenotype through blocking of IL6 by SOCS3.
Collapse
Affiliation(s)
- Naotoshi Iwahara
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Shin Hisahara
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Jun Kawamata
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Akihiro Matsumura
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Kazuki Yokokawa
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Taro Saito
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Mai Fujikura
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Tatsuo Manabe
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Hiromi Suzuki
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takashi Matsushita
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan.,Department of Pharmacology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Syuuichirou Suzuki
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Shun Shimohama
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| |
Collapse
|
129
|
Boondam Y, Cheepsunthorn P. Original article. Patterns of microglial innate immune responses elicited by amyloid β1–42 and lipopolysaccharide: the similarities of the differences. ASIAN BIOMED 2017. [DOI: 10.5372/1905-7415.0802.299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract
Background: As part of their innate immune response to changes in the central nervous system environment, normally quiescent microglia become activated and increase expression of pattern recognition receptors, scavenger receptors, and production of inflammatory cytokines, proteinases, reactive oxygen species (ROS), and free radicals. These molecules have been implicated in the pathogenesis and progression of several neurodegenerative disorders including Alzheimer disease (AD).
Objective: We compared patterns of microglial innate immune responses elicited by nonfibrillar amyloid β peptide (nfAβ1-42) to those elicited by lipopolysaccharide (LPS).
Methods: Murine BV-2 microglial cells were exposed to either nfAβ1-42 or LPS for 12 h. Then, total RNA from each condition was isolated and expression levels of Toll-like receptor (TLR)-4, scavenger receptor class A (SRMARCO) and class B (SR-BI), CD36, and matrix metalloproteinase (MMP)-9 were determined by reverse transcription-quantitative real-time polymerase chain reaction. The amount of hydrogen peroxide (H2O2) and nitric oxide (NO) in the cell-free supernatant at 24 h were determined using 10-acetyl-3,7-dihydroxyphenoxazine (Amplex Red) and Griess reagent, respectively.
Results: nfAβ1-42 and LPS significantly increased expression of TLR-4, SR-MARCO, CD36, and MMP-9 and production of H2O2 and NO in BV-2 microglial cells compared with that of unstimulated cells. However, expression of SR-BI was significantly induced only when the cells were exposed to nfAβ1-42.
Conclusion: These findings indicate that pattern of microglial innate immune responses elicited by nfAβ1-42 overlap with that elicited by LPS and suggest a specific role of microglial SR-BI expression in AD pathogenesis.
Collapse
Affiliation(s)
- Yingrak Boondam
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Poonlarp Cheepsunthorn
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
130
|
Ardura-Fabregat A, Boddeke EWGM, Boza-Serrano A, Brioschi S, Castro-Gomez S, Ceyzériat K, Dansokho C, Dierkes T, Gelders G, Heneka MT, Hoeijmakers L, Hoffmann A, Iaccarino L, Jahnert S, Kuhbandner K, Landreth G, Lonnemann N, Löschmann PA, McManus RM, Paulus A, Reemst K, Sanchez-Caro JM, Tiberi A, Van der Perren A, Vautheny A, Venegas C, Webers A, Weydt P, Wijasa TS, Xiang X, Yang Y. Targeting Neuroinflammation to Treat Alzheimer's Disease. CNS Drugs 2017; 31:1057-1082. [PMID: 29260466 PMCID: PMC5747579 DOI: 10.1007/s40263-017-0483-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Over the past few decades, research on Alzheimer's disease (AD) has focused on pathomechanisms linked to two of the major pathological hallmarks of extracellular deposition of beta-amyloid peptides and intra-neuronal formation of neurofibrils. Recently, a third disease component, the neuroinflammatory reaction mediated by cerebral innate immune cells, has entered the spotlight, prompted by findings from genetic, pre-clinical, and clinical studies. Various proteins that arise during neurodegeneration, including beta-amyloid, tau, heat shock proteins, and chromogranin, among others, act as danger-associated molecular patterns, that-upon engagement of pattern recognition receptors-induce inflammatory signaling pathways and ultimately lead to the production and release of immune mediators. These may have beneficial effects but ultimately compromise neuronal function and cause cell death. The current review, assembled by participants of the Chiclana Summer School on Neuroinflammation 2016, provides an overview of our current understanding of AD-related immune processes. We describe the principal cellular and molecular players in inflammation as they pertain to AD, examine modifying factors, and discuss potential future therapeutic targets.
Collapse
Affiliation(s)
- A. Ardura-Fabregat
- grid.5963.9Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - E. W. G. M. Boddeke
- 0000 0004 0407 1981grid.4830.fDepartment of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A. Boza-Serrano
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - S. Brioschi
- grid.5963.9Department of Psychiatry and Psychotherapy, Medical Center University of Freiburg, Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - S. Castro-Gomez
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Ceyzériat
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Dansokho
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - T. Dierkes
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dBiomedical Centre, Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - G. Gelders
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Michael T. Heneka
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - L. Hoeijmakers
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - L. Iaccarino
- grid.15496.3fVita-Salute San Raffaele University, Milan, Italy ,0000000417581884grid.18887.3eIn Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S. Jahnert
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Kuhbandner
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - G. Landreth
- 0000 0001 2287 3919grid.257413.6Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - N. Lonnemann
- 0000 0001 1090 0254grid.6738.aDepartment of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - R. M. McManus
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Paulus
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - K. Reemst
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - J. M. Sanchez-Caro
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Tiberi
- grid.6093.cBio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - A. Van der Perren
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - A. Vautheny
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Venegas
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - A. Webers
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - P. Weydt
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - T. S. Wijasa
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - X. Xiang
- 0000 0004 1936 973Xgrid.5252.0Biomedical Center (BMC), Biochemistry, Ludwig-Maximilians-University Munich, 81377 Munich, Germany ,0000 0004 1936 973Xgrid.5252.0Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University, Munich, 82152 Munich, Germany
| | - Y. Yang
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| |
Collapse
|
131
|
Sochocka M, Diniz BS, Leszek J. Inflammatory Response in the CNS: Friend or Foe? Mol Neurobiol 2016; 54:8071-8089. [PMID: 27889895 PMCID: PMC5684251 DOI: 10.1007/s12035-016-0297-1] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/09/2016] [Indexed: 12/19/2022]
Abstract
Inflammatory reactions could be both beneficial and detrimental to the brain, depending on strengths of their activation in various stages of neurodegeneration. Mild activation of microglia and astrocytes usually reveals neuroprotective effects and ameliorates early symptoms of neurodegeneration; for instance, released cytokines help maintain synaptic plasticity and modulate neuronal excitability, and stimulated toll-like receptors (TLRs) promote neurogenesis and neurite outgrowth. However, strong activation of glial cells gives rise to cytokine overexpression/dysregulation, which accelerates neurodegeneration. Altered mutual regulation of p53 protein, a major tumor suppressor, and NF-κB, the major regulator of inflammation, seems to be crucial for the shift from beneficial to detrimental effects of neuroinflammatory reactions in neurodegeneration. Therapeutic intervention in the p53-NF-κB axis and modulation of TLR activity are future challenges to cope with neurodegeneration.
Collapse
Affiliation(s)
- Marta Sochocka
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Breno Satler Diniz
- Department of Psychiatry and Behavioral Sciences, and The Consortium on Aging, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, Wybrzeże L. Pasteura 10, 50-367, Wroclaw, Poland.
| |
Collapse
|
132
|
Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol Neurobiol 2016; 54:7567-7584. [DOI: 10.1007/s12035-016-0245-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
|
133
|
Yamanaka D, Kawano T, Nishigaki A, Aoyama B, Tateiwa H, Shigematsu-Locatelli M, Locatelli FM, Yokoyama M. Preventive effects of dexmedetomidine on the development of cognitive dysfunction following systemic inflammation in aged rats. J Anesth 2016; 31:25-35. [DOI: 10.1007/s00540-016-2264-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/02/2016] [Indexed: 01/14/2023]
|
134
|
Mazzitelli S, Filipello F, Rasile M, Lauranzano E, Starvaggi-Cucuzza C, Tamborini M, Pozzi D, Barajon I, Giorgino T, Natalello A, Matteoli M. Amyloid-β 1-24 C-terminal truncated fragment promotes amyloid-β 1-42 aggregate formation in the healthy brain. Acta Neuropathol Commun 2016; 4:110. [PMID: 27724899 PMCID: PMC5057504 DOI: 10.1186/s40478-016-0381-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/24/2016] [Indexed: 01/21/2023] Open
Abstract
Substantial data indicate that amyloid-β (Aβ), the major component of senile plaques, plays a central role in Alzheimer’s Disease and indeed the assembly of naturally occurring amyloid peptides into cytotoxic aggregates is linked to the disease pathogenesis. Although Aβ42 is a highly aggregating form of Aβ, the co-occurrence of shorter Aβ peptides might affect the aggregation potential of the Aβ pool. In this study we aimed to assess whether the structural behavior of human Aβ42 peptide inside the brain is influenced by the concomitant presence of N-terminal fragments produced by the proteolytic activity of glial cells. We show that the occurrence of the human C-terminal truncated 1–24 Aβ fragment impairs Aβ42 clearance through blood brain barrier and promotes the formation of Aβ42 aggregates even in the healthy brain. By showing that Aβ1-24 has seeding properties for aggregate formation in intracranially injected wild type mice, our study provide the proof-of-concept that peptides produced upon Aβ42 cleavage by activated glial cells may cause phenotypic defects even in the absence of genetic mutations associated with Alzheimer’s Disease, possibly contributing to the development of the sporadic form of the pathology.
Collapse
|
135
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
136
|
Villemagne VL, Chételat G. Neuroimaging biomarkers in Alzheimer's disease and other dementias. Ageing Res Rev 2016; 30:4-16. [PMID: 26827785 DOI: 10.1016/j.arr.2016.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/16/2022]
Abstract
In vivo imaging of β-amyloid (Aβ) has transformed the assessment of Aβ pathology and its changes over time, extending our insight into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional or global Aβ burden in the brain. This knowledge is essential for therapeutic trial recruitment and for the evaluation of anti-Aβ treatments. Although cross sectional evaluation of Aβ burden does not strongly correlate with cognitive impairment, it does correlate with cognitive (especially memory) decline and with a higher risk for conversion to AD in the aging population and MCI subjects. This suggests that Aβ deposition is a protracted pathological process starting well before the onset of symptoms. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of Aβ are required to confirm this hypothesis and further elucidate the role of Aβ deposition in the course of Alzheimer's disease.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Victoria 3084, Australia; Department of Medicine, University of Melbourne, Austin Health, Victoria 3084, Australia; The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France.
| | - Gaël Chételat
- The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France; Université de Caen Basse-Normandie, Unité Mixte de Recherche (UMR), S1077 Caen, France; Ecole Pratique des Hautes Etudes, UMR-S1077, 14000 Caen, France; Unité 1077, Centre Hospitalier Universitaire de Caen, 14000 Caen, France
| |
Collapse
|
137
|
Su F, Bai F, Zhou H, Zhang Z. Reprint of: Microglial toll-like receptors and Alzheimer's disease. Brain Behav Immun 2016; 55:166-178. [PMID: 27255539 DOI: 10.1016/j.bbi.2016.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 01/04/2023] Open
Abstract
Microglial activation represents an important pathological hallmark of Alzheimer's disease (AD), and emerging data highlight the involvement of microglial toll-like receptors (TLRs) in the course of AD. TLRs have been observed to exert both beneficial and detrimental effects on AD-related pathologies, and transgenic animal models have provided direct and credible evidence for an association between TLRs and AD. Moreover, analyses of genetic polymorphisms have suggested interactions between genetic polymorphisms in TLRs and AD risk, further supporting the hypothesis that TLRs are involved in AD. In this review, we summarize the key evidence in this field. Future studies should focus on exploring the mechanisms underlying the potential roles of TLRs in AD.
Collapse
Affiliation(s)
- Fan Su
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Feng Bai
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Hong Zhou
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
138
|
Higuchi M, Ji B, Maeda J, Sahara N, Suhara T. In vivoimaging of neuroinflammation in Alzheimer's disease. ACTA ACUST UNITED AC 2016. [DOI: 10.1111/cen3.12308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology; Chiba Japan
| | - Bin Ji
- National Institutes for Quantum and Radiological Science and Technology; Chiba Japan
| | - Jun Maeda
- National Institutes for Quantum and Radiological Science and Technology; Chiba Japan
| | - Naruhiko Sahara
- National Institutes for Quantum and Radiological Science and Technology; Chiba Japan
| | - Tetsuya Suhara
- National Institutes for Quantum and Radiological Science and Technology; Chiba Japan
| |
Collapse
|
139
|
Calsolaro V, Edison P. Neuroinflammation in Alzheimer's disease: Current evidence and future directions. Alzheimers Dement 2016; 12:719-32. [DOI: 10.1016/j.jalz.2016.02.010] [Citation(s) in RCA: 738] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 02/14/2016] [Accepted: 02/25/2016] [Indexed: 01/19/2023]
Affiliation(s)
| | - Paul Edison
- Neurology Imaging Unit; Imperial College London; UK
| |
Collapse
|
140
|
Kreisl WC, Lyoo CH, Liow JS, Wei M, Snow J, Page E, Jenko KJ, Morse CL, Zoghbi SS, Pike VW, Turner RS, Innis RB. (11)C-PBR28 binding to translocator protein increases with progression of Alzheimer's disease. Neurobiol Aging 2016; 44:53-61. [PMID: 27318133 DOI: 10.1016/j.neurobiolaging.2016.04.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 02/08/2023]
Abstract
This longitudinal study sought to determine whether the 18 kDa translocator protein (TSPO), a marker of neuroinflammation, increases over time in Alzheimer's disease. Positron emission tomography imaging with the TSPO radioligand (11)C-PBR28 was performed at baseline and after a median follow-up of 2.7 years in 14 amyloid-positive patients and 8 amyloid-negative controls. Patients had a greater increase in TSPO binding than controls in inferior parietal lobule, precuneus, occipital cortex, hippocampus, entorhinal cortex, and combined middle and inferior temporal cortex. TSPO binding in temporoparietal regions increased from 3.9% to 6.3% per annum in patients, but ranged from -0.5% to 1% per annum in controls. The change in TSPO binding correlated with cognitive worsening on clinical dementia rating scale-sum of boxes and reduced cortical volume. The annual rate of increased TSPO binding in temporoparietal regions was about 5-fold higher in patients with clinical progression (n = 9) compared with those who did not progress (n = 5). TSPO may serve as a biomarker of Alzheimer's progression and response to anti-inflammatory therapies.
Collapse
Affiliation(s)
- William C Kreisl
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| | - Chul Hyoung Lyoo
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Monica Wei
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Joseph Snow
- Office of the Clinical Director, National Institute of Mental Health, Bethesda, MD, USA
| | - Emily Page
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Kimberly J Jenko
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - R Scott Turner
- Memory Disorders Program, Georgetown University, Washington, DC, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
141
|
Wang HL, Liu H, Xue ZG, Liao QW, Fang H. Minocycline attenuates post-operative cognitive impairment in aged mice by inhibiting microglia activation. J Cell Mol Med 2016; 20:1632-9. [PMID: 27061744 PMCID: PMC4988280 DOI: 10.1111/jcmm.12854] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/26/2016] [Indexed: 11/29/2022] Open
Abstract
Although it is known that isoflurane exposure or surgery leads to post‐operative cognitive dysfunction in aged rodents, there are few clinical interventions and treatments available to prevent this disorder. Minocycline (MINO) produces neuroprotection from several neurodegenerative diseases and various experimental animal models. Therefore, we set out to investigate the effects of MINO pre‐treatment on isoflurane or surgery induced cognitive impairment in aged mice by assessing the hippocampal‐dependent spatial memory performance using the Morris water maze task. Hippocampal tissues were isolated from mice and evaluated by Western blot analysis, immunofluorescence procedures and protein array system. Our results elucidate that MINO down‐regulated the isoflurane‐induced and surgery‐induced enhancement in the protein levels of pro‐inflammatory cytokine tumour necrosis factor alpha, interleukin (IL)‐1β, interferon‐γ and microglia marker Iba‐1, and up‐regulated protein levels of the anti‐inflammatory cytokine IL‐4 and IL‐10. These findings suggest that pre‐treatment with MINO attenuated isoflurane or surgery induced cognitive impairment by inhibiting the overactivation of microglia in aged mice.
Collapse
Affiliation(s)
- Hui-Lin Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hua Liu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhang-Gang Xue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing-Wu Liao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Anesthesiology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
142
|
Lauterbach EC. Repurposing psychiatric medicines to target activated microglia in anxious mild cognitive impairment and early Parkinson's disease. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2016; 5:29-51. [PMID: 27073741 PMCID: PMC4788730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/19/2016] [Indexed: 06/05/2023]
Abstract
Anxiety is common in the Mild Cognitive Impairment (MCI) stage of Alzheimer's disease (AD) and the pre-motor stages of Parkinson's disease (PD). A concomitant and possible cause of this anxiety is microglial activation, also considered a key promoter of neurodegeneration in MCI and early PD via inflammatory mechanisms and the generation of degenerative proinflammatory cytokines. Psychiatric disorders, prevalent in AD and PD, are often treated with psychiatric drugs (psychotropics), raising the question of whether psychotropics might therapeutically affect microglial activation, MCI, and PD. The literature of common psychotropics used in treating psychiatric disorders was reviewed for preclinical and clinical findings regarding microglial activation. Findings potentially compatible with reduced microglial activation or reduced microglial inflammogen release were evident for: antipsychotics including neuroleptics (chlorpromazine, thioridazine, loxapine) and atypicals (aripiprazole, olanzapine, quetiapine, risperidone, ziprasidone); mood stabilizers (carbamazepine, valproate, lithium); antidepressants including tricyclics (amitriptyline, clomipramine, imipramine, nortriptyline), SSRIs (citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline), venlafaxine, and bupropion; benzodiazepine anxiolytics (clonazepam, diazepam); cognitive enhancers (donepezil, galantamine, memantine); and other drugs (dextromethorphan, quinidine, amantadine). In contrast, pramipexole and methylphenidate might promote microglial activation. The most promising replicated findings of reduced microglial activation are for quetiapine, valproate, lithium, fluoxetine, donepezil, and memantine but further study is needed and translation of their microglial effects to human disease still requires investigation. In AD-relevant models, risperidone, valproate, lithium, fluoxetine, bupropion, donepezil, and memantine have therapeutic microglial effects in need of replication. Limited clinical data suggest some support for lithium and donepezil in reducing MCI progression, but other drugs have not been studied. In PD-relevant models, lamotrigine, valproate, fluoxetine, dextromethorphan, and amantadine have therapeutic microglial effects whereas methylphenidate induced microglial activation and pramipexole promoted NO release. Clinical data limited to pramipexole do not as of yet indicate faster progression of early PD while the other drugs remain to be investigated. These tantalizing psychotropic neuroprotective findings now invite replication and evidence in AD-and PD-specific models under chronic administration, followed by consideration for clinical trials in MCI and early stage PD. Psychiatric features in early disease may provide opportunities for clinical studies that also employ microglial PET biomarkers.
Collapse
Affiliation(s)
- Edward C Lauterbach
- Department of Psychiatry and Behavioral Sciences, Mercer University School of Medicine 655 First Street, Macon, Georgia, 31201, USA
| |
Collapse
|
143
|
Jawaid A, Krajewska J, Pawliczak F, Kandra V, Schulz PE. A Macro Role for Microglia in Poststroke Depression. J Am Geriatr Soc 2016; 64:459-61. [DOI: 10.1111/jgs.13974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ali Jawaid
- Brain Research Institute; Swiss Federal Institute of Technology; University of Zurich; Zurich Switzerland
| | | | | | | | - Paul E. Schulz
- Department of Neurology; University of Texas Health Science Center at Houston; Houston Texas
| |
Collapse
|
144
|
Chang YT, Huang CW, Chen NC, Lin KJ, Huang SH, Chang YH, Hsu SW, Chang WN, Lui CC, Hsu CW, Chang CC. Prefrontal Lobe Brain Reserve Capacity with Resistance to Higher Global Amyloid Load and White Matter Hyperintensity Burden in Mild Stage Alzheimer's Disease. PLoS One 2016; 11:e0149056. [PMID: 26872386 PMCID: PMC4752238 DOI: 10.1371/journal.pone.0149056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Amyloid deposition and white matter lesions (WMLs) in Alzheimer's disease (AD) are both considered clinically significant while a larger brain volume is thought to provide greater brain reserve (BR) against these pathological effects. This study identified the topography showing BR in patients with mild AD and explored the clinical balances among BR, amyloid, and WMLs burden. METHODS Thirty patients with AD were enrolled, and AV-45 positron emission tomography was conducted to measure the regional standardized uptake value ratio (SUVr) in 8 cortical volumes-of- interests (VOIs). The quantitative WMLs burden was measured from magnetic resonance imaging while the normalized VOIs volumes represented BR in this study. The cognitive test represented major clinical correlates. RESULTS Significant correlations between the prefrontal volume and global (r = 0.470, p = 0.024), but not regional (r = 0.264, p = 0.223) AV-45 SUVr were found. AD patients having larger regional volume in the superior- (r = 0.572, p = 0.004), superior medial- (r = 0.443, p = 0.034), and middle-prefrontal (r = 0.448, p = 0.032) regions had higher global AV-45 SUVr. For global WML loads, the prefrontal (r = -0.458, p = 0.019) and hippocampal volume (r = -0.469, p = 0.016) showed significant correlations while the prefrontal (r = -0.417, p = 0.043) or hippocampal volume (r = -0.422, p = 0.04) also predicted better composite memory scores. There were no interactions between amyloid SUVr and WML loads on the prefrontal volume. CONCLUSIONS BR of the prefrontal region might modulate the adverse global pathological burden caused by amyloid deposition. While prefrontal volume positively associated with hippocampal volume, WMLs had an adverse impact on the hippocampal volume that predicts memory performance in mild stage AD.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Wei Huang
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Nai-Ching Chen
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Hua Huang
- Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Hsiang Chang
- Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Wei Hsu
- Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Neng Chang
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chun-Chung Lui
- Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Che-Wei Hsu
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chiung-Chih Chang
- Cognition and Aging Center, Departments of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
145
|
Su F, Bai F, Zhou H, Zhang Z. Microglial toll-like receptors and Alzheimer's disease. Brain Behav Immun 2016; 52:187-198. [PMID: 26526648 DOI: 10.1016/j.bbi.2015.10.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 02/08/2023] Open
Abstract
Microglial activation represents an important pathological hallmark of Alzheimer's disease (AD), and emerging data highlight the involvement of microglial toll-like receptors (TLRs) in the course of AD. TLRs have been observed to exert both beneficial and detrimental effects on AD-related pathologies, and transgenic animal models have provided direct and credible evidence for an association between TLRs and AD. Moreover, analyses of genetic polymorphisms have suggested interactions between genetic polymorphisms in TLRs and AD risk, further supporting the hypothesis that TLRs are involved in AD. In this review, we summarize the key evidence in this field. Future studies should focus on exploring the mechanisms underlying the potential roles of TLRs in AD.
Collapse
Affiliation(s)
- Fan Su
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Feng Bai
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Hong Zhou
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
146
|
Ye M, Chung HS, Lee C, Yoon MS, Yu AR, Kim JS, Hwang DS, Shim I, Bae H. Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer's disease. J Neuroinflammation 2016; 13:10. [PMID: 26772975 PMCID: PMC4715334 DOI: 10.1186/s12974-016-0476-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/05/2016] [Indexed: 11/10/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a severe neuroinflammatory disease. CD4+Foxp3+ regulatory T cells (Tregs) modulate various inflammatory diseases via suppressing Th cell activation. There are increasing evidences that Tregs have beneficial roles in neurodegenerative diseases. Previously, we found the population of Treg cells was significantly increased by bee venom phospholipase A2 (bvPLA2) treatment in vivo and in vitro. Methods To examine the effects of bvPLA2 on AD, bvPLA2 was administered to 3xTg-AD mice, mouse model of Alzheimer’s disease. The levels of amyloid beta (Aβ) deposits in the hippocampus, glucose metabolism in the brain, microglia activation, and CD4+ T cell infiltration were analyzed to evaluate the neuroprotective effect of bvPLA2. Results bvPLA2 treatment significantly enhanced the cognitive function of the 3xTg-AD mice and increased glucose metabolism, as assessed with 18F-2 fluoro-2-deoxy-D-glucose ([F-18] FDG) positron emission tomography (PET). The levels of Aβ deposits in the hippocampus were dramatically decreased by bvPLA2 treatment. This neuroprotective effect of bvPLA2 was associated with microglial deactivation and reduction in CD4+ T cell infiltration. Interestingly, the neuroprotective effects of bvPLA2 were abolished in Treg-depleted mice. Conclusions The present studies strongly suggest that the increase of Treg population by bvPLA2 treatment might inhibit progression of AD in the 3xTg AD mice.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Hwan-Suck Chung
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea.
| | - Chanju Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Moon Sik Yoon
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - A Ram Yu
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, University of Science and Technology, #215-4 Gongneug-dong, Nowon-ku, Seoul, 139-241, Republic of Korea.
| | - Jin Su Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, University of Science and Technology, #215-4 Gongneug-dong, Nowon-ku, Seoul, 139-241, Republic of Korea.
| | - Deok-Sang Hwang
- Department of Obstetrics and Gynecology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medical Science Graduate School, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
147
|
Masdeu JC, Pascual B. Genetic and degenerative disorders primarily causing dementia. HANDBOOK OF CLINICAL NEUROLOGY 2016; 135:525-564. [PMID: 27432682 DOI: 10.1016/b978-0-444-53485-9.00026-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuroimaging comprises a powerful set of instruments to diagnose the different causes of dementia, clarify their neurobiology, and monitor their treatment. Magnetic resonance imaging (MRI) depicts volume changes with neurodegeneration and inflammation, as well as abnormalities in functional and structural connectivity. MRI arterial spin labeling allows for the quantification of regional cerebral blood flow, characteristically altered in Alzheimer's disease, diffuse Lewy-body disease, and the frontotemporal dementias. Positron emission tomography allows for the determination of regional metabolism, with similar abnormalities as flow, and for the measurement of β-amyloid and abnormal tau deposition in the brain, as well as regional inflammation. These instruments allow for the quantification in vivo of most of the pathologic features observed in disorders causing dementia. Importantly, they allow for the longitudinal study of these abnormalities, having revealed, for instance, that the deposition of β-amyloid in the brain can antecede by decades the onset of dementia. Thus, a therapeutic window has been opened and the efficacy of immunotherapies directed at removing β-amyloid from the brain of asymptomatic individuals is currently being tested. Tau and inflammation imaging, still in their infancy, combined with genomics, should provide powerful insights into these disorders and facilitate their treatment.
Collapse
Affiliation(s)
- Joseph C Masdeu
- Department of Neurology, Houston Methodist Hospital, Houston, TX, USA.
| | - Belen Pascual
- Department of Neurology, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
148
|
Waldron AM, Wintmolders C, Bottelbergs A, Kelley JB, Schmidt ME, Stroobants S, Langlois X, Staelens S. In vivo molecular neuroimaging of glucose utilization and its association with fibrillar amyloid-β load in aged APPPS1-21 mice. ALZHEIMERS RESEARCH & THERAPY 2015; 7:76. [PMID: 26666747 PMCID: PMC4678474 DOI: 10.1186/s13195-015-0158-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/21/2015] [Indexed: 11/10/2022]
Abstract
Introduction Radioligand imaging is a powerful in vivo method to assess the molecular basis of Alzheimer’s Disease. We therefore aimed to visualize the pathological deposition of fibrillar amyloid-β and neuronal dysfunction in aged double transgenic mice. Methods Using non-invasive positron emission tomography (PET) we assessed brain glucose utilization with [18F]FDG and fibrillar amyloidosis with [11C]PiB and [18F]AV45 in 12 month old APPPS1-21 (n = 10) mice and their age-matched wild-type controls (n = 15). PET scans were analyzed with statistical parametric mapping (SPM) to detect significant differences in tracer uptake between genotypes. After imaging, mice were sacrificed and ex vivo measures of amyloid-β burden with immunohistochemistry as well as glucose utilization with [14C]-2DG autoradiography were obtained as gold standards. Results Voxel-wise SPM analysis revealed significantly decreased [18F]FDG uptake in aged APPPS1-21 mice in comparison to WT with the thalamus (96.96 %, maxT = 3.35) and striatum (61.21 %, maxT = 3.29) demonstrating the most widespread reductions at the threshold of p < 0.01. [11C]PiB binding was significantly increased in APPPS1-21 mice, most notably in the hippocampus (87.84 %, maxT = 7.15) and cortex (69.08 %, maxT = 7.95), as detected by SPM voxel-wise analysis at the threshold of p < 0.01. Using the same threshold [18F]AV45 uptake was comparably lower with less significant differences. Compared to their respective ex vivo equivalents [18F]FDG demonstrated significant positive correlation to [14C]2-DG autoradiography (r = 0.67, p <0.0001) while [11C]PiB and [18F]AV45 binding did not correlate to ex vivo immunohistochemistry for amyloid-β (r = 0.25, p = 0.07 and r = 0.17, p = 0.26 respectively). Lastly no correlation was observed between regions of high amyloid burden and those with decreased glucose utilization (r = 0.001, p = 0.99). Conclusions Our findings support that fibrillar amyloid-β deposition and reduced glucose utilization can be visualized and quantified with in vivo μPET imaging in aged APPPS1-21 mice. Therefore, the combined use of [18F]FDG and amyloid μPET imaging can shed light on the underlying relationship between fibrillar amyloid-β pathology and neuronal dysfunction.
Collapse
Affiliation(s)
- Ann-Marie Waldron
- Molecular Imaging Center Antwerp, University of Antwerp, Campus Drie Eiken - UC, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| | - Cindy Wintmolders
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Astrid Bottelbergs
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Jonathan B Kelley
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Mark E Schmidt
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Campus Drie Eiken - UC, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium. .,Nuclear Medicine Department, University Hospital Antwerp, Antwerp, Belgium.
| | - Xavier Langlois
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Campus Drie Eiken - UC, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| |
Collapse
|
149
|
In-vivo imaging of grey and white matter neuroinflammation in Alzheimer's disease: a positron emission tomography study with a novel radioligand, [18F]-FEPPA. Mol Psychiatry 2015; 20:1579-87. [PMID: 25707397 PMCID: PMC8026116 DOI: 10.1038/mp.2015.1] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 11/09/2022]
Abstract
Our primary aim was to compare neuroinflammation in cognitively intact control subjects and patients with Alzheimer's disease (AD) by using positron emission tomography (PET) with translocator protein 18 kDa (TSPO)-specific radioligand [(18)F]-FEPPA. [(18)F]-FEPPA PET scans were acquired on a high-resolution research tomograph in 21 patients with AD (47- 81 years) and 21 control subjects (49-82 years). They were analyzed by using a 2-tissue compartment model with arterial plasma input function. Differences in neuroinflammation, indexed as [(18)F]-FEPPA binding were compared, adjusting for differences in binding affinity class as determined by a single polymorphism in the TSPO gene (rs6971). In grey matter areas, [(18)F]-FEPPA was significantly higher in AD compared with healthy control subjects. Large increases were seen in the hippocampus, prefrontal, temporal, parietal and occipital cortex (average Cohen's d= 0.89). Voxel-based analyses confirmed significant clusters of neuroinflammation in the frontal, temporal and parietal cortex in patients with AD. In white matter, [(18)F]-FEPPA binding was elevated in the posterior limb of the internal capsule, and the cingulum bundle. Higher neuroinflammation in the parietal cortex (r= -0.7, P= 0.005), and posterior limb of the internal capsule (r= -0.8, P=0.001) was associated with poorer visuospatial function. In addition, a higher [(18)F]-FEPPA binding in the posterior limb of the internal capsule was associated with a greater impairment in language ability (r= -0.7, P=0.004). Elevated neuroinflammation can be detected in AD patients throughout the brain grey and white matter by using [(18)F]-FEPPA PET. Our results also suggest that neuroinflammation is associated with some cognitive deficits.
Collapse
|
150
|
Caso F, Agosta F, Filippi M. Insights into White Matter Damage in Alzheimer's Disease: From Postmortem to in vivo Diffusion Tensor MRI Studies. NEURODEGENER DIS 2015; 16:26-33. [DOI: 10.1159/000441422] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/01/2015] [Indexed: 11/19/2022] Open
|