1
|
de Souza AA, Goularte KCM, Piccoli RC, Custódio SV, de Mello JE, Victor MG, Domingues WB, de Souza LP, Dos Santos Gonçalves L, Campos VF, Cunico W, Oses JP, Stefanello FM, de Aguiar MSS, Spanevello RM. 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one Attenuates Scopolamine-induced Cognitive Impairment in Rats: Insights Into Neuroprotective Effects. Mol Neurobiol 2025:10.1007/s12035-025-04887-5. [PMID: 40164887 DOI: 10.1007/s12035-025-04887-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's Disease (AD) is characterized by memory decline, dysregulation in cholinergic and purinergic signaling, neuroinflammation, and oxidative stress. Current treatments are limited, highlighting the need for new compounds to prevent or delay AD progression. Thiazolidinones have emerged as promising candidates due to their antioxidant, anti-inflammatory, and anticholinesterase properties. The aim of this study was to evaluate the effects of 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one (DS27) in a rat model of scopolamine-induced memory deficits. Male rats were divided into groups: I - Control, II - Scopolamine (SCO) (1 mg/kg), III - SCO and DS27 (5 mg/kg), IV - SCO and DS27 (10 mg/kg), V - SCO and donepezil (5 mg/kg). The animals were treated orally with DS27 or donepezil for seven days. On the 8th day, they underwent the open field test and inhibitory avoidance training, followed by intraperitoneal administration SCO. Twenty-four hours later, an inhibitory avoidance test was conducted. Acetylcholinesterase (AChE) activity, oxidative stress, and inflammatory and purinergic parameters were analyzed in the cerebral cortex, hippocampus, cerebrospinal fluid, serum, lymphocytes, and liver. DS27 prevented memory deficits, alterations in AChE activity, and oxidative damage induced by SCO in brain structures. Additionally, DS27 prevented SCO-induced decrease in IL-10 levels, and increase in IL-6, and TNF-α expression in the cerebral cortex, and normalized ATP and ADP hydrolysis in cerebrospinal fluid and lymphocytes. DS27 did not induce oxidative stress in the liver or alter serum biochemical parameters. These findings suggest that DS27 has significant neuroprotective properties and could be a promising alternative for treating neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Anita Avila de Souza
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Kelen Cristiane Machado Goularte
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Raphaela Cassol Piccoli
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Solange Vega Custódio
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Julia Eisenhardt de Mello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Melinda Gomes Victor
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - William Borges Domingues
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Lucas Petitemberte de Souza
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Laís Dos Santos Gonçalves
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Vinicius Farias Campos
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Wilson Cunico
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Jean Pierre Oses
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Francieli Moro Stefanello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Department of Clinical Medicine, Faculty of Medicine - Laboratory of Neuroscience and Behavior - Drug Research and Development Center, Federal University of Ceará, Fortaleza-CE, CEP 60020-181, Brazil
| | - Roselia Maria Spanevello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil.
| |
Collapse
|
2
|
Gan R, Xie H, Zhao Z, Wu X, Wang R, Wu B, Chen Q, Jia Z. Investigation of patterns and associations of neuroinflammation in cognitive impairment. Cereb Cortex 2025; 35:bhaf013. [PMID: 39917815 DOI: 10.1093/cercor/bhaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/29/2024] [Accepted: 01/19/2025] [Indexed: 03/17/2025] Open
Abstract
Neuroinflammation has been identified as an important pathological component of cognitive impairment, and translocator protein imaging has become a valuable tool for assessing its patterns. We aimed to obtain the exact distribution of neuroinflammation in cognitive impairment and its underlying mechanisms with amyloid-beta. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines, two investigators searched literature databases for studies that measured translocator protein binding levels. This measurement was performed between healthy controls and subjects with mild cognitive impairment or Alzheimer's disease via voxel-based positron emission tomography image analysis at the whole-brain level. This meta-analysis was performed with the anisotropic effect-size based algorithm. Neuroinflammation in patients with mild cognitive impairment was mainly concentrated in the left middle temporal gyrus and left amygdala. In Alzheimer's disease patients, the brain regions involved were the left inferior temporal gyrus, left calcarine fissure/surrounding cortex, left parahippocampal gyrus, right lingual gyrus, and right middle temporal gyrus. In addition, neuroinflammation in patients with cognitive impairment was highly correlated with amyloid-beta deposition in the cortex. This study deepens our understanding of the patterns of neuroinflammation in patients with cognitive impairment and its interaction with amyloid-beta, providing potential insights for therapeutic approaches targeting neuroinflammation in Alzheimer's disease.
Collapse
Affiliation(s)
- Ruoqiu Gan
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Department of Radiology, Sanya People's Hospital, No. 558 Jiefang Road, 572000, Sanya, Hainan, China
| | - Hongsheng Xie
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Ziru Zhao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Ruihan Wang
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Baolin Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guo Xue Alley, 610041, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Appleton J, Finn Q, Zanotti-Fregonara P, Yu M, Faridar A, Nakawah MO, Zarate C, Carrillo MC, Dickerson BC, Rabinovici GD, Apostolova LG, Masdeu JC, Pascual B. Brain inflammation co-localizes highly with tau in mild cognitive impairment due to early-onset Alzheimer's disease. Brain 2025; 148:119-132. [PMID: 39013020 PMCID: PMC11706285 DOI: 10.1093/brain/awae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Brain inflammation, with an increased density of microglia and macrophages, is an important component of Alzheimer's disease and a potential therapeutic target. However, it is incompletely characterized, particularly in patients whose disease begins before the age of 65 years and, thus, have few co-pathologies. Inflammation has been usefully imaged with translocator protein (TSPO) PET, but most inflammation PET tracers cannot image subjects with a low-binder TSPO rs6971 genotype. In an important development, participants with any TSPO genotype can be imaged with a novel tracer, 11C-ER176, that has a high binding potential and a more favourable metabolite profile than other TSPO tracers currently available. We applied 11C-ER176 to detect brain inflammation in mild cognitive impairment (MCI) caused by early-onset Alzheimer's disease. Furthermore, we sought to correlate the brain localization of inflammation, volume loss, elevated amyloid-β (Aβ)and tau. We studied brain inflammation in 25 patients with early-onset amnestic MCI (average age 59 ± 4.5 years, 10 female) and 23 healthy controls (average age 65 ± 6.0 years, 12 female), both groups with a similar proportion of all three TSPO-binding affinities. 11C-ER176 total distribution volume (VT), obtained with an arterial input function, was compared across patients and controls using voxel-wise and region-wise analyses. In addition to inflammation PET, most MCI patients had Aβ (n = 23) and tau PET (n = 21). For Aβ and tau tracers, standard uptake value ratios were calculated using cerebellar grey matter as region of reference. Regional correlations among the three tracers were determined. Data were corrected for partial volume effect. Cognitive performance was studied with standard neuropsychological tools. In MCI caused by early-onset Alzheimer's disease, there was inflammation in the default network, reaching statistical significance in precuneus and lateral temporal and parietal association cortex bilaterally, and in the right amygdala. Topographically, inflammation co-localized most strongly with tau (r = 0.63 ± 0.24). This correlation was higher than the co-localization of Aβ with tau (r = 0.55 ± 0.25) and of inflammation with Aβ (0.43 ± 0.22). Inflammation co-localized least with atrophy (-0.29 ± 0.26). These regional correlations could be detected in participants with any of the three rs6971 TSPO polymorphisms. Inflammation in Alzheimer's disease-related regions correlated with impaired cognitive scores. Our data highlight the importance of inflammation, a potential therapeutic target, in the Alzheimer's disease process. Furthermore, they support the notion that, as shown in experimental tissue and animal models, the propagation of tau in humans is associated with brain inflammation.
Collapse
Affiliation(s)
- Johanna Appleton
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Quentin Finn
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | | | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Carlos Zarate
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, IL 60603, USA
| | | | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Terada T, Kubota M, Miyata J, Obi T, Takashima H, Matsudaira T, Bunai T, Ouchi Y, Murai T. Frontal neurodegeneration associated with Frontal Assessment Battery in early Alzheimer's disease. J Neurol Sci 2024; 467:123327. [PMID: 39608296 DOI: 10.1016/j.jns.2024.123327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The Frontal Assessment Battery (FAB) is widely used to assess executive dysfunction in patients with amnestic mild cognitive impairments due to Alzheimer's disease (aMCI-AD), but its neurobiological meaning is unclear. To elucidate this, we examined the relationship between the FAB score and three key imaging biomarkers: gray matter volume, amyloid-beta (Aβ) deposition, and glucose metabolism. METHODS Twenty Aβ- and tau-positive aMCI-AD patients and age-matched controls underwent structural magnetic resonance imaging and positron emission tomography with [11C]PiB and [18F]FDG. Voxel-based morphometry and statistical parametric mapping analyses were performed to elucidate the relationships between FAB scores and regional gray matter volume, [11C]PiB uptake for Aβ deposition, and [18F]FDG uptake for glucose metabolism. RESULTS FAB scores were significantly lower in aMCI-AD than in controls (p < 0.001). In aMCI-AD, FAB was significantly correlated with right inferior frontal gray matter volume and right medial and left middle frontal glucose metabolism (family-wise error p < 0.05). However, there was no correlation between Aβ deposition and FAB (family-wise error p < 0.05). CONCLUSIONS The decreased FAB score is linked more with frontal-lobe neurodegeneration than with Aβ pathology in aMCI-AD. The FAB could be an early marker for neurodegeneration related to frontal-lobe executive dysfunction.
Collapse
Affiliation(s)
- Tatsuhiro Terada
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka 420-8688, Japan; Department of Biofunctional Imaging, Preeminent Bioimaging Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama Higashi-ku, Hamamatsu 431-3192, Japan.
| | - Manabu Kubota
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Brain Disorder Translational Research Group, Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Jun Miyata
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Psychiatry, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute-shi, Aichi 480-1195, Japan
| | - Tomokazu Obi
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka 420-8688, Japan
| | - Hirotsugu Takashima
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka 420-8688, Japan; Department of Biofunctional Imaging, Preeminent Bioimaging Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama Higashi-ku, Hamamatsu 431-3192, Japan
| | - Takashi Matsudaira
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka 420-8688, Japan; Department of Biofunctional Imaging, Preeminent Bioimaging Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama Higashi-ku, Hamamatsu 431-3192, Japan
| | - Tomoyasu Bunai
- Department of Biofunctional Imaging, Preeminent Bioimaging Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama Higashi-ku, Hamamatsu 431-3192, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Bioimaging Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama Higashi-ku, Hamamatsu 431-3192, Japan
| | - Toshiya Murai
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
5
|
Yasuno F, Kimura Y, Ogata A, Ikenuma H, Abe J, Minami H, Nihashi T, Yokoi K, Hattori S, Shimoda N, Watanabe A, Kasuga K, Ikeuchi T, Takeda A, Sakurai T, Ito K, Kato T. Trait-anxiety and glial-related neuroinflammation of the amygdala and its associated regions in Alzheimer's disease: A significant correlation. Brain Behav Immun Health 2024; 38:100795. [PMID: 38799793 PMCID: PMC11126804 DOI: 10.1016/j.bbih.2024.100795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/28/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
Background Positron emission tomography, which assesses the binding of translocator protein radiotracers, 11C-DPA-713, may be a sensitive method for determining glial-mediated neuroinflammation levels. This study investigated the relationship between regional 11C-DPA713 binding potential (BPND) and anxiety in patients with Alzheimer's disease (AD) continuum. Methods Nineteen patients with AD continuum determined to be amyloid-/p-tau 181-positive via cerebrospinal fluid analysis were included in this cross-sectional study (mild cognitive impairment [MCI, n = 5] and AD [n = 14]). Anxiety was evaluated using the State-Trait Anxiety Inventory (STAI). A whole-brain voxel-based analysis was performed to examine the relationship between 11C-DPA-713-BPND values at each voxel and the STAI score. Stepwise multiple regression analysis was performed to determine the predictors of STAI scores using independent variables, including 11C-DPA-713-BPND values within significant clusters. 11C-DPA-713-BPND values were compared between patients with AD continuum with low-to-moderate and high STAI scores. Results Voxel-based analysis revealed a positive correlation between trait anxiety severity and 11C-DPA713-BPND values in the centromedial amygdala and the left inferior occipital area [P < 0.001 (uncorrected) at the voxel-level]. 11C-DPA713-BPND values in these regions were a strong predictor of the STAI trait anxiety score. Specifically, patients with AD continuum and high trait anxiety had increased 11C-DPA713-BPND values in these regions. Conclusions The amygdala-occipital lobe circuit influences the control of emotional generation, and disruption of this network by AD pathology-induced inflammation may contribute to the expression of anxiety. Our findings suggest that suppression of inflammation can help effectively treat anxiety by attenuating damage to the amygdala and its associated areas.
Collapse
Affiliation(s)
- Fumihiko Yasuno
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuyuki Kimura
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Aya Ogata
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Kani, Japan
| | - Hiroshi Ikenuma
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Junichiro Abe
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroyuki Minami
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Nihashi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kastunori Yokoi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Saori Hattori
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Nobuyoshi Shimoda
- Functional Genomics Unit, Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akinori Takeda
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Sakurai
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kengo Ito
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
6
|
Zhang L, Gai Y, Liu Y, Meng D, Zeng Y, Luo Y, Zhang H, Wang Z, Yang M, Li Y, Liu Y, Lai Y, Yang J, Wu G, Chen Y, Zhu J, Liu S, Yu T, Zeng J, Wang J, Zhu D, Wang X, Lan X, Liu R. Tau induces inflammasome activation and microgliosis through acetylating NLRP3. Clin Transl Med 2024; 14:e1623. [PMID: 38488468 PMCID: PMC10941548 DOI: 10.1002/ctm2.1623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) and related Tauopathies are characterised by the pathologically hyperphosphorylated and aggregated microtubule-associated protein Tau, which is accompanied by neuroinflammation mediated by activated microglia. However, the role of Tau pathology in microglia activation or their causal relationship remains largely elusive. METHODS The levels of nucleotide-binding oligomerisation domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) acetylation and inflammasome activation in multiple cell models with Tau proteins treatment, transgenic mice with Tauopathy, and AD patients were measured by Western blotting and enzyme-linked immunosorbent assay. In addition, the acetyltransferase activity of Tau and NLRP3 acetylation sites were confirmed using the test-tube acetylation assay, co-immunoprecipitation, immunofluorescence (IF) staining, mass spectrometry and molecular docking. The Tau-overexpressing mouse model was established by overexpression of human Tau proteins in mouse hippocampal CA1 neurons through the adeno-associated virus injection. The cognitive functions of Tau-overexpressing mice were assessed in various behavioural tests, and microglia activation was analysed by Iba-1 IF staining and [18F]-DPA-714 positron emission tomography/computed tomography imaging. A peptide that blocks the interaction between Tau and NLRP3 was synthesised to determine the in vitro and in vivo effects of Tau-NLRP3 interaction blockade on NLRP3 acetylation, inflammasome activation, microglia activation and cognitive function. RESULTS Excessively elevated NLRP3 acetylation and inflammasome activation were observed in 3xTg-AD mice, microtubule-associated protein Tau P301S (PS19) mice and AD patients. It was further confirmed that mimics of 'early' phosphorylated-Tau proteins which increase at the initial stage of diseases with Tauopathy, including TauT181E, TauS199E, TauT217E and TauS262E, significantly promoted Tau-K18 domain acetyltransferase activity-dependent NLRP3 acetylation and inflammasome activation in HEK293T and BV-2 microglial cells. In addition, Tau protein could directly acetylate NLRP3 at the K21, K22 and K24 sites at its PYD domain and thereby induce inflammasome activation in vitro. Overexpression of human Tau proteins in mouse hippocampal CA1 neurons resulted in impaired cognitive function, Tau transmission to microglia and microgliosis with NLRP3 acetylation and inflammasome activation. As a targeted intervention, competitive binding of a designed Tau-NLRP3-binding blocking (TNB) peptide to block the interaction of Tau protein with NLRP3 inhibited the NLRP3 acetylation and downstream inflammasome activation in microglia, thereby alleviating microglia activation and cognitive impairment in mice. CONCLUSIONS In conclusion, our findings provide evidence for a novel role of Tau in the regulation of microglia activation through acetylating NLRP3, which has potential implications for early intervention and personalised treatment of AD and related Tauopathies.
Collapse
Affiliation(s)
- Lun Zhang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of Clinical LaboratoryWuhan Fourth HospitalWuhanChina
| | - Yongkang Gai
- Department of Nuclear MedicineHubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yushuang Liu
- Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Dongli Meng
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yi Zeng
- Department of Clinical LaboratoryThe Central Hospital of WuhanWuhanChina
| | - Yong Luo
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Huiliang Zhang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhuoqun Wang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Mengzhe Yang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yunfan Li
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yi Liu
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yiwen Lai
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jiayu Yang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Gang Wu
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yu Chen
- Department of PediatricsTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jingtan Zhu
- Britton Chance Center for Biomedical Photonics‐MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhanChina
| | - Shaojun Liu
- Britton Chance Center for Biomedical Photonics‐MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhanChina
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics‐MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhanChina
| | - Ji Zeng
- Department of Clinical LaboratoryWuhan Fourth HospitalWuhanChina
| | - Jianzhi Wang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics‐MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhanChina
| | - Xiaochuan Wang
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Shenzhen Huazhong University of Science and Technology Research InstituteShenzhenChina
| | - Xiaoli Lan
- Department of Nuclear MedicineHubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Rong Liu
- Department of PathophysiologySchool of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of PediatricsTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Shenzhen Huazhong University of Science and Technology Research InstituteShenzhenChina
| |
Collapse
|
7
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
8
|
Lee MY, Kim M. Effects of Red ginseng on neuroinflammation in neurodegenerative diseases. J Ginseng Res 2024; 48:20-30. [PMID: 38223824 PMCID: PMC10785270 DOI: 10.1016/j.jgr.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/17/2023] [Accepted: 08/25/2023] [Indexed: 01/16/2024] Open
Abstract
Red ginseng (RG) is widely used as a herbal medicine. As the human lifespan has increased, numerous diseases have developed, and RG has also been used to treat various diseases. Neurodegenerative diseases are major problems that modern people face through their lives. Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are featured by progressive nerve system damage. Recently, neuroinflammation has emerged as a degenerative factor and is an immune response in which cytokines with nerve cells that constitute the nervous system. RG, a natural herbal medicine with fewer side effects than chemically synthesized drugs, is currently in the spotlight. Therefore, we reviewed studies reporting the roles of RG in treating neuroinflammation and neurodegenerative diseases and found that RG might help alleviate neurodegenerative diseases by regulating neuroinflammation.
Collapse
Affiliation(s)
- Min Yeong Lee
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| |
Collapse
|
9
|
Abstract
This paper provides an overview of the role of neuroinflammation in Alzheimer's disease and other neurodegenerative diseases, highlighting the potential of anti-inflammatory treatments to slow or prevent decline. This research focuses on the use of positron emission tomography (PET) imaging to visualize and quantify molecular brain changes in patients, specifically microglial activation and reactive astrogliosis. We discuss the development and application of several PET radioligands, including first-generation ligands like PK11195 and Ro5-4864, as well as second- and third-generation ligands such as [11C]PBR28, [18F]DPA-714, [18F]GE-180, and [11C]ER176. These ligands target the 18-kDa translocator protein (TSPO), which is overexpressed in activated microglia and upregulated in astrocytes. We also address the limitations of these ligands, such as low brain uptake, poor penetration of the blood-brain barrier, short half-life, and variable kinetic behavior. Furthermore, we demonstrate the impact of genetic polymorphisms on ligand binding.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
10
|
Yasuno F, Kimura Y, Ogata A, Ikenuma H, Abe J, Minami H, Nihashi T, Yokoi K, Hattori S, Shimoda N, Watanabe A, Kasuga K, Ikeuchi T, Takeda A, Sakurai T, Ito K, Kato T. Neuroimaging biomarkers of glial activation for predicting the annual cognitive function decline in patients with Alzheimer's disease. Brain Behav Immun 2023; 114:214-220. [PMID: 37648003 DOI: 10.1016/j.bbi.2023.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Glial activation is central to the pathogenesis of Alzheimer's disease (AD). However, researchers have not demonstrated its relationship to longitudinal cognitive deterioration. We aimed to compare the prognostic effects of baseline positron emission tomography (PET) imaging of glial activation and amyloid/tau pathology on the successive annual cognitive decline in patients with AD. METHODS We selected 17 patients diagnosed with mild cognitive impairment or AD. We assessed the annual changes in global cognition and memory. Furthermore, we assessed the predictive effects of baseline amyloid and tau pathology indicated by cerebrospinal fluid (CSF) concentrations and PET imaging of glial activation (11C-DPA-713-binding potential in the area of Braak 1-3 [11C-DPA-713-BPND]) on global cognition and memory using a stepwise regression analysis. RESULTS The final multiple regression model of annual changes in global cognition and memory scores included 11C-DPA-713-BPND as the predictor. The CSF Aβ42/40 ratios and p-tau concentrations were removed from the final model. In stepwise Bayesian regression analysis, the Bayes factor-based model comparison suggested that the best model included 11C-DPA-713-BPND as the predictor of decline in global cognition and memory. CONCLUSIONS Translocator protein-PET imaging of glial activation is a stronger predictor of AD clinical progression than the amount of amyloid/tau pathology measured using CSF concentrations. Glial activation is the primary cause of tau-induced neuronal toxicity and cognitive deterioration, thereby highlighting the potential of blocking maladaptive microglial responses as a therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Fumihiko Yasuno
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan.
| | - Yasuyuki Kimura
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Aya Ogata
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Kani, Japan
| | - Hiroshi Ikenuma
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Junichiro Abe
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroyuki Minami
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Nihashi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kastunori Yokoi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Saori Hattori
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Nobuyoshi Shimoda
- Molecular Analysis Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akinori Takeda
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Sakurai
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kengo Ito
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan; Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
11
|
Gouilly D, Rafiq M, Nogueira L, Salabert AS, Payoux P, Péran P, Pariente J. Beyond the amyloid cascade: An update of Alzheimer's disease pathophysiology. Rev Neurol (Paris) 2023; 179:812-830. [PMID: 36906457 DOI: 10.1016/j.neurol.2022.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/02/2022] [Accepted: 12/02/2022] [Indexed: 03/13/2023]
Abstract
Alzheimer's disease (AD) is a multi-etiology disease. The biological system of AD is associated with multidomain genetic, molecular, cellular, and network brain dysfunctions, interacting with central and peripheral immunity. These dysfunctions have been primarily conceptualized according to the assumption that amyloid deposition in the brain, whether from a stochastic or a genetic accident, is the upstream pathological change. However, the arborescence of AD pathological changes suggests that a single amyloid pathway might be too restrictive or inconsistent with a cascading effect. In this review, we discuss the recent human studies of late-onset AD pathophysiology in an attempt to establish a general updated view focusing on the early stages. Several factors highlight heterogenous multi-cellular pathological changes in AD, which seem to work in a self-amplifying manner with amyloid and tau pathologies. Neuroinflammation has an increasing importance as a major pathological driver, and perhaps as a convergent biological basis of aging, genetic, lifestyle and environmental risk factors.
Collapse
Affiliation(s)
- D Gouilly
- Toulouse Neuroimaging Center, Toulouse, France.
| | - M Rafiq
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - L Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - A-S Salabert
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France
| | - P Payoux
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - P Péran
- Toulouse Neuroimaging Center, Toulouse, France
| | - J Pariente
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| |
Collapse
|
12
|
Hautecloque-Raysz G, Sellal F, Bousiges O, Phillipi N, Blanc F, Cretin B. Epileptic Prodromal Alzheimer's Disease Treated with Antiseizure Medications: Medium-Term Outcome of Seizures and Cognition. J Alzheimers Dis 2023:JAD221197. [PMID: 37355889 DOI: 10.3233/jad-221197] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2023]
Abstract
BACKGROUND The medium term outcome (over more than one year) of epileptic prodromal AD (epAD) patients treated with antiseizure medications (ASMs) is unknown in terms of seizure response, treatment tolerability, and cognitive and functional progression. OBJECTIVE To describe such medium term outcome over a mean of 5.1±2.1 years. METHODS We retrospectively compared 19 epAD patients with 16 non-epileptic prodromal AD (nepAD) patients: 1) at baseline for demographics, medical history, cognitive fluctuations (CFs), psychotropic medications, MMSE scores, visually rated hippocampal atrophy, CSF neurodegenerative biomarkers, and standard EEG recordings; 2) during follow-up (FU) for psychotropic medications, MMSE progression, and conversion to dementia. In the epAD group, we analyzed baseline and FU types of seizures as well as each line of ASM with the corresponding efficacy and tolerability. RESULTS At baseline, the epAD group had more CFs than the nepAD group (58% versus 20%, p = 0.03); focal impaired awareness seizures were the most common type (n = 12, 63.1%), occurring at a monthly to quarterly frequency (89.5%), and were well controlled with monotherapy in 89.5% of cases (including 63.1% seizure-free individuals). During FU, treated epAD patients did not differ significantly from nepAD patients in MMSE progression or in conversion to dementia. CONCLUSION Epilepsy is commonly controlled with ASMs over the medium term in epAD patients, with similar functional and cognitive outcomes to nepAD patients. Pathophysiologically, epilepsy is likely to be an ASM-modifiable cognitive aggravating factor at this stage of AD.
Collapse
Affiliation(s)
- Geoffroy Hautecloque-Raysz
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), France
- Service de Neurologie, Hospices Civils de Colmar, France
| | - François Sellal
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), France
- Service de Neurologie, Hospices Civils de Colmar, France
- Unité INSERM U-1118, Faculté de Médecine de Strasbourg, France
| | - Olivier Bousiges
- Service de Neurologie, Hospices Civils de Colmar, France
- University Hospital of Strasbourg, Laboratory of Biochemistry and Molecular Biology, and CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Strasbourg, France
| | - Nathalie Phillipi
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), France
- Unité de Neuropsychologie, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and CNRS, ICube laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS/Neurocrypto Strasbourg, France
- Centre de Compétences des démences rares des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Frédéric Blanc
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), France
- Unité de Neuropsychologie, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and CNRS, ICube laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS/Neurocrypto Strasbourg, France
- Centre de Compétences des démences rares des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Benjamin Cretin
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), France
- Unité de Neuropsychologie, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and CNRS, ICube laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS/Neurocrypto Strasbourg, France
- Centre de Compétences des démences rares des Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
13
|
Yang L, Pang X, Guo W, Zhu C, Yu L, Song X, Wang K, Pang C. An Exploration of the Coherent Effects between METTL3 and NDUFA10 on Alzheimer's Disease. Int J Mol Sci 2023; 24:10111. [PMID: 37373264 DOI: 10.3390/ijms241210111] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized primarily by a decline in cognitive function. However, the etiopathogenesis of AD is unclear. N6-methyladenosine (m6A) is abundant in the brain, and it is interesting to explore the relationship between m6A and AD causes. In this paper, the gene expression of METTL3 and NDUFA10 were found to correlate with the Mini-mental State Examination (MMSE), which is a clinical indicator of the degree of dementia. METTL3 is involved in post-transcriptional methylation and the formation of m6A. NDUFA10 encodes the protein with NADH dehydrogenase activity and oxidoreductase activity in the mitochondrial electron transport chain. The following three characteristics were observed in this paper: 1. The lower the expression level of NDUFA10, the smaller the MMSE, and the higher the degree of dementia. 2. If the expression level of METTL3 dropped below its threshold, the patient would have a risk of AD with a probability close to 100%, suggesting a basic necessity for m6A to protect mRNA. 3. The lower the expression levels of both METTL3 and NDUFA10, the more likely the patient would suffer from AD, implying the coherence between METTL3 and NDUFA10. Regarding the above discovery, the following hypothesis is presented: METTL3 expression level is downregulated, then the m6A modification level of NDUFA10 mRNA is also decreased, thereby reducing the expression level of NDUFA10-encoded protein. Furthermore, the abnormal expression of NDUFA10 contributes to the assembly disorder of mitochondrial complex I and affects the process of the electron respiratory chain, with the consequent development of AD. In addition, to confirm the above conclusions, the AI Ant Colony Algorithm was improved to be more suitable for discovering the characteristics of AD data, and the SVM diagnostic model was applied to mine the coherent effects on AD between METTL3 and NDUFA10. In conclusion, our findings suggest that dysregulated m6A leads to altered expression of its target genes, thereby affecting AD's development.
Collapse
Affiliation(s)
- Lin Yang
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| | - Xinping Pang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Wenbo Guo
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| | - Chengjiang Zhu
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| | - Lei Yu
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| | - Xianghu Song
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu 610101, China
| |
Collapse
|
14
|
Ferrari-Souza JP, Lussier FZ, Leffa DT, Therriault J, Tissot C, Bellaver B, Ferreira PC, Malpetti M, Wang YT, Povala G, Benedet AL, Ashton NJ, Chamoun M, Servaes S, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, O’Brien JT, Rowe JB, Cohen AD, Lopez OL, Tudorascu DL, Karikari TK, Klunk WE, Villemagne VL, Soucy JP, Gauthier S, Souza DO, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA. APOEε4 associates with microglial activation independently of Aβ plaques and tau tangles. SCIENCE ADVANCES 2023; 9:eade1474. [PMID: 37018391 PMCID: PMC10075966 DOI: 10.1126/sciadv.ade1474] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
Animal studies suggest that the apolipoprotein E ε4 (APOEε4) allele is a culprit of early microglial activation in Alzheimer's disease (AD). Here, we tested the association between APOEε4 status and microglial activation in living individuals across the aging and AD spectrum. We studied 118 individuals with positron emission tomography for amyloid-β (Aβ; [18F]AZD4694), tau ([18F]MK6240), and microglial activation ([11C]PBR28). We found that APOEε4 carriers presented increased microglial activation relative to noncarriers in early Braak stage regions within the medial temporal cortex accounting for Aβ and tau deposition. Furthermore, microglial activation mediated the Aβ-independent effects of APOEε4 on tau accumulation, which was further associated with neurodegeneration and clinical impairment. The physiological distribution of APOE mRNA expression predicted the patterns of APOEε4-related microglial activation in our population, suggesting that APOE gene expression may regulate the local vulnerability to neuroinflammation. Our results support that the APOEε4 genotype exerts Aβ-independent effects on AD pathogenesis by activating microglia in brain regions associated with early tau deposition.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Firoza Z. Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Douglas T. Leffa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- ADHD Outpatient Program and Development Psychiatry Program, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Maura Malpetti
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Guilherme Povala
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andréa L. Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Artificial Intelligence and Computational Neurosciences lab, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - John T. O’Brien
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B. Rowe
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Ann D. Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas K. Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Diogo O. Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Hoenig MC, Drzezga A. Clear-headed into old age: Resilience and resistance against brain aging-A PET imaging perspective. J Neurochem 2023; 164:325-345. [PMID: 35226362 DOI: 10.1111/jnc.15598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022]
Abstract
With the advances in modern medicine and the adaptation towards healthier lifestyles, the average life expectancy has doubled since the 1930s, with individuals born in the millennium years now carrying an estimated life expectancy of around 100 years. And even though many individuals around the globe manage to age successfully, the prevalence of aging-associated neurodegenerative diseases such as sporadic Alzheimer's disease has never been as high as nowadays. The prevalence of Alzheimer's disease is anticipated to triple by 2050, increasing the societal and economic burden tremendously. Despite all efforts, there is still no available treatment defeating the accelerated aging process as seen in this disease. Yet, given the advances in neuroimaging techniques that are discussed in the current Review article, such as in positron emission tomography (PET) or magnetic resonance imaging (MRI), pivotal insights into the heterogenous effects of aging-associated processes and the contribution of distinct lifestyle and risk factors already have and are still being gathered. In particular, the concepts of resilience (i.e. coping with brain pathology) and resistance (i.e. avoiding brain pathology) have more recently been discussed as they relate to mechanisms that are associated with the prolongation and/or even stop of the progressive brain aging process. Better understanding of the underlying mechanisms of resilience and resistance may one day, hopefully, support the identification of defeating mechanism against accelerating aging.
Collapse
Affiliation(s)
- Merle C Hoenig
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Alexander Drzezga
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases, Bonn/Cologne, Germany
| |
Collapse
|
16
|
Wang Q, Xie C. Microglia activation linking amyloid-β drive tau spatial propagation in Alzheimer's disease. Front Neurosci 2022; 16:951128. [PMID: 36033617 PMCID: PMC9417618 DOI: 10.3389/fnins.2022.951128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Neuropsychiatry, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
- The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- *Correspondence: Chunming Xie
| |
Collapse
|
17
|
Anti-Inflammatory Activity of 4-(4-(Heptyloxy)phenyl)-2,4-dihydro-3 H-1,2,4-triazol-3-one via Repression of MAPK/NF-κB Signaling Pathways in β-Amyloid-Induced Alzheimer's Disease Models. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155035. [PMID: 35956985 PMCID: PMC9370156 DOI: 10.3390/molecules27155035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 01/03/2023]
Abstract
Alzheimer’s disease (AD) is a major neurodegenerative disease, but so far, it can only be treated symptomatically rather than changing the process of the disease. Recently, triazoles and their derivatives have been shown to have potential for the treatment of AD. In this study, the neuroprotective effects of 4-(4-(heptyloxy)phenyl)-2,4-dihydro-3H-1,2,4-triazol-3-one (W112) against β-amyloid (Aβ)-induced AD pathology and its possible mechanism were explored both in vitro and in vivo. The results showed that W112 exhibits a neuroprotective role against Aβ-induced cytotoxicity in PC12 cells and improves the learning and memory abilities of Aβ-induced AD-like rats. In addition, the assays of the protein expression revealed that W112 reversed tau hyperphosphorylation and reduced the production of proinflammatory cytokines, tumor necrosis factor-α and interleukin-6, both in vitro and in vivo studies. Further study indicated that the regulation of mitogen-activated protein kinase/nuclear factor-κB pathways played a key role in mediating the neuroprotective effects of W112 against AD-like pathology. W112 may become a potential drug for AD intervention.
Collapse
|
18
|
Nozaki T, Sugiyama K, Asakawa T, Namba H, Yokokura M, Terada T, Bunai T, Ouchi Y. Increased anteroventral striatal dopamine transporter and motor recovery after subthalamic deep brain stimulation in Parkinson's disease. J Neurosurg 2022; 137:468-478. [PMID: 34972089 DOI: 10.3171/2021.10.jns211364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Subthalamic nucleus deep brain stimulation (STN-DBS) in Parkinson's disease is effective; however, its mechanism is unclear. To investigate the degree of neuronal terminal survival after STN-DBS, the authors examined the striatal dopamine transporter levels before and after treatment in association with clinical improvement using PET with [11C]2β-carbomethoxy-3β-(4-fluorophenyl)tropane ([11C]CFT). METHODS Ten patients with Parkinson's disease who had undergone bilateral STN-DBS were scanned twice with [11C]CFT PET just before and 1 year after surgery. Correlation analysis was conducted between [11C]CFT binding and off-period Unified Parkinson's Disease Rating Scale (UPDRS) scores assessed preoperatively and postoperatively. RESULTS [11C]CFT uptake reduced significantly in the posterodorsal putamen contralateral to the parkinsonism-dominant side after 1 year; however, an increase was noted in the contralateral anteroventral putamen and ipsilateral ventral caudate postoperatively (p < 0.05). The percentage increase in [11C]CFT binding was inversely correlated with the preoperative binding level in the bilateral anteroventral putamen, ipsilateral ventral caudate, contralateral anterodorsal putamen, contralateral posteroventral putamen, and contralateral nucleus accumbens. The percentage reduction in UPDRS-II score was significantly correlated with the percentage increase in [11C]CFT binding in the ipsilateral anteroventral putamen (p < 0.05). The percentage reduction in UPDRS-III score was significantly correlated with the percentage increase in [11C]CFT binding in the ipsilateral anteroventral putamen, ventral caudate, and nucleus accumbens (p < 0.05). CONCLUSIONS STN-DBS increases dopamine transporter levels in the anteroventral striatum, which is correlated with the motor recovery and possibly suggests the neuromodulatory effect of STN-DBS on dopaminergic terminals in Parkinson's disease patients. A preoperative level of anterior striatal dopamine transporter may predict reserve capacity of STN-DBS on motor recovery.
Collapse
Affiliation(s)
- Takao Nozaki
- 1Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Sugiyama
- 2Department of Neurosurgery, Toyoda Eisei Hospital, Iwata, Japan
| | - Tetsuya Asakawa
- 3Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Hiroki Namba
- 4Department of Neurosurgery, JA Shizuoka Kohseiren Enshu Hospital, Hamamatsu, Japan
| | - Masamichi Yokokura
- 5Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuhiro Terada
- 6Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
- 9Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyasu Bunai
- 7Department of Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- 9Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuomi Ouchi
- 8Hamamatsu PET Imaging Center, Hamamatsu Medical Photonics Foundation, Hamamatsu, Japan; and
- 9Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
19
|
Chen SM, Hsu TC, Chew CH, Huang WT, Chen AL, Lin YF, Eddarkaoui S, Buee L, Chen CC. Microtube Array Membrane Encapsulated Cell Therapy: A Novel Platform Technology Solution for Treatment of Alzheimer's Disease. Int J Mol Sci 2022; 23:6855. [PMID: 35743295 PMCID: PMC9224941 DOI: 10.3390/ijms23126855] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease is the most frequent form of dementia in aging population and is presently the world's sixth largest cause of mortality. With the advancement of therapies, several solutions have been developed such as passive immunotherapy against these misfolded proteins, thereby resulting in the clearance. Within this segment, encapsulated cell therapy (ECT) solutions that utilize antibody releasing cells have been proposed with a multitude of techniques under development. Hence, in this study, we utilized our novel and patented Microtube Array Membranes (MTAMs) as an encapsulating platform system with anti-pTau antibody-secreting hybridoma cells to study the impact of it on Alzheimer's disease. In vivo results revealed that in the water maze, the mice implanted with hybridoma cell MTAMs intracranially (IN) and subcutaneously (SC) showed improvement in the time spent the goal quadrant and escape latency. In passive avoidance, hybridoma cell loaded MTAMs (IN and SC) performed significantly well in step-through latency. At the end of treatment, animals with hybridoma cell loaded MTAMs had lower phosphorylated tau (pTau) expression than empty MTAMs had. Combining both experimental results unveiled that the clearance of phosphorylated tau might rescue the cognitive impairment associated with AD.
Collapse
Affiliation(s)
- Shu-Mei Chen
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-Chin Hsu
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11052, Taiwan; (T.-C.H.); (C.-H.C.); (W.-T.H.)
| | - Chee-Ho Chew
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11052, Taiwan; (T.-C.H.); (C.-H.C.); (W.-T.H.)
| | - Wan-Ting Huang
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11052, Taiwan; (T.-C.H.); (C.-H.C.); (W.-T.H.)
| | - Amanda Lin Chen
- Department of Biology, University of Washington, Seattle, WA 98195, USA;
| | - Yung-Feng Lin
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 11052, Taiwan;
| | - Sabiha Eddarkaoui
- Lille Neuroscience & Cognition, Inserm, CHU-Lille, Université de Lille, 59045 Lille, France; (S.E.); (L.B.)
| | - Luc Buee
- Lille Neuroscience & Cognition, Inserm, CHU-Lille, Université de Lille, 59045 Lille, France; (S.E.); (L.B.)
- NeuroTMU, Lille International Laboratory, Université de Lille, 59000 Lille, France
| | - Chien-Chung Chen
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11052, Taiwan; (T.-C.H.); (C.-H.C.); (W.-T.H.)
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- The Ph.D. Program for Translational Medicine, Taipei Medical University, Taipei 11052, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
20
|
Okazawa H, Ikawa M, Tsujikawa T, Mori T, Makino A, Kiyono Y, Nakamoto Y, Kosaka H, Yoneda M. Cerebral Oxidative Stress in Early Alzheimer's Disease Evaluated by 64Cu-ATSM PET/MRI: A Preliminary Study. Antioxidants (Basel) 2022; 11:1022. [PMID: 35624886 PMCID: PMC9138060 DOI: 10.3390/antiox11051022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress imaging using diacetyl-bis (N4-methylthiosemicarbazone) (Cu-ATSM) was applied to the evaluation of patients with early Alzheimer's disease (eAD). Ten eAD patients (72 ± 9 years) and 10 age-matched healthy controls (HCs) (73 ± 9 years) participated in this study. They underwent dynamic PET/MRI using 11C-PiB and 64Cu-ATSM with multiple MRI sequences. To evaluate cerebral oxidative stress, three parameters of 64Cu-ATSM PET were compared: standardized uptake value (SUV), tracer influx rate (Kin), and a rate constant k3. The input functions were estimated by the image-derived input function method. The relative differences were analyzed by statistical parametric mapping (SPM) using SUV and Kin images. All eAD patients had positive and HC subjects had negative PiB accumulation, and MMSE scores were significantly different between them. The 64Cu-ATSM accumulation tended to be higher in eAD than in HCs for both SUV and Kin. When comparing absolute values, eAD patients had a greater Kin in the posterior cingulate cortex and a greater k3 in the hippocampus compared with lobar cortical values of HCs. In SPM analysis, eAD had an increased left operculum and decreased bilateral hippocampus and anterior cingulate cortex compared to HCs. 64Cu-ATSM PET/MRI and tracer kinetic analysis elucidated cerebral oxidative stress in the eAD patients, particularly in the cingulate cortex and hippocampus.
Collapse
Affiliation(s)
- Hidehiko Okazawa
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
| | - Masamichi Ikawa
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
| | - Tetsuya Tsujikawa
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
| | - Tetsuya Mori
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
| | - Akira Makino
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
| | - Makoto Yoneda
- Biomedical Imaging Research Center, University of Fukui, Fukui 910-1193, Japan; (M.I.); (T.T.); (T.M.); (A.M.); (Y.K.); (M.Y.)
- Faculty of Nursing and Social Welfare Science, Fukui Prefectural University, Fukui 910-1195, Japan
| |
Collapse
|
21
|
Wang Q, Chen G, Schindler SE, Christensen J, McKay NS, Liu J, Wang S, Sun Z, Hassenstab J, Su Y, Flores S, Hornbeck R, Cash L, Cruchaga C, Fagan AM, Tu Z, Morris JC, Mintun MA, Wang Y, Benzinger TL. Baseline Microglial Activation Correlates With Brain Amyloidosis and Longitudinal Cognitive Decline in Alzheimer Disease. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:e1152. [PMID: 35260470 PMCID: PMC8906187 DOI: 10.1212/nxi.0000000000001152] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/06/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVES This study aims to quantify microglial activation in individuals with Alzheimer disease (AD) using the 18-kDa translocator protein (TSPO) PET imaging in the hippocampus and precuneus, the 2 AD-vulnerable regions, and to evaluate the association of baseline neuroinflammation with amyloidosis, tau, and longitudinal cognitive decline. METHODS Twenty-four participants from the Knight Alzheimer Disease Research Center (Knight ADRC) were enrolled and classified into stable cognitively normal, progressor, and symptomatic AD groups based on clinical dementia rating (CDR) at 2 or more clinical assessments. The baseline TSPO radiotracer [11C]PK11195 was used to image microglial activation. Baseline CSF concentrations of Aβ42, Aβ42/Aβ40 ratio, tau phosphorylated at position 181 (p-tau181), and total tau (t-tau) were measured. Clinical and cognitive decline were examined with longitudinal CDR and cognitive composite scores (Global and Knight ADRC-Preclinical Alzheimer Cognitive Composite [Knight ADRC-PACC] Score). RESULTS Participants in the progressor and symptomatic AD groups had significantly elevated [11C]PK11195 standard uptake value ratios (SUVRs) in the hippocampus but not in the precuneus region. In the subcohort with CSF biomarkers (16 of the 24), significant negative correlations between CSF Aβ42 or Aβ42/Aβ40 and [11C]PK11195 SUVR were observed in the hippocampus and precuneus. No correlations were observed between [11C]PK11195 SUVR and CSF p-tau181 or t-tau at baseline in those regions. Higher baseline [11C]PK11195 SUVR averaged in the whole cortical regions predicted longitudinal decline on cognitive tests. DISCUSSION Microglial activation is increased in individuals with brain amyloidosis and predicts worsening cognition in AD. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that in patients with AD, higher baseline [11C]PK11195 SUVR averaged in the whole cortical regions was associated with longitudinal decline on cognitive tests.
Collapse
Affiliation(s)
- Qing Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Gengsheng Chen
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Suzanne E. Schindler
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Jon Christensen
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Nicole S. McKay
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Jingxia Liu
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Sicheng Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Zhexian Sun
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Jason Hassenstab
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Yi Su
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Shaney Flores
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Russ Hornbeck
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Lisa Cash
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Carlos Cruchaga
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Anne M. Fagan
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Zhude Tu
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - John C. Morris
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Mark A. Mintun
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Yong Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Tammie L.S. Benzinger
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., J.C., S.F., R.H., Z.T., Y.W., T.L.S.B.), Washington University School of Medicine; Knight Alzheimer Disease Research Center (Q.W., G.C., S.E.S., J.H., L.C., A.M.F., J.C.M., T.L.S.B.), Washington University School of Medicine; Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Washington University School of Medicine; Department of Surgery (J.L.), Washington University School of Medicine; Department of Electrical and System Engineering (S.W., Y.W.), Washington University School of Med-icine; Department of Biomedical Engineering (Z.S., Y.W.), Washington University School of Medicine, St. Louis, MO; Banner Alzheimer's Institute and Arizona Alzheimer's Consortium (Y.S.), Phoenix, AZ; Department of Psychiatry (C.C.), Washington University School of Medicine, St. Louis, MO; Avid Radiopharmaceuticals (M.A.M.), Philadelphia, PA; Department of Obstetrics and Gynecology (Y.W.), Washington University School of Medicine; and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
22
|
Yousefzadeh-Nowshahr E, Winter G, Bohn P, Kneer K, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Strobel J, Kletting P, Riepe MW, Higuchi M, Glatting G, Ludolph A, Beer AJ. Quantitative analysis of regional distribution of tau pathology with 11C-PBB3-PET in a clinical setting. PLoS One 2022; 17:e0266906. [PMID: 35404966 PMCID: PMC9045369 DOI: 10.1371/journal.pone.0266906] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 03/29/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE The recent developments of tau-positron emission tomography (tau-PET) enable in vivo assessment of neuropathological tau aggregates. Among the tau-specific tracers, the application of 11C-pyridinyl-butadienyl-benzothiazole 3 (11C-PBB3) in PET shows high sensitivity to Alzheimer disease (AD)-related tau deposition. The current study investigates the regional tau load in patients within the AD continuum, biomarker-negative individuals (BN) and patients with suspected non-AD pathophysiology (SNAP) using 11C-PBB3-PET. MATERIALS AND METHODS A total of 23 memory clinic outpatients with recent decline of episodic memory were examined using 11C-PBB3-PET. Pittsburg compound B (11C-PIB) PET was available for 17, 18F-flurodeoxyglucose (18F-FDG) PET for 16, and cerebrospinal fluid (CSF) protein levels for 11 patients. CSF biomarkers were considered abnormal based on Aβ42 (< 600 ng/L) and t-tau (> 450 ng/L). The PET biomarkers were classified as positive or negative using statistical parametric mapping (SPM) analysis and visual assessment. Using the amyloid/tau/neurodegeneration (A/T/N) scheme, patients were grouped as within the AD continuum, SNAP, and BN based on amyloid and neurodegeneration status. The 11C-PBB3 load detected by PET was compared among the groups using both atlas-based and voxel-wise analyses. RESULTS Seven patients were identified as within the AD continuum, 10 SNAP and 6 BN. In voxel-wise analysis, significantly higher 11C-PBB3 binding was observed in the AD continuum group compared to the BN patients in the cingulate gyrus, tempo-parieto-occipital junction and frontal lobe. Compared to the SNAP group, patients within the AD continuum had a considerably increased 11C-PBB3 uptake in the posterior cingulate cortex. There was no significant difference between SNAP and BN groups. The atlas-based analysis supported the outcome of the voxel-wise quantification analysis. CONCLUSION Our results suggest that 11C-PBB3-PET can effectively analyze regional tau load and has the potential to differentiate patients in the AD continuum group from the BN and SNAP group.
Collapse
Affiliation(s)
- Elham Yousefzadeh-Nowshahr
- Department of Nuclear Medicine, Medical Radiation Physics, Ulm
University, Ulm, Germany
- Department of Nuclear Medicine, Medical Center—University of Freiburg,
Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | - Peter Bohn
- Department of Nuclear Medicine, Inselspital Bern—University of Bern,
Bern, Switzerland
| | - Katharina Kneer
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | - Christine A. F. von Arnim
- Department of Neurology, Ulm University, Ulm, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen,
Germany
| | - Markus Otto
- Department of Neurology, University Hospital Halle (Saale), Halle,
Germany
| | | | | | - Dörte Polivka
- Department of Neurology, Ulm University, Ulm, Germany
| | - Patrick Fissler
- Department of Neurology, Ulm University, Ulm, Germany
- Psychiatric Services of Thurgovia (Academic Teaching Hospital of Medical
University Salzburg), Münsterlingen, Switzerland
| | - Joachim Strobel
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Medical Radiation Physics, Ulm
University, Ulm, Germany
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, Ulm,
Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba,
Japan
| | - Gerhard Glatting
- Department of Nuclear Medicine, Medical Radiation Physics, Ulm
University, Ulm, Germany
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegerative Diseases (DZNE), Ulm,
Germany
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University, Ulm,
Germany
| | | |
Collapse
|
23
|
Gouilly D, Saint-Aubert L, Ribeiro MJ, Salabert AS, Tauber C, Péran P, Arlicot N, Pariente J, Payoux P. Neuroinflammation PET imaging of the translocator protein (TSPO) in Alzheimer's disease: an update. Eur J Neurosci 2022; 55:1322-1343. [PMID: 35083791 DOI: 10.1111/ejn.15613] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is a significant contributor to Alzheimer's disease (AD). Until now, PET imaging of the translocator protein (TSPO) has been widely used to depict the neuroimmune endophenotype of AD. The aim of this review was to provide an update to the results from 2018 and to advance the characterization of the biological basis of TSPO imaging in AD by re-examining TSPO function and expression and the methodological aspects of interest. Although the biological basis of the TSPO PET signal is obviously related to microglia and astrocytes in AD, the observed process remains uncertain and might not be directly related to neuroinflammation. Further studies are required to re-examine the cellular significance underlying a variation in the PET signal in AD and how it can be impacted by a disease-modifying treatment.
Collapse
Affiliation(s)
- Dominique Gouilly
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Laure Saint-Aubert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Maria-Joao Ribeiro
- Department of Nuclear Medicine, CHU, Tours, France.,UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| | | | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Jérémie Pariente
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU, Toulouse, France.,Center of Clinical Investigations (CIC1436), CHU, Toulouse, France
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| |
Collapse
|
24
|
Terada T, Therriault J, Kang MS, Savard M, Pascoal TA, Lussier F, Tissot C, Wang YT, Benedet A, Poltronetti NM, Ottoy J, Arias JF, Bezgin G, Matsudaira T, Bunai T, Obi T, Tsukada H, Ouchi Y, Rosa-Neto P. Mitochondrial complex-I abnormalities underlie neurodegeneration and cognitive decline in Alzheimer's disease. Eur J Neurol 2022; 29:1324-1334. [PMID: 35007366 DOI: 10.1111/ene.15246] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Abnormal mitochondrial metabolism has been described in Alzheimer's disease (AD) brain. However, the relationship between AD pathophysiology and key mitochondrial processes remains elusive. The purpose of this study is to investigate whether mitochondrial complex I dysfunction is associated with amyloid aggregation, or glucose metabolism and brain atrophy in patients with mild AD using positron emission tomography (PET). METHODS Amyloid and tau positive symptomatic AD patients with clinical dementia rating 0.5 or 1 (N=30; mean age ± standard deviation: 71.8 ± 7.6 years) underwent magnetic resonance imaging and PET scans with [18 F]BCPP-EF, [11 C]PiB and [18 F]FDG for assessing brain atrophy, mitochondrial complex I dysfunction, amyloid deposition, and glucose metabolism, respectively. Local cortical associations among these biomarkers and gray matter volume were evaluated with voxel-based regressions models. RESULTS [18 F]BCPP-EF standardized uptake value ratio (SUVR) was positively correlated with [18 F]FDG SUVR in the widespread brain area, while its associations with gray matter volume were restricted to the parahippocampal gyrus. Reductions in [18 F]BCPP-EF SUVR were associated with domain-specific cognitive performance. We did not observe regional associations between mitochondrial dysfunction and amyloid burden. CONCLUSIONS In symptomatic cases, although mitochondrial complex I reduction is linked to a wide range of downstream neurodegenerative processes such as hypometabolism, atrophy, and cognitive decline, the link to amyloid was not observable. The data presented here support [18 F]BCPP-EF as an excellent imaging tool to investigate mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Tatsuhiro Terada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada.,Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.,Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Tharick Ali Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Firoza Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Andrea Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Julie Ottoy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Jaime Frenandez Arias
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Takashi Matsudaira
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.,Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Tomoyasu Bunai
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Tomokazu Obi
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita-ku, Hamamatsu, 434-0041, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| |
Collapse
|
25
|
Chauveau F, Becker G, Boutin H. Have (R)-[ 11C]PK11195 challengers fulfilled the promise? A scoping review of clinical TSPO PET studies. Eur J Nucl Med Mol Imaging 2021; 49:201-220. [PMID: 34387719 PMCID: PMC8712292 DOI: 10.1007/s00259-021-05425-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The prototypical TSPO radiotracer (R)-[11C]PK11195 has been used in humans for more than thirty years to visualize neuroinflammation in several pathologies. Alternative radiotracers have been developed to improve signal-to-noise ratio and started to be tested clinically in 2008. Here we examined the scientific value of these "(R)-[11C]PK11195 challengers" in clinical research to determine if they could supersede (R)-[11C]PK11195. METHODS A systematic MEDLINE (PubMed) search was performed (up to end of year 2020) to extract publications reporting TSPO PET in patients with identified pathologies, excluding studies in healthy subjects and methodological studies. RESULTS Of the 288 publications selected, 152 used 13 challengers, and 142 used (R)-[11C]PK11195. Over the last 20 years, the number of (R)-[11C]PK11195 studies remained stable (6 ± 3 per year), but was surpassed by the total number of challenger studies for the last 6 years. In total, 3914 patients underwent a TSPO PET scan, and 47% (1851 patients) received (R)-[11C]PK11195. The 2 main challengers were [11C]PBR28 (24%-938 patients) and [18F]FEPPA (11%-429 patients). Only one-in-ten patients (11%-447) underwent 2 TSPO scans, among whom 40 (1%) were scanned with 2 different TSPO radiotracers. CONCLUSIONS Generally, challengers confirmed disease-specific initial (R)-[11C]PK11195 findings. However, while their better signal-to-noise ratio seems particularly useful in diseases with moderate and widespread neuroinflammation, most challengers present an allelic-dependent (Ala147Thr polymorphism) TSPO binding and genetic stratification is hindering their clinical implementation. As new challengers, insensitive to TSPO human polymorphism, are about to enter clinical evaluation, we propose this systematic review to be regularly updated (living review).
Collapse
Affiliation(s)
- Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, University Lyon 1, Lyon, France.
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University Liege, Liege, Belgium
- University of Lyon, CarMeN Laboratory, INSERM U1060, University Lyon 1, Hospices Civils Lyon, Lyon, France
| | - Hervé Boutin
- Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
26
|
Frigerio I, Boon BDC, Lin CP, Galis-de Graaf Y, Bol J, Preziosa P, Twisk J, Barkhof F, Hoozemans JJM, Bouwman FH, Rozemuller AJM, van de Berg WDJ, Jonkman LE. Amyloid-β, p-tau and reactive microglia are pathological correlates of MRI cortical atrophy in Alzheimer’s disease. Brain Commun 2021; 3:fcab281. [PMID: 34927073 PMCID: PMC8677327 DOI: 10.1093/braincomms/fcab281] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease is characterized by cortical atrophy on MRI and abnormal depositions of amyloid-beta, phosphorylated-tau and inflammation pathologically. However, the relative contribution of these pathological hallmarks to cortical atrophy, a widely used MRI biomarker in Alzheimer’s disease, is yet to be defined. Therefore, the aim of this study was to identify the histopathological correlates of MRI cortical atrophy in Alzheimer’s disease donors, and its typical amnestic and atypical non-amnestic phenotypes. Nineteen Alzheimer’s disease (of which 10 typical and 9 atypical) and 10 non-neurological control brain donors underwent post-mortem in situ 3T 3D-T1, from which cortical thickness was calculated with Freesurfer. Upon subsequent autopsy, 12 cortical brain regions from the right hemisphere and 9 from the left hemisphere were dissected and immunostained for amyloid-beta, phosphorylated-tau and reactive microglia, and percentage area load was calculated for each marker using ImageJ. In addition, post-mortem MRI was compared to ante-mortem MRI of the same Alzheimer’s disease donors when available. MRI-pathology associations were assessed using linear mixed models. Higher amyloid-beta load weakly correlated with higher cortical thickness globally (r = 0.22, P = 0.022). Phosphorylated-tau strongly correlated with cortical atrophy in temporal and frontal regions (−0.76 < r < −1.00, all P < 0.05). Reactive microglia load strongly correlated with cortical atrophy in the parietal region (r = −0.94, P < 0.001). Moreover, post-mortem MRI scans showed high concordance with ante-mortem scans acquired <1 year before death. In conclusion, distinct histopathological markers differently correlated with cortical atrophy, highlighting their different roles in the neurodegenerative process, and therefore contributing to the understanding of the pathological underpinnings of MRI atrophic patterns in Alzheimer’s disease. In our cohort, no or only subtle differences were found in MRI-pathology associations in Alzheimer’s disease phenotypes, indicating that the histopathological correlates of cortical atrophy in typical and atypical phenotypes might be similar. Moreover, we show that post-mortem in situ MRI can be used as proxy for ante-mortem in vivo MRI.
Collapse
Affiliation(s)
- Irene Frigerio
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Baayla D C Boon
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Chen-Pei Lin
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Yvon Galis-de Graaf
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - John Bol
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 60-20132 Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 60-20132 Milan, Italy
| | - Jos Twisk
- Department of Epidemiology and Biostatistics, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, the Netherlands
- Institutes of Neurology and Healthcare Engineering, University College London, London WC1E, UK
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Femke H Bouwman
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Location VUmc, Alzheimer Centrum Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| | - Laura E Jonkman
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
27
|
Kim S, Nam Y, Ham MJ, Park C, Moon M, Yoo DH. Neurological Mechanisms of Animal-Assisted Intervention in Alzheimer's Disease: A Hypothetical Review. Front Aging Neurosci 2021; 13:682308. [PMID: 34335229 PMCID: PMC8317687 DOI: 10.3389/fnagi.2021.682308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/14/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative brain disorder with aggregation of amyloid-beta (Aβ) and tau as the pathological hallmarks. AD is the most common form of dementia and is characterized by a progressive decline of cognition. The failure of pharmacological approaches to treat AD has resulted in an increased focus on non-pharmacological interventions that can mitigate cognitive decline and delay disease progression in patients with AD. Animal-assisted intervention (AAI), a non-pharmacological intervention, improves emotional, social, and cognitive dysfunction in patients with neurodegenerative diseases. In particular, AAI is reported to mitigate the effects of cognitive impairment in patients with AD. Despite the positive effects of AAI on cognitive dysfunction in patients with AD, there have been no studies on how AAI affects AD-related pathologies. This review postulates potential neurological mechanisms of emotional or social interaction through AAI in countering AD-related pathologies, such as Aβ deposition, tau hyperphosphorylation, neuroinflammation, and impaired adult hippocampal neurogenesis (AHN), and proposes insights for future research by organizing accumulated previous evidence.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, South Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Min-Joo Ham
- Department of Occupational Therapy, Konyang University, Daejeon, South Korea
| | - Chisoo Park
- Department of Occupational Therapy, Konyang University, Daejeon, South Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, South Korea
| | - Doo-Han Yoo
- Research Institute for Dementia Science, Konyang University, Daejeon, South Korea
- Department of Occupational Therapy, Konyang University, Daejeon, South Korea
| |
Collapse
|
28
|
Zhang PF, Hu H, Tan L, Yu JT. Microglia Biomarkers in Alzheimer's Disease. Mol Neurobiol 2021; 58:3388-3404. [PMID: 33713018 DOI: 10.1007/s12035-021-02348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Early detection and clinical diagnosis of Alzheimer's disease (AD) have become an extremely important link in the prevention and treatment of AD. Because of the occult onset, the diagnosis and treatment of AD based on clinical symptoms are increasingly challenged by current severe situations. Therefore, molecular diagnosis models based on early AD pathological markers have received more attention. Among the possible pathological mechanisms, microglia which are necessary for normal brain function are highly expected and have been continuously studied in various models. Several AD biomarkers already exist, but currently there is a paucity of specific and sensitive microglia biomarkers which can accurately measure preclinical AD. Bringing microglia biomarkers into the molecular diagnostic system which is based on fluid and neuroimaging will play an important role in future scientific research and clinical practice. Furthermore, developing novel, more specific, and sensitive microglia biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial microglial functions in response to AD pathology. This review discusses microglia biomarkers in the context of AD.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
29
|
Terada T, Therriault J, Kang MSP, Savard M, Pascoal TA, Lussier F, Tissot C, Wang YT, Benedet A, Matsudaira T, Bunai T, Obi T, Tsukada H, Ouchi Y, Rosa-Neto P. Mitochondrial complex I abnormalities is associated with tau and clinical symptoms in mild Alzheimer's disease. Mol Neurodegener 2021; 16:28. [PMID: 33902654 PMCID: PMC8074456 DOI: 10.1186/s13024-021-00448-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
Background Mitochondrial electron transport chain abnormalities have been reported in postmortem pathological specimens of Alzheimer’s disease (AD). However, it remains unclear how amyloid and tau are associated with mitochondrial dysfunction in vivo. The purpose of this study is to assess the local relationships between mitochondrial dysfunction and AD pathophysiology in mild AD using the novel mitochondrial complex I PET imaging agent [18F]BCPP-EF. Methods Thirty-two amyloid and tau positive mild stage AD dementia patients (mean age ± SD: 71.1 ± 8.3 years) underwent a series of PET measurements with [18F]BCPP-EF mitochondrial function, [11C]PBB3 for tau deposition, and [11C] PiB for amyloid deposition. Age-matched normal control subjects were also recruited. Inter and intrasubject comparisons of levels of mitochondrial complex I activity, amyloid and tau deposition were performed. Results The [18F]BCPP-EF uptake was significantly lower in the medial temporal area, highlighting the importance of the mitochondrial involvement in AD pathology. [11C]PBB3 uptake was greater in the temporo-parietal regions in AD. Region of interest analysis in the Braak stage I-II region showed significant negative correlation between [18F]BCPP-EF SUVR and [11C]PBB3 BPND (R = 0.2679, p = 0.04), but not [11C] PiB SUVR. Conclusions Our results indicated that mitochondrial complex I is closely associated with tau load evaluated by [11C]PBB3, which might suffer in the presence of its off-target binding. The absence of association between mitochondrial complex I dysfunction with amyloid load suggests that mitochondrial dysfunction in the trans-entorhinal and entorhinal region is a reflection of neuronal injury occurring in the brain of mild AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00448-1.
Collapse
Affiliation(s)
- Tatsuhiro Terada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada.,Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.,Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Min Su Peter Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Tharick Ali Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Firoza Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Andrea Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada
| | - Takashi Matsudaira
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.,Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Tomoyasu Bunai
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Tomokazu Obi
- Department of Neurology, Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi-ku, Shizuoka, 420-8688, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita-ku, Hamamatsu, 434-0041, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan. .,Hamamatsu PET Imaging Center, Hamamatsu Medical Photonics Foundation, 5000 Hirakuchi, Hamakita-ku, Hamamatsu, 434-0041, Japan.
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, 6875 Boulevard LaSalle, Montreal, H4H 1R3, Canada.
| |
Collapse
|
30
|
Clinical validity of increased cortical binding of tau ligands of the THK family and PBB3 on PET as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework. Eur J Nucl Med Mol Imaging 2021; 48:2086-2096. [PMID: 33723628 PMCID: PMC8175292 DOI: 10.1007/s00259-021-05277-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/21/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE The research community has focused on defining reliable biomarkers for the early detection of the pathological hallmarks of Alzheimer's disease (AD). In 2017, the Geneva AD Biomarker Roadmap initiative adapted the framework for the systematic validation of oncological biomarkers to AD, with the aim to accelerate their development and implementation in clinical practice. The aim of this work was to assess the validation status of tau PET ligands of the THK family and PBB3 as imaging biomarkers for AD, based on the Biomarker Roadmap methodology. METHODS A panel of experts in AD biomarkers convened in November 2019 at a 2-day workshop in Geneva. The level of clinical validity of tau PET ligands of the THK family and PBB3 was assessed based on the 5-phase development framework before the meeting and discussed during the workshop. RESULTS PET radioligands of the THK family discriminate well between healthy controls and patients with AD dementia (phase 2; partly achieved) and recent evidence suggests an accurate diagnostic accuracy at the mild cognitive impairment (MCI) stage of the disease (phase 3; partly achieved). The phases 2 and 3 were considered not achieved for PBB3 since no evidence exists about the ligand's diagnostic accuracy. Preliminary evidence exists about the secondary aims of each phase for all ligands. CONCLUSION Much work remains for completing the aims of phases 2 and 3 and replicating the available evidence. However, it is unlikely that the validation process for these tracers will be completed, given the presence of off-target binding and the development of second-generation tracers with improved binding and pharmacokinetic properties.
Collapse
|
31
|
Kawamura K, Hashimoto H, Furutsuka K, Ohkubo T, Fujishiro T, Togashi T, Arashi D, Sakai T, Muto M, Ogawa M, Kurihara Y, Nengaki N, Takei M, Nemoto K, Higuchi M, Zhang MR. Radiosynthesis and quality control testing of the tau imaging positron emission tomography tracer [ 18 F]PM-PBB3 for clinical applications. J Labelled Comp Radiopharm 2021; 64:109-119. [PMID: 33067819 DOI: 10.1002/jlcr.3890] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022]
Abstract
Recently, we produced 11 C-labeled 2-((1E,3E)-4-(6-(methylamino)pyridin-3-yl)buta-1,3-dienyl)benzo[d]thiazol-6-ol ([11 C]PBB3) as a clinically useful positron emission tomography (PET) tracer for in vivo imaging of tau pathologies in the human brain. To overcome the limitations (i.e., rapid in vivo metabolism and short half-life) of [11 C]PBB3, we further synthesized 18 F-labeled 1-fluoro-3-((2-((1E,3E)-4-(6-(methylamino)pyridine-3-yl)buta-1,3-dien-1-yl)benzo[d]thiazol-6-yl)oxy)propan-2-ol ([18 F]PM-PBB3). [18 F]PM-PBB3 is also a useful tau PET tracer for imaging tau pathologies. In this study, we developed a routine radiosynthesis and quality control testing of [18 F]PM-PBB3 for clinical applications. [18 F]PM-PBB3 was synthesized by direct 18 F-fluorination of the tosylated derivative, followed by removal of the protecting group. [18 F]PM-PBB3 was obtained with sufficient radioactivity (25 ± 6.0% of the nondecay-corrected radiochemical yield at the end of synthesis, EOS), radiochemical purity (98 ± 0.6%), and molar activity (350 ± 94 GBq/μmol at EOS; n = 53). Moreover, [18 F]PM-PBB3 consistently retained >95% of radiochemical purity for 60 min without undergoing photoisomerization using a new UV-cutoff light (yellow light) fixed in the hot cell to monitor the synthesis. All the results of the quality control testing for the [18 F]PM-PBB3 injection complied with our in-house quality control and quality assurance specifications. We have accomplished >200 production runs of [18 F]PM-PBB3 in our facility for various research purposes.
Collapse
Affiliation(s)
- Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroki Hashimoto
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kenji Furutsuka
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- SHI Accelerator Service Ltd., Tokyo, Japan
| | - Takayuki Ohkubo
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- SHI Accelerator Service Ltd., Tokyo, Japan
| | - Tomoya Fujishiro
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Tokyo Nuclear Services Co. Ltd., Tokyo, Japan
| | - Takahiro Togashi
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Tokyo Nuclear Services Co. Ltd., Tokyo, Japan
| | - Daisuke Arashi
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Tokyo Nuclear Services Co. Ltd., Tokyo, Japan
| | - Toshiyuki Sakai
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Tokyo Nuclear Services Co. Ltd., Tokyo, Japan
| | - Masatoshi Muto
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Tokyo Nuclear Services Co. Ltd., Tokyo, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- SHI Accelerator Service Ltd., Tokyo, Japan
| | - Yusuke Kurihara
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- SHI Accelerator Service Ltd., Tokyo, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- SHI Accelerator Service Ltd., Tokyo, Japan
| | - Makoto Takei
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kazuyoshi Nemoto
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
32
|
Xu XJ, Yang MS, Zhang B, Niu F, Dong JQ, Liu BY. Glucose metabolism: A link between traumatic brain injury and Alzheimer's disease. Chin J Traumatol 2021; 24:5-10. [PMID: 33358332 PMCID: PMC7878452 DOI: 10.1016/j.cjtee.2020.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI), a growing public health problem, is a leading cause of death and disability worldwide, although its prevention measures and clinical cares are substantially improved. Increasing evidence shows that TBI may increase the risk of mood disorders and neurodegenerative diseases, including Alzheimer's disease (AD). However, the complex relationship between TBI and AD remains elusive. Metabolic dysfunction has been the common pathology in both TBI and AD. On the one hand, TBI perturbs the glucose metabolism of the brain, and causes energy crisis and subsequent hyperglycolysis. On the other hand, glucose deprivation promotes amyloidogenesis via β-site APP cleaving enzyme-1 dependent mechanism, and triggers tau pathology and synaptic function. Recent findings suggest that TBI might facilitate Alzheimer's pathogenesis by altering metabolism, which provides clues to metabolic link between TBI and AD. In this review, we will explore how TBI-induced metabolic changes contribute to the development of AD.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, 100070, China,China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China,Corresponding author. Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
33
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
34
|
Ju Y, Tam KY. 9R, the cholinesterase and amyloid beta aggregation dual inhibitor, as a multifunctional agent to improve cognitive deficit and neuropathology in the triple-transgenic Alzheimer's disease mouse model. Neuropharmacology 2020; 181:108354. [DOI: 10.1016/j.neuropharm.2020.108354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
|
35
|
Osama A, Zhang J, Yao J, Yao X, Fang J. Nrf2: a dark horse in Alzheimer's disease treatment. Ageing Res Rev 2020; 64:101206. [PMID: 33144124 DOI: 10.1016/j.arr.2020.101206] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), an age-dependent neurodegenerative disorder, is the main cause of dementia. Common hallmarks of AD include the amyloid β-peptide (Aβ) aggregation, high levels of hyperphosphorylated tau protein (p-tau) and failure in redox homeostasis. To date, all proposed drugs affecting Aβ and/or p-tau have been failed in clinical trials. A decline in the expression of the transcription factor Nrf2 (nuclear factor-erythroid 2-p45 derived factor 2) and its driven genes (NQO1, HO-1, and GCLC), and alteration of the Nrf2-related pathways have been observed in AD brains. Nrf2 plays a critical role in maintaining cellular redox homeostasis and regulating inflammation response. Nrf2 activation also provides cytoprotection against increasing pathologies including neurodegenerative diseases. These lines of evidence imply that Nrf2 activation may be a novel AD treatment option. Interestingly, recent studies have also demonstrated that Nrf2 interferes with several key pathogenic processes in AD including Aβ and p-tau pathways. The current review aims to provide insights into the role of Nrf2 in AD. Also, we discuss the progress and challenges regarding the Nrf2 activators for AD treatment.
Collapse
Affiliation(s)
- Alsiddig Osama
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Juan Yao
- School of pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China.
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
36
|
Kreisl WC, Kim MJ, Coughlin JM, Henter ID, Owen DR, Innis RB. PET imaging of neuroinflammation in neurological disorders. Lancet Neurol 2020; 19:940-950. [PMID: 33098803 PMCID: PMC7912433 DOI: 10.1016/s1474-4422(20)30346-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
A growing need exists for reliable in-vivo measurement of neuroinflammation to better characterise the inflammatory processes underlying various diseases and to inform the development of novel therapeutics that target deleterious glial activity. PET is well suited to quantify neuroinflammation and has the potential to discriminate components of the neuroimmune response. However, there are several obstacles to the reliable quantification of neuroinflammation by PET imaging. Despite these challenges, PET studies have consistently identified associations between neuroimmune responses and pathophysiology in brain disorders such as Alzheimer's disease. Tissue studies have also begun to clarify the meaning of changes in PET signal in some diseases. Furthermore, although PET imaging of neuroinflammation does not have an established clinical application, novel targets are under investigation and a small but growing number of studies have suggested that this imaging modality could have a role in drug development. Future studies are needed to further improve our knowledge of the cellular mechanisms that underlie changes in PET signal, how immune response contributes to neurological disease, and how it might be therapeutically modified.
Collapse
Affiliation(s)
- William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Abstract
Sleep is evolutionarily conserved across all species, and impaired sleep is a common trait of the diseased brain. Sleep quality decreases as we age, and disruption of the regular sleep architecture is a frequent antecedent to the onset of dementia in neurodegenerative diseases. The glymphatic system, which clears the brain of protein waste products, is mostly active during sleep. Yet the glymphatic system degrades with age, suggesting a causal relationship between sleep disturbance and symptomatic progression in the neurodegenerative dementias. The ties that bind sleep, aging, glymphatic clearance, and protein aggregation have shed new light on the pathogenesis of a broad range of neurodegenerative diseases, for which glymphatic failure may constitute a therapeutically targetable final common pathway.
Collapse
Affiliation(s)
- Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
38
|
Nicastro N, Malpetti M, Mak E, Williams GB, Bevan-Jones WR, Carter SF, Passamonti L, Fryer TD, Hong YT, Aigbirhio FI, Rowe JB, O'Brien JT. Gray matter changes related to microglial activation in Alzheimer's disease. Neurobiol Aging 2020; 94:236-242. [PMID: 32663716 PMCID: PMC7456794 DOI: 10.1016/j.neurobiolaging.2020.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is increasingly recognized as playing a key pathogenetic role in Alzheimer's disease (AD). We examined the relationship between in vivo neuroinflammation and gray matter (GM) changes. Twenty-eight subjects with clinically probable AD (n = 14) and amyloid-positive mild cognitive impairment (n = 14) (age 71.9 ± 8.4 years, 46% female) and 24 healthy controls underwent structural 3T brain MRI. AD/mild cognitive impairment participants exhibited GM atrophy and cortical thinning in AD-related temporoparietal regions (false discovery rate-corrected p < 0.05). Patients also showed increased microglial activation in temporal cortices. Higher 11C-PK11195 binding in these regions was associated with reduced volume and cortical thickness in parietal, occipital, and cingulate areas (false discovery rate p < 0.05). Hippocampal GM atrophy and parahippocampal cortical thinning were related to worse cognition (p < 0.05), but these effects were not mediated by microglial activation. This study demonstrates an association between in vivo microglial activation and markers of GM damage in AD, positioning neuroinflammation as a potential target for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nicolas Nicastro
- Department of Psychiatry, University of Cambridge, Cambridge, UK; Department of Clinical Neurosciences, Geneva University Hospitals, Switzerland.
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Wolfson Brain Imaging Centre, Cognition University of Cambridge, Cambridge, UK
| | | | - Stephen F Carter
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Consiglio Nazionale delle Ricerche (CNR), Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Milano, Italy
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Wolfson Brain Imaging Centre, Cognition University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Wolfson Brain Imaging Centre, Cognition University of Cambridge, Cambridge, UK
| | | | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
39
|
Steroids and Alzheimer's Disease: Changes Associated with Pathology and Therapeutic Potential. Int J Mol Sci 2020; 21:ijms21134812. [PMID: 32646017 PMCID: PMC7370115 DOI: 10.3390/ijms21134812] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial age-related neurodegenerative disease that today has no effective treatment to prevent or slow its progression. Neuroactive steroids, including neurosteroids and sex steroids, have attracted attention as potential suitable candidates to alleviate AD pathology. Accumulating evidence shows that they exhibit pleiotropic neuroprotective properties that are relevant for AD. This review focuses on the relationship between selected neuroactive steroids and the main aspects of AD disease, pointing out contributions and gaps with reference to sex differences. We take into account the regulation of brain steroid concentrations associated with human AD pathology. Consideration is given to preclinical studies in AD models providing current knowledge on the neuroprotection offered by neuroactive (neuro)steroids on major AD pathogenic factors, such as amyloid-β (Aβ) and tau pathology, mitochondrial impairment, neuroinflammation, neurogenesis and memory loss. Stimulating endogenous steroid production opens a new steroid-based strategy to potentially overcome AD pathology. This article is part of a Special Issue entitled Steroids and the Nervous System.
Collapse
|
40
|
Mestre H, Mori Y, Nedergaard M. The Brain's Glymphatic System: Current Controversies. Trends Neurosci 2020; 43:458-466. [PMID: 32423764 DOI: 10.1016/j.tins.2020.04.003] [Citation(s) in RCA: 341] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/16/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
The glymphatic concept along with the discovery of meningeal lymphatic vessels have, in recent years, highlighted that fluid is directionally transported within the central nervous system (CNS). Imaging studies, as well as manipulations of fluid transport, point to a key role of the glymphatic-lymphatic system in clearance of amyloid-β and other proteins. As such, the glymphatic-lymphatic system represents a new target in combating neurodegenerative diseases. Not unexpectedly, introduction of a new plumbing system in the brain has stirred controversies. This opinion article will highlight what we know about the brain's fluid transport systems, where experimental data are lacking, and what is still debated.
Collapse
Affiliation(s)
- Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
41
|
Malpetti M, Kievit RA, Passamonti L, Jones PS, Tsvetanov KA, Rittman T, Mak E, Nicastro N, Bevan-Jones WR, Su L, Hong YT, Fryer TD, Aigbirhio FI, O’Brien JT, Rowe JB. Microglial activation and tau burden predict cognitive decline in Alzheimer's disease. Brain 2020; 143:1588-1602. [PMID: 32380523 PMCID: PMC7241955 DOI: 10.1093/brain/awaa088] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 02/07/2020] [Indexed: 11/12/2022] Open
Abstract
Tau pathology, neuroinflammation, and neurodegeneration are key aspects of Alzheimer's disease. Understanding whether these features predict cognitive decline, alone or in combination, is crucial to develop new prognostic measures and enhanced stratification for clinical trials. Here, we studied how baseline assessments of in vivo tau pathology (measured by 18F-AV-1451 PET), neuroinflammation (measured by 11C-PK11195 PET) and brain atrophy (derived from structural MRI) predicted longitudinal cognitive changes in patients with Alzheimer's disease pathology. Twenty-six patients (n = 12 with clinically probable Alzheimer's dementia and n = 14 with amyloid-positive mild cognitive impairment) and 29 healthy control subjects underwent baseline assessment with 18F-AV-1451 PET, 11C-PK11195 PET, and structural MRI. Cognition was examined annually over the subsequent 3 years using the revised Addenbrooke's Cognitive Examination. Regional grey matter volumes, and regional binding of 18F-AV-1451 and 11C-PK11195 were derived from 15 temporo-parietal regions characteristically affected by Alzheimer's disease pathology. A principal component analysis was used on each imaging modality separately, to identify the main spatial distributions of pathology. A latent growth curve model was applied across the whole sample on longitudinal cognitive scores to estimate the rate of annual decline in each participant. We regressed the individuals' estimated rate of cognitive decline on the neuroimaging components and examined univariable predictive models with single-modality predictors, and a multi-modality predictive model, to identify the independent and combined prognostic value of the different neuroimaging markers. Principal component analysis identified a single component for the grey matter atrophy, while two components were found for each PET ligand: one weighted to the anterior temporal lobe, and another weighted to posterior temporo-parietal regions. Across the whole-sample, the single-modality models indicated significant correlations between the rate of cognitive decline and the first component of each imaging modality. In patients, both stepwise backward elimination and Bayesian model selection revealed an optimal predictive model that included both components of 18F-AV-1451 and the first (i.e. anterior temporal) component for 11C-PK11195. However, the MRI-derived atrophy component and demographic variables were excluded from the optimal predictive model of cognitive decline. We conclude that temporo-parietal tau pathology and anterior temporal neuroinflammation predict cognitive decline in patients with symptomatic Alzheimer's disease pathology. This indicates the added value of PET biomarkers in predicting cognitive decline in Alzheimer's disease, over and above MRI measures of brain atrophy and demographic data. Our findings also support the strategy for targeting tau and neuroinflammation in disease-modifying therapy against Alzheimer's disease.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rogier A Kievit
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Institute of Molecular Bioimaging and Physiology, National Research Council, Milano, Italy
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kamen A Tsvetanov
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Nicolas Nicastro
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, Geneva University Hospitals, Switzerland
| | | | - Li Su
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Trust, Cambridge, UK
| |
Collapse
|
42
|
Penke B, Szűcs M, Bogár F. Oligomerization and Conformational Change Turn Monomeric β-Amyloid and Tau Proteins Toxic: Their Role in Alzheimer's Pathogenesis. Molecules 2020; 25:molecules25071659. [PMID: 32260279 PMCID: PMC7180792 DOI: 10.3390/molecules25071659] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/29/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The structural polymorphism and the physiological and pathophysiological roles of two important proteins, β-amyloid (Aβ) and tau, that play a key role in Alzheimer's disease (AD) are reviewed. Recent results demonstrate that monomeric Aβ has important physiological functions. Toxic oligomeric Aβ assemblies (AβOs) may play a decisive role in AD pathogenesis. The polymorph fibrillar Aβ (fAβ) form has a very ordered cross-β structure and is assumed to be non-toxic. Tau monomers also have several important physiological actions; however, their oligomerization leads to toxic oligomers (TauOs). Further polymerization results in probably non-toxic fibrillar structures, among others neurofibrillary tangles (NFTs). Their structure was determined by cryo-electron microscopy at atomic level. Both AβOs and TauOs may initiate neurodegenerative processes, and their interactions and crosstalk determine the pathophysiological changes in AD. TauOs (perhaps also AβO) have prionoid character, and they may be responsible for cell-to-cell spreading of the disease. Both extra- and intracellular AβOs and TauOs (and not the previously hypothesized amyloid plaques and NFTs) may represent the novel targets of AD drug research.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (M.S.); (F.B.)
- Correspondence:
| | - Mária Szűcs
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (M.S.); (F.B.)
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (M.S.); (F.B.)
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
43
|
Lin J, Lin H, Ma C, Dong F, Hu Y, Li H. MiR-149 Aggravates Pyroptosis in Myocardial Ischemia-Reperfusion Damage via Silencing FoxO3. Med Sci Monit 2019; 25:8733-8743. [PMID: 31741467 PMCID: PMC6880628 DOI: 10.12659/msm.918410] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs), which modulate the expression of their target genes, are commonly involved in stimulating and adjusting of many processes that result in cardiovascular diseases, contain cardiac ischemia/reperfusion (I/R) damage. However, the expression and role of miR-149 in pyroptosis mediated myocardial I/R damage remains unclear. MATERIAL AND METHODS Real-time polymerase chain reaction was performed to measure the miR-149 and FoxO3 expression in I/R stimulated H9C2 cells. The cell proliferation, pyroptosis-related inflammatory genes in I/R-treated H9C2 cells transfected miR-149 mimics or miR-149 inhibitor were both explored. We predicted and confirmed miR-149 targets by using bioinformatics analyses and luciferase reporter assay. In addition, the potential relationship between miR-149 and FoxO3 in pyroptosis from I/R treated H9C2 cells was analyzed. RESULTS Our results showed that miR-149 was upregulated, while FoxO3 was downregulated in I/R stimulated H9C2 cells. Over-expression of miR-149 inhibited cell viability and promote pyroptosis, however, down-expression of miR-149 had an opposite effect in I/R treated H9C2 cells. Furthermore, miR-149 could negatively regulate FoxO3 expression by binding 3'UTR, whereas silencing of FoxO3 attenuated the effect of miR-149-mimics on cell proliferation and pyroptosis in I/R treated H9C2 cells. CONCLUSIONS Our study found that miR-149 played a critical role in pyroptosis during cardiac I/R injury, and thus, might provide a novel therapeutic target.
Collapse
Affiliation(s)
- Jie Lin
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Haihuan Lin
- Laboratory Medicine Humboldt University, Charité University Medicine, Berlin, China (mainland)
| | - Chao Ma
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Berlin, China (mainland)
| | - Fengquan Dong
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Yingchun Hu
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Haiying Li
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| |
Collapse
|
44
|
Prospects and challenges of imaging neuroinflammation beyond TSPO in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2019; 46:2831-2847. [PMID: 31396666 PMCID: PMC6879435 DOI: 10.1007/s00259-019-04462-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation, as defined by the activation of microglia and astrocytes, has emerged in the last years as a key element of the pathogenesis of neurodegenerative diseases based on genetic findings and preclinical and human studies. This has raised the need for new methodologies to assess and follow glial activation in patients, prompting the development of PET ligands for molecular imaging of glial cells and novel structural MRI and DTI tools leading to a multimodal approach. The present review describes the recent advancements in microglia and astrocyte biology in the context of health, ageing, and Alzheimer's disease, the most common dementia worldwide. The review further delves in molecular imaging discussing the challenges associated with past and present targets, including conflicting findings, and finally, presenting novel methodologies currently explored to improve our in vivo knowledge of the neuroinflammatory patterns in Alzheimer's disease. With glial cell activation as a potential therapeutic target in neurodegenerative diseases, the translational research between cell biologists, chemists, physicists, radiologists, and neurologists should be strengthened.
Collapse
|