1
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
2
|
Zhou Y, Li C, Wu R, Yin H, Liu G, Meng H, Xie W, Birar VC, Wang C, Wu X, Bai P. Molecular Imaging Reveals Antineuroinflammatory Effects of HDAC6 Inhibition in Stroke Models. Mol Pharm 2024. [PMID: 39504500 DOI: 10.1021/acs.molpharmaceut.4c01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Ischemic stroke is a devastating disease that causes neuronal death, neuroinflammation, and other cerebral damage. However, effective therapeutic strategies for ischemic stroke are still lacking. Histone deacetylase 6 (HDAC6) has been implicated in the pathogenesis of ischemic stroke, and the pharmacological inhibition of HDAC6 has shown promising neuroprotective effects. In this study, we utilized positron emission tomography (PET) imaging with the HDAC6-specific radioligand [18F]PB118 to investigate the dynamic changes of HDAC6 expression in the brain after ischemic injury. The results revealed a significant decline in [18F]PB118 uptake in the ipsilateral hemisphere on the first day after ischemia, followed by a gradual increase on days 4 and 7. To evaluate the therapeutic potential of HDAC6 inhibitors, we developed a novel brain-permeable and potent HDAC6 inhibitor, PB131, and assessed its neuroprotective effects in an ischemic stroke mouse model. PET imaging studies demonstrated that PB131 treatment alleviated the decline in [18F]PB118 uptake and reduced the infarct size in middle cerebral artery occlusion mice. Furthermore, PET imaging with the TSPO-specific radioligand [18F]FEPPA revealed that PB131 significantly suppressed neuroinflammation in the ischemic brain. These findings provide insights into the dynamic changes of HDAC6 in ischemic stroke and the potential of HDAC6 inhibitors as novel therapeutic agents for this condition.
Collapse
Affiliation(s)
- Yanting Zhou
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu 61000, China
| | - Rui Wu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Honghai Yin
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Liu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Meng
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weiyao Xie
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Vishal C Birar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Bai
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
3
|
Guttuso T, Zhu J, Wilding GE. Lithium Aspartate for Long COVID Fatigue and Cognitive Dysfunction: A Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2436874. [PMID: 39356507 PMCID: PMC11447566 DOI: 10.1001/jamanetworkopen.2024.36874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Importance Neurologic post-COVID-19 condition (PCC), or long COVID, symptoms of fatigue and cognitive dysfunction continue to affect millions of people who have been infected with SARS-CoV-2. There currently are no effective evidence-based therapies available for treating neurologic PCC. Objective To assess the effects of lithium aspartate therapy on PCC fatigue and cognitive dysfunction. Design, Setting, and Participants A randomized, double-blind, placebo-controlled trial (RCT) enrolling participants in a neurology clinic from November 28, 2022, to June 29, 2023, with 3 weeks of follow-up, was conducted. Subsequently, an open-label lithium dose-finding study with 6 weeks of follow-up was performed among the same participants enrolled in the RCT. Eligible individuals needed to report new, bothersome fatigue or cognitive dysfunction persisting for more than 4 weeks after a self-reported positive test for COVID-19, Fatigue Severity Scale-7 (FSS-7) or Brain Fog Severity Scale (BFSS) score of 28 or greater, Beck Depression Inventory-II score less than 24, and no history of a condition known to cause fatigue or cognitive dysfunction. All participants in the RCT were eligible for the dose-finding study, except for those who responded to the placebo. Intention-to-treat analysis was used. Intervention Lithium aspartate, 10 to 15 mg/d, or identically appearing placebo for 3 weeks followed by open-label lithium aspartate, 10 to 15 mg/d, for 2 weeks. In the subsequent dose-finding study, open-label lithium aspartate dosages up to 45 mg/d for 6 weeks were given. Main Outcomes and Measures Change in sum of FSS-7 and BFSS scores. The scores for each measure range from 7 to 49, with higher scores indicating more severe symptoms. Secondary outcomes included changes from baseline in the scores of additional questionnaires. Results Fifty-two participants were enrolled (30 [58%] males; mean [SD] age, 58.54 [14.34] years) and 26 were randomized to treatment with lithium aspartate (10 females) and 26 to placebo (12 female). Two participants assigned to lithium aspartate were lost to follow-up and none withdrew. No adverse events were attributable to lithium therapy. There were no significant intergroup differences for the primary outcome (-3.6; 95% CI, -16.6 to 9.5; P = .59) or any secondary outcomes. Among 3 patients completing a subsequent dose-finding study, open-label lithium aspartate, 40 to 45 mg/d, was associated with numerically greater reductions in fatigue and cognitive dysfunction scores than 15 mg/d, particularly in 2 patients with serum lithium levels of 0.18 and 0.49 mEq/L compared with 1 patient with a level of 0.10 mEq/L. Conclusions and Relevance In this RCT, therapy with lithium aspartate, 10 to 15 mg/d, was ineffective for neurologic PCC fatigue and cognitive dysfunction. Another RCT is required to assess the potential benefits of higher lithium dosages for treating neurologic PCC. Trial Registration ClinicalTrials.gov Identifier: NCT05618587 and NCT06108297.
Collapse
Affiliation(s)
- Thomas Guttuso
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Williamsville, New York
| | - Jingtao Zhu
- Department of Biostatistics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Williamsville, New York
| | - Gregory E. Wilding
- Department of Biostatistics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Williamsville, New York
| |
Collapse
|
4
|
Swann P, Mirza-Davies A, O'Brien J. Associations Between Neuropsychiatric Symptoms and Inflammation in Neurodegenerative Dementia: A Systematic Review. J Inflamm Res 2024; 17:6113-6141. [PMID: 39262651 PMCID: PMC11389708 DOI: 10.2147/jir.s385825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Background Neuropsychiatric symptoms are common in dementia and linked to adverse outcomes. Inflammation is increasingly recognized as playing a role as a driver of early disease progression in Alzheimer's disease (AD) and related dementias. Inflammation has also been linked to primary psychiatric disorders, however its association with neuropsychiatric symptoms in neurodegenerative dementias remains uncertain. Methods We conducted a systematic literature review investigating associations between inflammation and neuropsychiatric symptoms in neurodegenerative dementias, including AD, Lewy body, Frontotemporal, Parkinson's (PD) and Huntington's disease dementias. Results Ninety-nine studies met our inclusion criteria, and the majority (n = 59) investigated AD and/or mild cognitive impairment (MCI). Thirty-five studies included PD, and only 6 investigated non-AD dementias. Inflammation was measured in blood, CSF, by genotype, brain tissue and PET imaging. Overall, studies exhibited considerable heterogeneity and evidence for specific inflammatory markers was inconsistent, with lack of replication and few longitudinal studies with repeat biomarkers. Depression was the most frequently investigated symptom. In AD, some studies reported increases in peripheral IL-6, TNF-a associated with depressive symptoms. Preliminary investigations using PET measures of microglial activation found an association with agitation. In PD, studies reported positive associations between TNF-a, IL-6, CRP, MCP-1, IL-10 and depression. Conclusion Central and peripheral inflammation may play a role in neuropsychiatric symptoms in neurodegenerative dementias; however, the evidence is inconsistent. There is a need for multi-site longitudinal studies with detailed assessments of neuropsychiatric symptoms combined with replicable peripheral and central markers of inflammation.
Collapse
Affiliation(s)
- Peter Swann
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Anastasia Mirza-Davies
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
5
|
Salerno S, Viviano M, Baglini E, Poggetti V, Giorgini D, Castagnoli J, Barresi E, Castellano S, Da Settimo F, Taliani S. TSPO Radioligands for Neuroinflammation: An Overview. Molecules 2024; 29:4212. [PMID: 39275061 PMCID: PMC11397380 DOI: 10.3390/molecules29174212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
The translocator protein (TSPO) is predominately localized on the outer mitochondrial membrane in steroidogenic cells. In the brain, TSPO expression, low under normal conditions, results upregulated in response to glial cell activation, that occurs in neuroinflammation. As a consequence, TSPO has been extensively studied as a biomarker of such conditions by means of TSPO-targeted radiotracers. Although [11C]-PK11195, the prototypical TSPO radioligand, is still widely used for in vivo studies, it is endowed with severe limitations, mainly low sensitivity and poor amenability to quantification. Consequently, several efforts have been focused on the design of new radiotracers for the in vivo imaging of TSPO. The present review will provide an outlook on the latest advances in TSPO radioligands for neuroinflammation imaging. The final goal is to pave the way for (radio)chemists in the future design and development of novel effective and sensitive radiopharmaceuticals targeting TSPO.
Collapse
Affiliation(s)
- Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Doralice Giorgini
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Jacopo Castagnoli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| |
Collapse
|
6
|
Mei J, Li Y, Niu L, Liang R, Tang M, Cai Q, Xu J, Zhang D, Yin X, Liu X, Shen Y, Liu J, Xu M, Xia P, Ling J, Wu Y, Liang J, Zhang J, Yu P. SGLT2 inhibitors: a novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl Neurodegener 2024; 13:41. [PMID: 39123214 PMCID: PMC11312905 DOI: 10.1186/s40035-024-00431-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.
Collapse
Affiliation(s)
- Jiaqi Mei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Yi Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Ruikai Liang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyue Tang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingdong Xu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
7
|
Xu J, Xie L, Yin J, Shi X, Dong K, Tao J, Xu W, Ma D, Zhang S, Chen J, Yang Y. A High-Carbohydrate Diet Induces Cognitive Impairment and Promotes Amyloid Burden and Tau Phosphorylation via PI3K/Akt/GSK-3β Pathway in db/db Mice. Biomedicines 2024; 12:1701. [PMID: 39200168 PMCID: PMC11351503 DOI: 10.3390/biomedicines12081701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND Cognitive impairment is a prevalent complication of type 2 diabetes, influenced significantly by various dietary patterns. High-carbohydrate diets (HCDs) are commonly consumed nowadays; however, the specific impact of HCDs on cognitive function in diabetes remains unclear. METHODS The objective of this study was to investigate whether an HCD has effects on cognition in diabetes. Eight-week-old diabetic (db/db) mice and wild-type (WT) mice underwent a twelve-week dietary intervention, including a normal diet (ND), an HCD, or a high-fat diet (HFD). Following this, behavioral tests were conducted, and related hippocampal pathology was evaluated. RESULTS Our results demonstrated that an HCD exacerbated cognitive decline in db/db mice compared to an ND. Additionally, an HCD increased amyloid-β burden and expression of β-site APP cleaving enzyme-1. An HCD was also found to promote the phosphorylation of tau protein via the PI3K/Akt/GSK-3β pathway. Furthermore, an HCD markedly induced neuroinflammation and increased the quantity of microglia and astrocytes. However, these damages induced by an HCD were less severe than those caused by an HFD. CONCLUSIONS Collectively, our findings indicate that a high intake of carbohydrates can have an adverse impact on cognitive function in diabetes.
Collapse
Affiliation(s)
- Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Jiaxin Yin
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Jing Tao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Delin Ma
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Juan Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| |
Collapse
|
8
|
Cai Y, Zhang Y, Leng S, Ma Y, Jiang Q, Wen Q, Ju S, Hu J. The relationship between inflammation, impaired glymphatic system, and neurodegenerative disorders: A vicious cycle. Neurobiol Dis 2024; 192:106426. [PMID: 38331353 DOI: 10.1016/j.nbd.2024.106426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
The term "glymphatic" emerged roughly a decade ago, marking a pivotal point in neuroscience research. The glymphatic system, a glial-dependent perivascular network distributed throughout the brain, has since become a focal point of investigation. There is increasing evidence suggesting that impairment of the glymphatic system appears to be a common feature of neurodegenerative disorders, and this impairment exacerbates as disease progression. Nevertheless, the common factors contributing to glymphatic system dysfunction across most neurodegenerative disorders remain unclear. Inflammation, however, is suspected to play a pivotal role. Dysfunction of the glymphatic system can lead to a significant accumulation of protein and waste products, which can trigger inflammation. The interaction between the glymphatic system and inflammation appears to be cyclical and potentially synergistic. Yet, current research is limited, and there is a lack of comprehensive models explaining this association. In this perspective review, we propose a novel model suggesting that inflammation, impaired glymphatic function, and neurodegenerative disorders interconnected in a vicious cycle. By presenting experimental evidence from the existing literature, we aim to demonstrate that: (1) inflammation aggravates glymphatic system dysfunction, (2) the impaired glymphatic system exacerbated neurodegenerative disorders progression, (3) neurodegenerative disorders progression promotes inflammation. Finally, the implication of proposed model is discussed.
Collapse
Affiliation(s)
- Yu Cai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yangqiqi Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Shuo Leng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Yuanyuan Ma
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, 2799 W Grand Blvd, Detroit, MI 48202, USA
| | - Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W.16th Street, Indianapolis, IN 46202-5188, USA
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Jiani Hu
- Department of Radiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
9
|
Zhou L, Yang C, Liu Z, Chen L, Wang P, Zhou Y, Yuan M, Zhou LT, Wang X, Zhu LQ. Neuroprotective effect of the traditional decoction Tian-Si-Yin against Alzheimer's disease via suppression of neuroinflammation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117569. [PMID: 38086513 DOI: 10.1016/j.jep.2023.117569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is the most prevalent neurodegenerative disease among old adults. As a traditional Chinese medicine, the herbal decoction Tian-Si-Yin consists of Morinda officinalis How. and Cuscuta chinensis Lam., which has been widely used to nourish kidney. Interestingly, Tian-Si-Yin has also been used to treat dementia, depression and other neurological conditions. However, its therapeutic potential for neurodegenerative diseases such as AD and the underlying mechanisms remain unclear. AIM OF THE STUDY To evaluate the therapeutic effect of the herbal formula Tian-Si-Yin against AD and to explore the underlying mechanisms. MATERIALS AND METHODS The N2a cells treated with amyloid β (Aβ) peptide or overexpressing amyloid precursor protein (APP) were used to establish cellular models of AD. The in vivo anti-AD effects were evaluated by using Caenorhabditis elegans and 3 × Tg-AD mouse models. Tian-Si-Yin was orally administered to the mice for 8 weeks at a dose of 10, 15 or 20 mg/kg/day, respectively. Its protective role on memory deficits of mice was examined using the Morris water maze and fear conditioning tests. Network pharmacology, proteomic analysis and ultra-high performance liquid chromatography-mass spectrometry/mass spectrometry (UHPLC-MS/MS) were used to explore the underlying molecular mechanisms, which were further investigated by Western blotting and immunohistochemistry. RESULTS Tian-Si-Yin was shown to improve cell viability of Aβ-treated N2a cells and APP-expressing N2a-APP cells. Tian-Si-Yin was also found to reduce ROS level and extend lifespan of transgenic AD-like C. elegans model. Oral administration of Tian-Si-Yin at medium dose was able to effectively rescue memory impairment in 3 × Tg mice. Tian-Si-Yin was further shown to suppress neuroinflammation by inhibition of glia cell activation and downregulation of inflammatory cytokines, diminishing tau phosphoralytion and Aβ deposition in the mice. Using UHPLC-MS/MS and network pharmacology technologies, 17 phytochemicals from 68 components of Tian-Si-Yin were identified as potential anti-AD components. MAPK1, BRAF, TTR and Fyn were identified as anti-AD targets of Tian-Si-Yin from network pharmacology and mass spectrum. CONCLUSIONS This study has established the protective effect of Tian-Si-Yin against AD and demonstrates that Tian-Si-Yin is capable of improving Aβ level, tau pathology and synaptic disorder by regulating inflammatory response.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chunqing Yang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhiqiang Liu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Linlin Chen
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, PR China
| | - Ping Wang
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, PR China
| | - Yuan Zhou
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, PR China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, PR China
| | - Lan-Ting Zhou
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, PR China; Neuroscience and Brainscience Institute of Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China.
| | - Xueren Wang
- Department of Anesthesiology, Shanxi Bethune Hospital, Taiyuan, PR China; Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
10
|
Wang Q, Schindler SE, Chen G, Mckay NS, McCullough A, Flores S, Liu J, Sun Z, Wang S, Wang W, Hassenstab J, Cruchaga C, Perrin RJ, Fagan AM, Morris JC, Wang Y, Benzinger TLS. Investigating White Matter Neuroinflammation in Alzheimer Disease Using Diffusion-Based Neuroinflammation Imaging. Neurology 2024; 102:e208013. [PMID: 38315956 PMCID: PMC10890836 DOI: 10.1212/wnl.0000000000208013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/13/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Alzheimer disease (AD) is primarily associated with accumulations of amyloid plaques and tau tangles in gray matter, however, it is now acknowledged that neuroinflammation, particularly in white matter (WM), significantly contributes to the development and progression of AD. This study aims to investigate WM neuroinflammation in the continuum of AD and its association with AD pathologies and cognition using diffusion-based neuroinflammation imaging (NII). METHODS This is a cross-sectional, single-center, retrospective evaluation conducted on an observational study of 310 older research participants who were enrolled in the Knight Alzheimer's Disease Research Center cohort. Hindered water ratio (HR), an index of WM neuroinflammation, was quantified by a noninvasive diffusion MRI method, NII. The alterations of NII-HR were investigated at different AD stages, classified based on CSF concentrations of β-amyloid (Aβ) 42/Aβ40 for amyloid and phosphorylated tau181 (p-tau181) for tau. On the voxel and regional levels, the relationship between NII-HR and CSF markers of amyloid, tau, and neuroinflammation were examined, as well as cognition. RESULTS This cross-sectional study included 310 participants (mean age 67.1 [±9.1] years), with 52 percent being female. Subgroups included 120 individuals (38.7%) with CSF measures of soluble triggering receptor expressed on myeloid cells 2, 80 participants (25.8%) with CSF measures of chitinase-3-like protein 1, and 110 individuals (35.5%) with longitudinal cognitive measures. The study found that cognitively normal individuals with positive CSF Aβ42/Aβ40 and p-tau181 had higher HR than healthy controls and those with positive CSF Aβ42/Aβ40 but negative p-tau181. WM tracts with elevated NII-HR in individuals with positive CSF Aβ42/Aβ40 and p-tau181 were primarily located in the posterior brain regions while those with elevated NII-HR in individuals with positive CSF Aβ42/Aβ40 and p-tau181 connected the posterior and anterior brain regions. A significant negative correlation between NII-HR and CSF Aβ42/Aβ40 was found in individuals with positive CSF Aβ42/Aβ40. Baseline NII-HR correlated with baseline cognitive composite score and predicted longitudinal cognitive decline. DISCUSSION Those findings suggest that WM neuroinflammation undergoes alterations before the onset of AD clinical symptoms and that it interacts with amyloidosis. This highlights the potential value of noninvasive monitoring of WM neuroinflammation in AD progression and treatment.
Collapse
Affiliation(s)
- Qing Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Austin McCullough
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Wenshang Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Richard J Perrin
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), 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., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
11
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Hsieh HH, Chu PA, Lin YH, Kao YCJ, Chung YH, Hsu ST, Mo JM, Wu CY, Peng SL. Imaging diabetic cardiomyopathy in a type 1 diabetic rat model using 18F-FEPPA PET. Nucl Med Biol 2024; 128-129:108878. [PMID: 38324923 DOI: 10.1016/j.nucmedbio.2024.108878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Diabetic patients often experience chronic inflammation and fibrosis in their cardiac tissues, highlighting the pressing need for the development of sensitive diagnostic methods for longitudinal assessment of diabetic cardiomyopathy. This study aims to evaluate the significance of an inflammatory marker known as translocator protein (TSPO) in a positron emission tomography (PET) protocol for longitudinally monitoring cardiac dysfunction in a diabetic animal model. Additionally, we compared the commonly used radiotracer, 18F-fluoro-2-deoxy-d-glucose (18F-FDG). METHODS Fourteen 7-week-old female Sprague-Dawley rats were used in this study. Longitudinal PET experiments were conducted using 18F-N-2-(2-fluoroethoxy)benzyl)-N-(4-phenoxypyridin-3-yl)acetamide (18F-FEPPA) (n = 3), the TSPO radiotracer, and 18F-FDG (n = 3), both before and after the onset of diabetes. Histological and immunohistochemical staining assays were also conducted in both the control (n = 4) and diabetes (n = 4) groups. RESULTS Results indicated a significant increase in cardiac tissue uptake of 18F-FEPPA after the onset of diabetes (P < 0.05), aligning with elevated TSPO levels observed in diabetic animals according to histological data. Conversely, the uptake of 18F-FDG in cardiac tissue significantly decreased after the onset of diabetes (P < 0.05). CONCLUSION These findings suggest that 18F-FEPPA can function as a sensitive probe for detecting chronic inflammation and fibrosis in the cardiac tissues of diabetic animals.
Collapse
Affiliation(s)
- Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan
| | - Pei-An Chu
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Chieh Jill Kao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan
| | - Yi-Hsiu Chung
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Ting Hsu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jia-Min Mo
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan.
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan; Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
13
|
Złotek M, Kurowska A, Herbet M, Piątkowska-Chmiel I. GLP-1 Analogs, SGLT-2, and DPP-4 Inhibitors: A Triad of Hope for Alzheimer's Disease Therapy. Biomedicines 2023; 11:3035. [PMID: 38002034 PMCID: PMC10669527 DOI: 10.3390/biomedicines11113035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's is a prevalent, progressive neurodegenerative disease marked by cognitive decline and memory loss. The disease's development involves various pathomechanisms, including amyloid-beta accumulation, neurofibrillary tangles, oxidative stress, inflammation, and mitochondrial dysfunction. Recent research suggests that antidiabetic drugs may enhance neuronal survival and cognitive function in diabetes. Given the well-documented correlation between diabetes and Alzheimer's disease and the potential shared mechanisms, this review aimed to comprehensively assess the potential of new-generation anti-diabetic drugs, such as GLP-1 analogs, SGLT-2 inhibitors, and DPP-4 inhibitors, as promising therapeutic approaches for Alzheimer's disease. This review aims to comprehensively assess the potential therapeutic applications of novel-generation antidiabetic drugs, including GLP-1 analogs, SGLT-2 inhibitors, and DPP-4 inhibitors, in the context of Alzheimer's disease. In our considered opinion, antidiabetic drugs offer a promising avenue for groundbreaking developments and have the potential to revolutionize the landscape of Alzheimer's disease treatment.
Collapse
Affiliation(s)
| | | | | | - Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland; (M.Z.); (A.K.); (M.H.)
| |
Collapse
|
14
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
15
|
Syed AU, Liang C, Patel KK, Mondal R, Kamalia VM, Moran TR, Ahmed ST, Mukherjee J. Comparison of Monoamine Oxidase-A, Aβ Plaques, Tau, and Translocator Protein Levels in Postmortem Human Alzheimer's Disease Brain. Int J Mol Sci 2023; 24:10808. [PMID: 37445985 PMCID: PMC10341404 DOI: 10.3390/ijms241310808] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Increased monoamine oxidase-A (MAO-A) activity in Alzheimer's disease (AD) may be detrimental to the point of neurodegeneration. To assess MAO-A activity in AD, we compared four biomarkers, Aβ plaques, tau, translocator protein (TSPO), and MAO-A in postmortem AD. Radiotracers were [18F]FAZIN3 for MAO-A, [18F]flotaza and [125I]IBETA for Aβ plaques, [124/125I]IPPI for tau, and [18F]FEPPA for TSPO imaging. Brain sections of the anterior cingulate (AC; gray matter GM) and corpus callosum (CC; white matter WM) from cognitively normal control (CN, n = 6) and AD (n = 6) subjects were imaged using autoradiography and immunostaining. Using competition with clorgyline and (R)-deprenyl, the binding of [18F]FAZIN3 was confirmed to be selective to MAO-A levels in the AD brain sections. Increases in MAO-A, Aβ plaque, tau, and TSPO activity were found in the AD brains compared to the control brains. The [18F]FAZIN3 ratio in AD GM versus CN GM was 2.80, suggesting a 180% increase in MAO-A activity. Using GM-to-WM ratios of AD versus CN, a >50% increase in MAO-A activity was observed (AD/CN = 1.58). Linear positive correlations of [18F]FAZIN3 with [18F]flotaza, [125I]IBETA, and [125I]IPPI were measured and suggested an increase in MAO-A activity with increases in Aβ plaques and tau activity. Our results support the finding that MAO-A activity is elevated in the anterior cingulate cortex in AD and thus may provide a new biomarker for AD in this brain region.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
16
|
Ayton S, Janelidze S, Kalinowski P, Palmqvist S, Belaidi AA, Stomrud E, Roberts A, Roberts B, Hansson O, Bush AI. CSF ferritin in the clinicopathological progression of Alzheimer's disease and associations with APOE and inflammation biomarkers. J Neurol Neurosurg Psychiatry 2023; 94:211-219. [PMID: 36357168 PMCID: PMC9992756 DOI: 10.1136/jnnp-2022-330052] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/23/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUND A putative role for iron in driving Alzheimer's disease (AD) progression is complicated by previously reported associations with neuroinflammation, apolipoprotein E and AD proteinopathy. To establish how iron interacts with clinicopathological features of AD and at what disease stage iron influences cognitive outcomes, we investigated the association of cerebrospinal fluid (CSF) biomarkers of iron (ferritin), inflammation (acute phase response proteins) and apolipoproteins with pathological biomarkers (CSF Aβ42/t-tau, p-tau181), clinical staging and longitudinal cognitive deterioration in subjects from the BioFINDER cohort, with replication of key results in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. METHODS Ferritin, acute phase response proteins (n=9) and apolipoproteins (n=6) were measured in CSF samples from BioFINDER (n=1239; 4 years cognitive follow-up) participants stratified by cognitive status (cognitively unimpaired, mild cognitive impairment, AD) and for the presence of amyloid and tangle pathology using CSF Aβ42/t-tau (A+) and p-tau181 (T+). The ferritin and apolipoprotein E associations were replicated in the ADNI (n=264) cohort. RESULTS In both cohorts, ferritin and apoE were elevated in A-T+ and A+T+ subjects (16%-40%), but not clinical diagnosis. Other apolipoproteins and acute phase response proteins increased with clinical diagnosis, not pathology. CSF ferritin was positively associated with p-tau181, which was mediated by apolipoprotein E. An optimised threshold of ferritin predicted cognitive deterioration in mild cognitive impairment subjects in the BioFINDER cohort, especially those people classified as A-T- and A+T-. CONCLUSIONS CSF markers of iron and neuroinflammation have distinct associations with disease stages, while iron may be more intimately associated with apolipoprotein E and tau pathology.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Abdel A. Belaidi
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Anne Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Blaine Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I. Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | | | | |
Collapse
|
17
|
Li QY, Li XM, Hu HY, Ma YH, Ou YN, Wang AY, Tan L, Yu JT. Associations of Lung Function Decline with Risks of Cognitive Impairment and Dementia: A Meta-Analysis and Systematic Review. J Alzheimers Dis 2023; 92:853-873. [PMID: 36806509 DOI: 10.3233/jad-221136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND There are controversies surrounding the effects of lung function decline on cognitive impairment and dementia. OBJECTIVE We conducted a meta-analysis and systematic review to explore the associations of lung function decline with the risks of cognitive impairment and dementia. METHODS The PubMed, EMBASE, and the Cochrane Library were searched to identify prospective studies published from database inception through January 10, 2023. We pooled relative risk (RR) and 95% confidence intervals (CI) using random-effects models. The Egger test, funnel plots, meta-regression, sensitivity, and subgroup analyses were conducted to detect publication bias and investigate the source of heterogeneity. RESULTS Thirty-three articles with a total of 8,816,992 participants were subjected to meta-analysis. Poorer pulmonary function was associated with an increased risk of dementia (FEV: RR = 1.25 [95% CI, 1.17-1.33]; FVC: RR = 1.40 [95% CI, 1.16-1.69]; PEF: RR = 1.84 [95% CI, 1.37-2.46]). The results of the subgroup analyses were similar to the primary results. Individuals with lung diseases had a higher combined risk of dementia and cognitive impairment (RR = 1.39 [95% CI, 1.20-1.61]). Lung disease conferred an elevated risk of cognitive impairment (RR = 1.37 [95% CI, 1.14-1.65]). The relationship between lung disease and an increased risk of dementia was only shown in total study participants (RR = 1.32 [95% CI, 1.11-1.57]), but not in the participants with Alzheimer's disease (RR = 1.39 [95% CI, 1.00-1.93]) or vascular dementia (RR = 2.11 [95% CI, 0.57-7.83]). CONCLUSION Lung function decline was significantly associated with higher risks of cognitive impairment and dementia. These findings might provide implications for the prevention of cognitive disorders and the promotion of brain health.
Collapse
Affiliation(s)
- Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Mei Li
- Department of Outpatient, Qingdao Municipal Hospital, Qingdao University, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - An-Yi Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Spano M, Roytman M, Aboian M, Saboury B, Franceschi A, Chiang GC. Brain PET Imaging: Approach to Cognitive Impairment and Dementia. PET Clin 2023; 18:103-113. [PMID: 36442959 PMCID: PMC9713600 DOI: 10.1016/j.cpet.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alzheimer disease (AD) is the most common cause of dementia, accounting for 50% to 60% of cases and affecting nearly 6 million people in the United States. Definitive diagnosis requires either antemortem brain biopsy or postmortem autopsy. However, clinical neuroimaging has been playing a greater role in the diagnosis and management of AD, and several PET tracers approach the sensitivity of tissue diagnosis in identifying AD pathologic condition. This review will focus on the utility of PET imaging in the setting of cognitive impairment, with an emphasis on its role in the diagnosis of AD.
Collapse
Affiliation(s)
- Matthew Spano
- Department of Radiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, USA
| | - Michelle Roytman
- Department of Radiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, USA
| | - Mariam Aboian
- Department of Radiology, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, NIH Clinical Center, 10 Center Dr, Bethesda, MD 20892, USA
| | - Ana Franceschi
- Department of Radiology, Northwell Health/Donald and Barbara Zucker School of Medicine, Lenox Hill Hospital, 100 East 77th Street, 3rd Floor, New York, NY 10075, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, USA.
| |
Collapse
|
19
|
Kim J, Kim YK. Molecular Imaging of Neuroinflammation in Alzheimer's Disease and Mild Cognitive Impairment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:301-326. [PMID: 36949316 DOI: 10.1007/978-981-19-7376-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurocognitive disorder. Due to the ineffectiveness of treatments targeting the amyloid cascade, molecular biomarkers for neuroinflammation are attracting attention with increasing knowledge about the role of neuroinflammation in the pathogenesis of AD. This chapter will explore the results of studies using molecular imaging for diagnosing AD and mild cognitive impairment (MCI). Because it is critical to interpreting the data to understand which substances are targeted in molecular imaging, this chapter will discuss the two most significant targets, microglia and astrocytes, as well as the best-known radioligands for each. Then, neuroimaging results with PET neuroinflammation imaging will be reviewed for AD and MCI. Although a growing body of evidence has suggested that these molecular imaging biomarkers for neuroinflammation may have a role in the diagnosis of AD and MCI, the findings are inconsistent or cross-sectional, which indicates that it is difficult to apply the contents in practice due to the need for additional study. In particular, because the results of multiple interventions targeting neuroinflammation were inconclusive, molecular imaging markers for neuroinflammation can be used in combination with conventional markers to select appropriate patients for early intervention for neuroinflammation rather than as a single marker.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Psychiatry, Korea University College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
20
|
Current Pharmacotherapy and Multi-Target Approaches for Alzheimer's Disease. Pharmaceuticals (Basel) 2022; 15:ph15121560. [PMID: 36559010 PMCID: PMC9781592 DOI: 10.3390/ph15121560] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by decreased synaptic transmission and cerebral atrophy with appearance of amyloid plaques and neurofibrillary tangles. Cognitive, functional, and behavioral alterations are commonly associated with the disease. Different pathophysiological pathways of AD have been proposed, some of which interact and influence one another. Current treatment for AD mainly involves the use of therapeutic agents to alleviate the symptoms in AD patients. The conventional single-target treatment approaches do not often cause the desired effect in the disease due to its multifactorial origin. Thus, multi-target strategies have since been undertaken, which aim to simultaneously target multiple targets involved in the development of AD. In this review, we provide an overview of the pathogenesis of AD and the current drug therapies for the disease. Additionally, rationales of the multi-target approaches and examples of multi-target drugs with pharmacological actions against AD are also discussed.
Collapse
|
21
|
18F-Radiolabeled Translocator Protein (TSPO) PET Tracers: Recent Development of TSPO Radioligands and Their Application to PET Study. Pharmaceutics 2022; 14:pharmaceutics14112545. [PMID: 36432736 PMCID: PMC9697781 DOI: 10.3390/pharmaceutics14112545] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a transmembrane protein in the mitochondrial membrane, which has been identified as a peripheral benzodiazepine receptor. TSPO is generally present at high concentrations in steroid-producing cells and plays an important role in steroid synthesis, apoptosis, and cell proliferation. In the central nervous system, TSPO expression is relatively modest under normal physiological circumstances. However, some pathological disorders can lead to changes in TSPO expression. Overexpression of TSPO is associated with several diseases, such as neurodegenerative diseases, neuroinflammation, brain injury, and cancers. TSPO has therefore become an effective biomarker of related diseases. Positron emission tomography (PET), a non-invasive molecular imaging technique used for the clinical diagnosis of numerous diseases, can detect diseases related to TSPO expression. Several radiolabeled TSPO ligands have been developed for PET. In this review, we describe recent advances in the development of TSPO ligands, and 18F-radiolabeled TSPO in particular, as PET tracers. This review covers pharmacokinetic studies, preclinical and clinical trials of 18F-labeled TSPO PET ligands, and the synthesis of TSPO ligands.
Collapse
|
22
|
Huang J. Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Front Immunol 2022; 13:1010946. [PMID: 36211392 PMCID: PMC9537554 DOI: 10.3389/fimmu.2022.1010946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease with a concealed onset and continuous deterioration. Mild cognitive impairment (MCI) is the prodromal stage of AD. Molecule-based imaging with positron emission tomography (PET) is critical in tracking pathophysiological changes among AD and MCI patients. PET with novel targets is a promising approach for diagnostic imaging, particularly in AD patients. Our present review overviews the current status and applications of in vivo molecular imaging toward neuroinflammation. Although radiotracers can remarkably diagnose AD and MCI patients, a variety of limitations prevent the recommendation of a single technique. Recent studies examining neuroinflammation PET imaging suggest an alternative approach to evaluate disease progression. This review concludes that PET imaging towards neuroinflammation is considered a promising approach to deciphering the enigma of the pathophysiological process of AD and MCI.
Collapse
|
23
|
Viviano M, Barresi E, Siméon FG, Costa B, Taliani S, Da Settimo F, Pike VW, Castellano S. Essential Principles and Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging. Curr Med Chem 2022; 29:4862-4890. [PMID: 35352645 PMCID: PMC10080361 DOI: 10.2174/0929867329666220329204054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The translocator protein 18kDa (TSPO) is expressed in the outer mitochondrial membrane and is implicated in several functions, including cholesterol transport and steroidogenesis. Under normal physiological conditions, TSPO is present in very low concentrations in the human brain but is markedly upregulated in response to brain injury and inflammation. This upregulation is strongly associated with activated microglia. Therefore, TSPO is particularly suited for assessing active gliosis associated with brain lesions following injury or disease. For over three decades, TSPO has been studied as a biomarker. Numerous radioligands for positron emission tomography (PET) that target TSPO have been developed for imaging inflammatory progression in the brain. Although [11C]PK11195, the prototypical first-generation PET radioligand, is still widely used for in vivo studies, mainly now as its single more potent R-enantiomer, it has severe limitations, including low sensitivity and poor amenability to quantification. Second-generation radioligands are characterized by higher TSPO specific signals but suffer from other drawbacks, such as sensitivity to the TSPO single nucleotide polymorphism (SNP) rs6971. Therefore, their applications in human studies have the burden of needing to genotype subjects. Consequently, recent efforts are focused on developing improved radioligands that combine the optimal features of the second generation with the ability to overcome the differences in binding affinities across the population. This review presents essential principles in the design and development of TSPO PET ligands and discusses prominent examples among the main chemotypes.
Collapse
Affiliation(s)
- Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | | | - Fabrice G. Siméon
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| |
Collapse
|
24
|
Neuroinflammation in Low-Level PM2.5-Exposed Rats Illustrated by PET via an Improved Automated Produced [18F]FEPPA: A Feasibility Study. Mol Imaging 2022; 2022:1076444. [PMID: 35903248 PMCID: PMC9328187 DOI: 10.1155/2022/1076444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 05/18/2022] [Indexed: 11/26/2022] Open
Abstract
Background [18F]FEPPA is a potent TSPO imaging agent that has been found to be a potential tracer for imaging neuroinflammation. In order to fulfill the demand of this tracer for preclinical and clinical studies, we have developed a one-pot automated synthesis with simplified HPLC purification of this tracer, which was then used for PET imaging of neuroinflammation in fine particulate matter- (PM2.5-) exposed rats. Results Using this automated synthesis method, the RCY of the [18F]FEPPA was 38 ± 4% (n = 17, EOB) in a synthesis time of 83 ± 8 min from EOB. The radiochemical purity and molar activities were greater than 99% and 209 ± 138 GBq/μmol (EOS, n = 15), respectively. The quality of the [18F]FEPPA synthesized by this method met the U.S. Pharmacopoeia (USP) criteria. The stability test showed that the [18F]FEPPA was stable at 21 ± 2°C for up to 4 hr after the end of synthesis (EOS). Moreover, microPET imaging showed that increased tracer activity of [18F]FEPPA in the brain of PM2.5-exposed rats (n = 6) were higher than that of normal controls (n = 6) and regional-specific. Conclusions Using the improved semipreparative HPLC purification, [18F]FEPPA has been produced in high quantity, high quality, and high reproducibility and, for the first time, used for PET imaging the effects of PM2.5 in the rat brain. It is ready to be used for imaging inflammation in various clinical or preclinical studies, especially for nearby PET centers without cyclotrons.
Collapse
|
25
|
Young JW, Barback CV, Stolz LA, Groman SM, Vera DR, Hoh C, Kotta KK, Minassian A, Powell SB, Brody AL. MicroPET evidence for a hypersensitive neuroinflammatory profile of gp120 mouse model of HIV. Psychiatry Res Neuroimaging 2022; 321:111445. [PMID: 35101828 DOI: 10.1016/j.pscychresns.2022.111445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Despite increased survivability for people living with HIV (PLWH), HIV-related cognitive deficits persist. Determining biological mechanism(s) underlying abnormalities is critical to minimize the long-term impact of HIV. Positron emission tomography (PET) studies reveal that PLWH exhibit elevated neuroinflammation, potentially contributing to these problems. PLWH are hypersensitive to environmental insults that drive elevated inflammatory profiles. Gp120 is an envelope glycoprotein exposed on the surface of the HIV envelope which enables HIV entry into a cell contributing to HIV-related neurotoxicity. In vivo evidence for mice overexpressing gp120 (transgenic) mice exhibiting neuroinflammation remains unclear. Here, we conducted microPET imaging in gp120 transgenic and wildtype mice, using the radiotracer [(18)F]FEPPA (binds to the translocator protein expressed by activated microglial serving as a neuroinflammatory marker). Imaging was performed at baseline and 24 h after lipopolysaccharide (LPS; 5 mg/kg) treatment (endotoxin that triggers an immune response). Gp120 transgenic mice exhibited elevated [(18F)]FEPPA in response to LPS vs. wildtype mice throughout the brain including dorsal and ventral striata, hypothalamus, and hippocampus. Gp120 transgenic mice are hypersensitive to environmental inflammatory insults, consistent with PLWH, measurable in vivo. It remains to-be-determined whether this heightened sensitivity is connected to the behavioral abnormalities of these mice or sensitive to any treatments.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Christopher V Barback
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Louise A Stolz
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA
| | - Stephanie M Groman
- Department of Neuroscience, Medical Discovery Team on Addiction, University of Minnesota
| | - David R Vera
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Carl Hoh
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Kishore K Kotta
- Department of Radiology, University of California, San Diego, La Jolla California; UCSD In Vivo Cancer and Molecular Imaging Program
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Arthur L Brody
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
26
|
Comparison of 18F-FDG, 18F-Fluoroacetate, and 18F-FEPPA for Imaging Liver Fibrosis in a Bile Duct-Ligated Rat Model. Mol Imaging 2021; 2021:7545284. [PMID: 34934405 PMCID: PMC8654319 DOI: 10.1155/2021/7545284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/30/2022] Open
Abstract
Developing sensitive diagnostic methods for a longitudinal evaluation of the status of liver fibrosis is a priority. This study is aimed at assessing the significance of longitudinal positron emission tomography (PET) imaging with 18F-labeling tracers for assessing liver fibrosis in a rat model with bile duct ligation (BDL). Twenty-one 6-week-old Sprague-Dawley male rats were used in this study. Longitudinal PET images using [18F]N-2-(2-fluoroethoxy)benzyl)-N-(4-phenoxypyridin-3-yl)acetamide ([18F]FEPPA) (n = 3), [18F]fluoroacetate ([18F]FAc) (n = 3), and 18F-fluoro-2-deoxy-D-glucose ([18F]FDG) (n = 3) were obtained at 0, 1, and 2 weeks after BDL. Biochemical assays, histological assays, immunohistochemical staining assays, and next generation sequencing analyses were also performed at 0 (n = 3), 1 (n = 3), 2 (n = 3), and 3 (n = 3) weeks after BDL, which demonstrated the severe damage in rat livers after BDL. Regarding [18F]FEPPA and [18F]FDG, there was a significantly higher uptake in the liver after BDL (both P < 0.05), which lasted until week 2. However, the uptake of [18F]FAc in the liver was not significantly different before and after BDL (P = 0.28). Collectively, both [18F]FEPPA and [18F]FDG can serve as sensitive probes for detecting the liver fibrosis. However, [18F]FAc is not recommended to diagnose liver fibrosis.
Collapse
|
27
|
Pawlos A, Broncel M, Woźniak E, Gorzelak-Pabiś P. Neuroprotective Effect of SGLT2 Inhibitors. Molecules 2021; 26:7213. [PMID: 34885795 PMCID: PMC8659196 DOI: 10.3390/molecules26237213] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with diabetes are at higher risk of cardiovascular diseases and cognitive impairment. SGLT2 inhibitors (Empagliflozin, Canagliflozin, Dapagliflozin, Ertugliflozin, Sotagliflozin) are newer hypoglycemic agents with many pleiotropic effects. In this review, we discuss their neuroprotective potential. SGLT2 inhibitors (SGLT2i) are lipid-soluble and reach the brain/serum ratio from 0.3 to 0.5. SGLT receptors are present in the central nervous system (CNS). Flozins are not fully SGLT2-selective and have an affinity for the SGLT1 receptor, which is associated with protection against ischemia/reperfusion brain damage. SGLT2i show an anti-inflammatory and anti-atherosclerotic effect, including reduction of proinflammatory cytokines, M2 macrophage polarization, JAK2/STAT1 and NLRP3 inflammasome inhibition, as well as cIMT regression. They also mitigate oxidative stress. SGLT2i improve endothelial function, prevent remodeling and exert a protective effect on the neurovascular unit, blood-brain barrier, pericytes, astrocytes, microglia, and oligodendrocytes. Flozins are also able to inhibit AChE, which contributes to cognitive improvement. Empagliflozin significantly increases the level of cerebral BDNF, which modulates neurotransmission and ensures growth, survival, and plasticity of neurons. Moreover, they may be able to restore the circadian rhythm of mTOR activation, which is quite a novel finding in the field of research on metabolic diseases and cognitive impairment. SGLT2i have a great potential to protect against atherosclerosis and cognitive impairment in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | - Marlena Broncel
- Laboratory of Tissue Immunopharmacology, Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland; (A.P.); (E.W.); (P.G.-P.)
| | | | | |
Collapse
|
28
|
Cisbani G, Rivest S. Targeting innate immunity to protect and cure Alzheimer's disease: opportunities and pitfalls. Mol Psychiatry 2021; 26:5504-5515. [PMID: 33854189 DOI: 10.1038/s41380-021-01083-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/10/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Innate immunity has been the focus of many new directions to understand the mechanisms involved in the aetiology of brain diseases, especially Alzheimer's disease (AD). AD is a multifactorial disorder, with the innate immune response and neuroinflammation at the forefront of the pathology. Thus, microglial cells along with peripheral circulating monocytes and more generally the innate immune response have been the target of several pre-clinical and clinical studies. More than a decade ago, inhibiting innate immune cells was considered to be the critical angle for preventing and treating brain diseases. After the failing of numerous clinical trials and the discovery that it may actually be the opposite in various pre-clinical models, the field has changed considerably. Here, we present both sides of the story with a particular emphasis on the beneficial properties of innate immune cells and how they can be targeted to have neuroprotective properties.
Collapse
Affiliation(s)
- Giulia Cisbani
- Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Serge Rivest
- CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada.
| |
Collapse
|
29
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
30
|
van Aalst J, Ceccarini J, Sunaert S, Dupont P, Koole M, Van Laere K. In vivo synaptic density relates to glucose metabolism at rest in healthy subjects, but is strongly modulated by regional differences. J Cereb Blood Flow Metab 2021; 41:1978-1987. [PMID: 33444094 PMCID: PMC8327121 DOI: 10.1177/0271678x20981502] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Preclinical and postmortem studies have suggested that regional synaptic density and glucose consumption (CMRGlc) are strongly related. However, the relation between synaptic density and cerebral glucose metabolism in the human brain has not directly been assessed in vivo. Using [11C]UCB-J binding to synaptic vesicle glycoprotein 2 A (SV2A) as indicator for synaptic density and [18F]FDG for measuring cerebral glucose consumption, we studied twenty healthy female subjects (age 29.6 ± 9.9 yrs) who underwent a single-day dual-tracer protocol (GE Signa PET-MR). Global measures of absolute and relative CMRGlc and specific binding of [11C]UCB-J were indeed highly significantly correlated (r > 0.47, p < 0.001). However, regional differences in relative [18F]FDG and [11C]UCB-J uptake were observed, with up to 19% higher [11C]UCB-J uptake in the medial temporal lobe (MTL) and up to 17% higher glucose metabolism in frontal and motor-related areas and thalamus. This pattern has a considerable overlap with the brain regions showing different levels of aerobic glycolysis. Regionally varying energy demands of inhibitory and excitatory synapses at rest may also contribute to this difference. Being unaffected by astroglial and/or microglial energy demands, changes in synaptic density in the MTL may therefore be more sensitive to early detection of pathological conditions compared to changes in glucose metabolism.
Collapse
Affiliation(s)
- June van Aalst
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jenny Ceccarini
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Stefan Sunaert
- Translational MRI, Department of Imaging and Pathology, Leuven, Belgium.,Radiology, UZ Leuven, Leuven, Belgium
| | - Patrick Dupont
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Nuclear Medicine, UZ Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Peters van Ton AM, Duindam HB, van Tuijl J, Li WW, Dieker HJ, Riksen NP, Meijer FA, Kessels RP, Kohn N, van der Hoeven JG, Pickkers P, Rijpkema M, Abdo WF. Neuroinflammation in cognitive decline post-cardiac surgery (the FOCUS study): an observational study protocol. BMJ Open 2021; 11:e044062. [PMID: 33980522 PMCID: PMC8118022 DOI: 10.1136/bmjopen-2020-044062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Postoperative cognitive dysfunction occurs frequently after coronary artery bypass grafting (CABG). The underlying mechanisms remain poorly understood, but neuroinflammation might play a pivotal role. We hypothesise that systemic inflammation induced by the surgical trauma could activate the innate immune (glial) cells of the brain. This could lead to an exaggerated neuroinflammatory cascade, resulting in neuronal dysfunction and loss of neuronal cells. Therefore, the aims of this study are to assess neuroinflammation in vivo presurgery and postsurgery in patients undergoing major cardiac surgery and investigate whether there is a relationship of neuroinflammation to cognitive outcomes, changes to brain structure and function, and systemic inflammation. METHODS AND ANALYSIS The FOCUS study is a prospective, single-centre observational study, including 30 patients undergoing elective on-pump CABG. Translocator protein (TSPO) positron emission tomography neuroimaging will be performed preoperatively and postoperatively using the second generation tracer 18F-DPA-714 to assess the neuroinflammatory response. In addition, a comprehensive cerebral MRI will be performed presurgery and postsurgery, in order to discover newly developed brain and vascular wall lesions. Up to 6 months postoperatively, serial extensive neurocognitive assessments will be performed and blood will be obtained to quantify systemic inflammatory responses and peripheral immune cell activation. ETHICS AND DISSEMINATION Patients do not benefit directly from engaging in the study, but imaging neuroinflammation is considered safe and no side effects are expected. The study protocol obtained ethical approval by the Medical Research Ethics Committee region Arnhem-Nijmegen. This work will be published in peer-reviewed international medical journals and presented at medical conferences. TRIAL REGISTRATION NUMBER NCT04520802.
Collapse
Affiliation(s)
- Annemieke M Peters van Ton
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Harmke B Duindam
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Julia van Tuijl
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Wilson Wl Li
- Department of Cardiothoracic Surgery, Radboud university medical center, Nijmegen, The Netherlands
| | - Hendrik-Jan Dieker
- Department of Cardiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Niels P Riksen
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Fj Anton Meijer
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Roy Pc Kessels
- Department of Medical Psychology, Radboud university medical center, Nijmegen, The Netherlands
- Donders Center for Cognition, Radboud University, Nijmegen, The Netherlands
| | - Nils Kohn
- Donders Institute for Brain, Cognition and Behaviour, Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Mark Rijpkema
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Wilson F Abdo
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Bao W, Xie F, Zuo C, Guan Y, Huang YH. PET Neuroimaging of Alzheimer's Disease: Radiotracers and Their Utility in Clinical Research. Front Aging Neurosci 2021; 13:624330. [PMID: 34025386 PMCID: PMC8134674 DOI: 10.3389/fnagi.2021.624330] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD), the leading cause of senile dementia, is a progressive neurodegenerative disorder affecting millions of people worldwide and exerting tremendous socioeconomic burden on all societies. Although definitive diagnosis of AD is often made in the presence of clinical manifestations in late stages, it is now universally believed that AD is a continuum of disease commencing from the preclinical stage with typical neuropathological alterations appearing decades prior to its first symptom, to the prodromal stage with slight symptoms of amnesia (amnestic mild cognitive impairment, aMCI), and then to the terminal stage with extensive loss of basic cognitive functions, i.e., AD-dementia. Positron emission tomography (PET) radiotracers have been developed in a search to meet the increasing clinical need of early detection and treatment monitoring for AD, with reference to the pathophysiological targets in Alzheimer's brain. These include the pathological aggregations of misfolded proteins such as β-amyloid (Aβ) plagues and neurofibrillary tangles (NFTs), impaired neurotransmitter system, neuroinflammation, as well as deficient synaptic vesicles and glucose utilization. In this article we survey the various PET radiotracers available for AD imaging and discuss their clinical applications especially in terms of early detection and cognitive relevance.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
33
|
Huang M, Chen X, Yu Y, Lai H, Feng Q. Imaging Genetics Study Based on a Temporal Group Sparse Regression and Additive Model for Biomarker Detection of Alzheimer's Disease. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:1461-1473. [PMID: 33556003 DOI: 10.1109/tmi.2021.3057660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Imaging genetics is an effective tool used to detect potential biomarkers of Alzheimer's disease (AD) in imaging and genetic data. Most existing imaging genetics methods analyze the association between brain imaging quantitative traits (QTs) and genetic data [e.g., single nucleotide polymorphism (SNP)] by using a linear model, ignoring correlations between a set of QTs and SNP groups, and disregarding the varied associations between longitudinal imaging QTs and SNPs. To solve these problems, we propose a novel temporal group sparsity regression and additive model (T-GSRAM) to identify associations between longitudinal imaging QTs and SNPs for detection of potential AD biomarkers. We first construct a nonparametric regression model to analyze the nonlinear association between QTs and SNPs, which can accurately model the complex influence of SNPs on QTs. We then use longitudinal QTs to identify the trajectory of imaging genetic patterns over time. Moreover, the SNP information of group and individual levels are incorporated into the proposed method to boost the power of biomarker detection. Finally, we propose an efficient algorithm to solve the whole T-GSRAM model. We evaluated our method using simulation data and real data obtained from AD neuroimaging initiative. Experimental results show that our proposed method outperforms several state-of-the-art methods in terms of the receiver operating characteristic curves and area under the curve. Moreover, the detection of AD-related genes and QTs has been confirmed in previous studies, thereby further verifying the effectiveness of our approach and helping understand the genetic basis over time during disease progression.
Collapse
|
34
|
Zhang L, Hu K, Shao T, Hou L, Zhang S, Ye W, Josephson L, Meyer JH, Zhang MR, Vasdev N, Wang J, Xu H, Wang L, Liang SH. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B 2021; 11:373-393. [PMID: 33643818 PMCID: PMC7893127 DOI: 10.1016/j.apsb.2020.08.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is predominately localized to the outer mitochondrial membrane in steroidogenic cells. Brain TSPO expression is relatively low under physiological conditions, but is upregulated in response to glial cell activation. As the primary index of neuroinflammation, TSPO is implicated in the pathogenesis and progression of numerous neuropsychiatric disorders and neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), major depressive disorder (MDD) and obsessive compulsive disorder (OCD). In this context, numerous TSPO-targeted positron emission tomography (PET) tracers have been developed. Among them, several radioligands have advanced to clinical research studies. In this review, we will overview the recent development of TSPO PET tracers, focusing on the radioligand design, radioisotope labeling, pharmacokinetics, and PET imaging evaluation. Additionally, we will consider current limitations, as well as translational potential for future application of TSPO radiopharmaceuticals. This review aims to not only present the challenges in current TSPO PET imaging, but to also provide a new perspective on TSPO targeted PET tracer discovery efforts. Addressing these challenges will facilitate the translation of TSPO in clinical studies of neuroinflammation associated with central nervous system diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
- ANT, adenine nucleotide transporter
- Am, molar activities
- BBB, blood‒brain barrier
- BMSC, bone marrow stromal cells
- BP, binding potential
- BPND, non-displaceable binding potential
- BcTSPO, Bacillus cereus TSPO
- CBD, corticobasal degeneration
- CNS disorders
- CNS, central nervous system
- CRAC, cholesterol recognition amino acid consensus sequence
- DLB, Lewy body dementias
- EP, epilepsy
- FTD, frontotemporal dementia
- HAB, high-affinity binding
- HD, Huntington's disease
- HSE, herpes simplex encephalitis
- IMM, inner mitochondrial membrane
- KA, kainic acid
- LAB, low-affinity binding
- LPS, lipopolysaccharide
- MAB, mixed-affinity binding
- MAO-B, monoamine oxidase B
- MCI, mild cognitive impairment
- MDD, major depressive disorder
- MMSE, mini-mental state examination
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- MSA, multiple system atrophy
- Microglial activation
- NAA/Cr, N-acetylaspartate/creatine
- Neuroinflammation
- OCD, obsessive compulsive disorder
- OMM, outer mitochondrial membrane
- P2X7R, purinergic receptor P2X7
- PAP7, RIa-associated protein
- PBR, peripheral benzodiazepine receptor
- PCA, posterior cortical atrophy
- PD, Parkinson's disease
- PDD, PD dementia
- PET, positron emission tomography
- PKA, protein kinase A
- PRAX-1, PBR-associated protein 1
- PSP, progressive supranuclear palsy
- Positron emission tomography (PET)
- PpIX, protoporphyrin IX
- QA, quinolinic acid
- RCYs, radiochemical yields
- ROS, reactive oxygen species
- RRMS, relapsing remitting multiple sclerosis
- SA, specific activity
- SAH, subarachnoid hemorrhage
- SAR, structure–activity relationship
- SCIDY, spirocyclic iodonium ylide
- SNL, selective neuronal loss
- SNR, signal to noise ratio
- SUV, standard uptake volume
- SUVR, standard uptake volume ratio
- TBAH, tetrabutyl ammonium hydroxide
- TBI, traumatic brain injury
- TLE, temporal lobe epilepsy
- TSPO
- TSPO, translocator protein
- VDAC, voltage-dependent anion channel
- VT, distribution volume
- d.c. RCYs, decay-corrected radiochemical yields
- dMCAO, distal middle cerebral artery occlusion
- fP, plasma free fraction
- n.d.c. RCYs, non-decay-corrected radiochemical yields
- p.i., post-injection
Collapse
|
35
|
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
|
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: 124] [Impact Index Per Article: 24.8] [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
|
Al-Khishman NU, Qi Q, Roseborough AD, Levit A, Allman BL, Anazodo UC, Fox MS, Whitehead SN, Thiessen JD. TSPO PET detects acute neuroinflammation but not diffuse chronically activated MHCII microglia in the rat. EJNMMI Res 2020; 10:113. [PMID: 32990808 PMCID: PMC7524910 DOI: 10.1186/s13550-020-00699-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Background Accurate and sensitive imaging biomarkers are required to study the progression of white matter (WM) inflammation in neurodegenerative diseases. Radioligands targeting the translocator protein (TSPO) are considered sensitive indicators of neuroinflammation, but it is not clear how well the expression of TSPO coincides with major histocompatibility complex class II (MHCII) molecules in WM. This study aimed to test the ability of TSPO to detect activated WM microglia that are immunohistochemically positive for MHCII in rat models of prodromal Alzheimer’s disease and acute subcortical stroke. Methods Fischer 344 wild-type (n = 12) and TgAPP21 (n = 11) rats were imaged with [18F]FEPPA PET and MRI to investigate TSPO tracer uptake in the corpus callosum, a WM region known to have high levels of MHCII activated microglia in TgAPP21 rats. Wild-type rats subsequently received an endothelin-1 (ET1) subcortical stroke and were imaged at days 7 and 28 post-stroke before immunohistochemistry of TSPO, GFAP, iNOS, and the MHCII rat antigen, OX6. Results [18F]FEPPA PET was not significantly affected by genotype in WM and only detected increases near the ET1 infarct (P = 0.033, infarct/cerebellum uptake ratio: baseline = 0.94 ± 0.16; day 7 = 2.10 ± 0.78; day 28 = 1.77 ± 0.35). Immunohistochemistry confirmed that only the infarct (TSPO cells/mm2: day 7 = 555 ± 181; day 28 = 307 ± 153) and WM that is proximal to the infarct had TSPO expression (TSPO cells/mm2: day 7 = 113 ± 93; day 28 = 5 ± 7). TSPO and iNOS were not able to detect the chronic WM microglial activation that was detected with MHCII in the contralateral corpus callosum (day 28 OX6% area: saline = 0.62 ± 0.38; stroke = 4.30 ± 2.83; P = .029). Conclusion TSPO was only expressed in the stroke-induced insult and proximal tissue and therefore was unable to detect remote and non-insult-related chronically activated microglia overexpressing MHCII in WM. This suggests that research in neuroinflammation, particularly in the WM, would benefit from MHCII-sensitive radiotracers.
Collapse
Affiliation(s)
- Nassir U Al-Khishman
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Qi Qi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Austyn D Roseborough
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alexander Levit
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Brian L Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Udunna C Anazodo
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Matthew S Fox
- Department of Physics and Astronomy, Western University, London, ON, Canada.,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jonathan D Thiessen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Lawson Health Research Institute, B5-003a, 268 Grosvenor St, Stn. B, P.O. Box 5777, London, ON, N6A 4V2, Canada.
| |
Collapse
|
38
|
Disrupted white matter functional connectivity in aMCI APOEε4 carriers: a resting-state study. Brain Imaging Behav 2020; 15:1739-1747. [PMID: 32715396 DOI: 10.1007/s11682-020-00367-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ε4 allele of the APOE gene is thought to increase risk from amnestic mild cognitive impairment (aMCI) to Alzheimer's disease. Cognitive decline in the condition is increasingly considered to worsen functional disconnections in brain network composed of gray matter and white matter. Nevertheless, Whether APOEε4 targets specific white matter functional connectivity in patients with aMCI remains mostly unexplored, mainly due to the challenges of detecting BOLD signals in white matter. Here, we applied a novel approach to investigate APOEε4-related specific bundles and cortical area alterations in aMCI subjects, in order to characterize white matter-gray matter functional connectivity differences throughout the brain. We analyzed 75 patients with aMCI and 76 demographically matched normal controls. The aMCI APOEε4 carriers showed decreased functional connectivity located at left corticospinal tract, bilateral posterior limb of internal capsule, and right temporopolaris, which was different from the regions of aMCI-related changes. We further found that recognition scores were positively associated with the right temporopolaris in aMCI APOEε4 carriers. Collectively, the data provide new evidence that APOEε4 genotype exerts a negative impact on neural activity in both gray and white matter in aMCI, which potentially contributes to functional disconnection and memory decline. A novel method provides full-scale measuring effect of disease conditions on functional architecture throughout the brain. Trial registration: https://www.ClinicalTrials.gov (Identifier: NCT02225964). Registered January 2014.
Collapse
|
39
|
Cisbani G, Koppel A, Knezevic D, Suridjan I, Mizrahi R, Bazinet RP. Peripheral cytokine and fatty acid associations with neuroinflammation in AD and aMCI patients: An exploratory study. Brain Behav Immun 2020; 87:679-688. [PMID: 32135194 DOI: 10.1016/j.bbi.2020.02.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/10/2020] [Accepted: 02/26/2020] [Indexed: 01/01/2023] Open
Abstract
Neuroinflammation is thought to be important in the progression of Alzheimer's disease (AD). To evaluate cerebral inflammation radioligands that target TSPO, a translocator protein strongly expressed in microglia and macrophages during inflammation, can be used in conjunction with positron emission tomography (PET) imaging. In AD patients, neuroinflammation is up-regulated compared to both healthy volunteers as well as to subjects with amnestic Mild Cognitive Impairment. Peripheral biomarkers, such as serum cytokines and total fatty acids (FAs), can also be indicative of the inflammatory state of subjects with neurodegenerative disorders. To understand whether peripheral biomarkers are predictive of neuroinflammation we conducted a secondary exploratory analysis of two TSPO imaging studies conducted in subjects with AD, aMCI and aged matched healthy volunteers. We examined the association between candidate peripheral biomarkers (including amyloid beta, cytokines and serum total fatty acids) with brain TSPO levels. Our results showed that serum IL-6 and IL-10 are higher in AD compared to the aMCI and healthy volunteers while levels of some fatty acids are modulated during the disease. A limited number of associations were observed between region-specific inflammation and fatty acids in aMCI patients, and between amyloid beta 42 and brain inflammation in AD, however no associations were present with systemic cytokines. Our study suggests that while TSPO binding and systemic IL-6 and IL-10 were elevated in AD, serum amyloid beta, cytokines and fatty acids were generally not predictive of the disease nor correlated with neuroinflammation.
Collapse
Affiliation(s)
- Giulia Cisbani
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Canada
| | - Alex Koppel
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Dunja Knezevic
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Ivonne Suridjan
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Romina Mizrahi
- Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Canada
| |
Collapse
|
40
|
Berdyyeva T, Xia C, Taylor N, He Y, Chen G, Huang C, Zhang W, Kolb H, Letavic M, Bhattacharya A, Szardenings AK. PET Imaging of the P2X7 Ion Channel with a Novel Tracer [ 18F]JNJ-64413739 in a Rat Model of Neuroinflammation. Mol Imaging Biol 2020; 21:871-878. [PMID: 30632003 DOI: 10.1007/s11307-018-01313-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The P2X7 receptor, an adenosine triphosphate (ATP)-gated purinoreceptor, has emerged as one of the key players in neuroinflammatory processes. Therefore, developing a positron emission tomography (PET) tracer for imaging of P2X7 receptors in vivo presents a promising approach to diagnose, monitor, and study neuroinflammation in a variety of brain disorders. To fulfill the goal of developing a P2X7 PET ligand as a biomarker of neuroinflammation, [18F]JNJ-64413739 has been recently disclosed. PROCEDURES We evaluated [18F]JNJ-64413739 in a rat model of neuroinflammation induced by an intracerebral injection of lipopolysaccharide (LPS). In vivo brain uptake was determined by PET imaging. Upregulation of neuroinflammatory biomarkers was determined by quantitative polymerase chain reaction (qPCR). Distribution of the tracer in the brain was determined by ex vivo autoradiography (ARG). The specificity of [18F]JNJ-64413739 was confirmed by performing blocking experiments with the P2X7 antagonist JNJ-54175446. RESULTS Brain regions of rats injected with LPS had a significantly increased uptake (34 % ± 3 % s.e.m., p = 0.036, t test, standardized uptake value measured over the entire scanning period) of [18F]JNJ-64413739 relative to the corresponding brain regions of control animals injected with phosphate-buffered saline (PBS). The uptake in the contralateral regions and cerebellum was not significantly different between the groups of animals. The increase in uptake of [18F]JNJ-64413739 at the LPS-injected site observed by PET imaging was concordant with ex vivo ARG, upregulation of neuroinflammatory biomarkers, and elevated P2X7 expression levels. CONCLUSIONS While further work is needed to study [18F]JNJ-64413739 in other types of neuroinflammation, the current results favorably characterize [18F]JNJ-64413739 as a potential PET tracer of central neuroinflammation.
Collapse
Affiliation(s)
- Tamara Berdyyeva
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| | - Chunfang Xia
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Natalie Taylor
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Yingbo He
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Gang Chen
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Chaofeng Huang
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Wei Zhang
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Hartmuth Kolb
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Michael Letavic
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Anindya Bhattacharya
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | | |
Collapse
|
41
|
Benek O, Korabecny J, Soukup O. A Perspective on Multi-target Drugs for Alzheimer's Disease. Trends Pharmacol Sci 2020; 41:434-445. [PMID: 32448557 DOI: 10.1016/j.tips.2020.04.008] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) has a complex pathophysiology that includes aggregation of pathological proteins, impaired neurotransmission, increased oxidative stress, or microglia-mediated neuroinflammation. Therapeutics targeting only one of these AD-related subpathologies have not yet been successful in the search for a disease-modifying treatment. Therefore, multi-target drugs (MTDs) aiming simultaneously at several subpathologies are expected to be a better approach. However, the concept of MTD is inherently connected with several limitations, which are often ignored during MTD design and development. Here, we provide an overview of the MTD approach and discuss its potential pitfalls in the context of AD treatment. We also put forward ideas to be used in the rational design of MTDs to obtain drugs that are effective against AD.
Collapse
Affiliation(s)
- Ondrej Benek
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Jan Korabecny
- University Hospital Hradec Kralove, Biomedical Research Centre, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.
| | - Ondrej Soukup
- University Hospital Hradec Kralove, Biomedical Research Centre, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.
| |
Collapse
|
42
|
Tong J, Williams B, Rusjan PM, Mizrahi R, Lacapère JJ, McCluskey T, Furukawa Y, Guttman M, Ang LC, Boileau I, Meyer JH, Kish SJ. Concentration, distribution, and influence of aging on the 18 kDa translocator protein in human brain: Implications for brain imaging studies. J Cereb Blood Flow Metab 2020; 40:1061-1076. [PMID: 31220997 PMCID: PMC7181090 DOI: 10.1177/0271678x19858003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Positron emission tomography (PET) imaging of the translocator protein (TSPO) is widely used as a biomarker of microglial activation. However, TSPO protein concentration in human brain has not been optimally quantified nor has its regional distribution been compared to TSPO binding. We determined TSPO protein concentration, change with age, and regional distribution by quantitative immunoblotting in autopsied human brain. Brain TSPO protein concentration (>0.1 ng/µg protein) was higher than those reported by in vitro binding assays by at least 2 to 70 fold. TSPO protein distributed widely in both gray and white matter regions, with distribution in major gray matter areas ranked generally similar to that of PET binding in second-generation radiotracer studies. TSPO protein concentration in frontal cortex was high at birth, declined precipitously during the first three months, and increased modestly during adulthood/senescence (10%/decade; vs. 30% for comparison astrocytic marker GFAP). As expected, TSPO protein levels were significantly increased (+114%) in degenerating putamen in multiple system atrophy, providing further circumstantial support for TSPO as a gliosis marker. Overall, findings show some similarities between TSPO protein and PET binding characteristics in the human brain but also suggest that part of the TSPO protein pool might be less available for radioligand binding.
Collapse
Affiliation(s)
- Junchao Tong
- Preclinical Imaging, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Junchao Tong, Preclinical Imaging, Centre
for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8,
Canada.
| | - Belinda Williams
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Pablo M. Rusjan
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Romina Mizrahi
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Jean-Jacques Lacapère
- Sorbonne Universités-UPMC University of
Paris 06, Département de Chimie, École Normale Supérieure-PSL Research University,
Paris, France
| | - Tina McCluskey
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Yoshiaki Furukawa
- Department of Neurology, Juntendo Tokyo
Koto Geriatric Medical Center, and Faculty of Medicine, University & Post
Graduate University of Juntendo, Tokyo, Japan
| | - Mark Guttman
- Centre for Movement Disorders, Toronto,
Ontario, Canada
| | - Lee-Cyn Ang
- Division of Neuropathology, London
Health Science Centre, University of Western Ontario, London, Ontario, Canada
| | - Isabelle Boileau
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Jeffrey H Meyer
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Stephen J Kish
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| |
Collapse
|
43
|
Vo SV, Banister SD, Freelander I, Werry EL, Reekie TA, Ittner LM, Kassiou M. Reversing binding sensitivity to A147T translocator protein. RSC Med Chem 2020; 11:511-517. [PMID: 33479652 PMCID: PMC7489257 DOI: 10.1039/c9md00580c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/22/2020] [Indexed: 11/21/2022] Open
Abstract
The translocator protein (TSPO) is a target for the development of neuroinflammation imaging agents. Clinical translation of TSPO PET ligands, such as [11C]DPA-713, has been hampered by the presence of a common polymorphism (A147T TSPO), at which all second-generation TSPO ligands lose affinity. Little is known about what drives binding at A147T compared to WT TSPO. This study aimed to identify moieties in DPA-713, and related derivatives, that influence binding at A147T compared to WT TSPO. We found changes to the nitrogen position and number in the heterocyclic core influences affinity to WT and A147T to a similar degree. Hydrogen bonding groups in molecules with an indole core improve binding at A147T compared to WT, a strategy that generated compounds that possess up to ten-times greater affinity for A147T. These results should inform the future design of compounds that bind both A147T and WT TSPO for use in neuroinflammation imaging.
Collapse
Affiliation(s)
- Sophie V Vo
- Faculty of Medicine and Health , The University of Sydney , Sydney , New South Wales 2006 , Australia
| | - Samuel D Banister
- The Lambert Initiative for Cannabinoid Therapeutics , Brain and Mind Centre , The University of Sydney , Camperdown , NSW 2050 , Australia
- School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia .
| | - Isaac Freelander
- School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia .
| | - Eryn L Werry
- School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia .
| | - Tristan A Reekie
- Research School of Chemistry , The Australian National University , Canberra , Australian Capital Territory 2600 , Australia
| | - Lars M Ittner
- Department of Biomedical Sciences , Faculty of Medicine and Health Sciences , Macquarie University , 2 Technology Place , North Ryde , New South Wales 2109 , Australia
| | - Michael Kassiou
- School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia .
| |
Collapse
|
44
|
Chandra A, Valkimadi PE, Pagano G, Cousins O, Dervenoulas G, Politis M. Applications of amyloid, tau, and neuroinflammation PET imaging to Alzheimer's disease and mild cognitive impairment. Hum Brain Mapp 2019; 40:5424-5442. [PMID: 31520513 PMCID: PMC6864887 DOI: 10.1002/hbm.24782] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/29/2019] [Accepted: 08/18/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and progressive neurodegenerative disease for which there is no cure. Mild cognitive impairment (MCI) is considered a prodromal stage of the disease. Molecular imaging with positron emission tomography (PET) allows for the in vivo visualisation and tracking of pathophysiological changes in AD and MCI. PET is a very promising methodology for differential diagnosis and novel targets of PET imaging might also serve as biomarkers for disease-modifying therapeutic interventions. This review provides an overview of the current status and applications of in vivo molecular imaging of AD pathology, specifically amyloid, tau, and microglial activation. PET imaging studies were included and evaluated as potential biomarkers and for monitoring disease progression. Although the majority of radiotracers showed the ability to discriminate AD and MCI patients from healthy controls, they had various limitations that prevent the recommendation of a single technique or tracer as an optimal biomarker. Newer research examining amyloid, tau, and microglial PET imaging in combination suggest an alternative approach in studying the disease process.
Collapse
Affiliation(s)
- Avinash Chandra
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Polytimi-Eleni Valkimadi
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Oliver Cousins
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - George Dervenoulas
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| |
Collapse
|
45
|
Beaurain M, Salabert AS, Ribeiro MJ, Arlicot N, Damier P, Le Jeune F, Demonet JF, Payoux P. Innovative Molecular Imaging for Clinical Research, Therapeutic Stratification, and Nosography in Neuroscience. Front Med (Lausanne) 2019; 6:268. [PMID: 31828073 PMCID: PMC6890558 DOI: 10.3389/fmed.2019.00268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/01/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past few decades, several radiotracers have been developed for neuroimaging applications, especially in PET. Because of their low steric hindrance, PET radionuclides can be used to label molecules that are small enough to cross the blood brain barrier, without modifying their biological properties. As the use of 11C is limited by its short physical half-life (20 min), there has been an increasing focus on developing tracers labeled with 18F for clinical use. The first such tracers allowed cerebral blood flow and glucose metabolism to be measured, and the development of molecular imaging has since enabled to focus more closely on specific targets such as receptors, neurotransmitter transporters, and other proteins. Hence, PET and SPECT biomarkers have become indispensable for innovative clinical research. Currently, the treatment options for a number of pathologies, notably neurodegenerative diseases, remain only supportive and symptomatic. Treatments that slow down or reverse disease progression are therefore the subject of numerous studies, in which molecular imaging is proving to be a powerful tool. PET and SPECT biomarkers already make it possible to diagnose several neurological diseases in vivo and at preclinical stages, yielding topographic, and quantitative data about the target. As a result, they can be used for assessing patients' eligibility for new treatments, or for treatment follow-up. The aim of the present review was to map major innovative radiotracers used in neuroscience, and explain their contribution to clinical research. We categorized them according to their target: dopaminergic, cholinergic or serotoninergic systems, β-amyloid plaques, tau protein, neuroinflammation, glutamate or GABA receptors, or α-synuclein. Most neurological disorders, and indeed mental disorders, involve the dysfunction of one or more of these targets. Combinations of molecular imaging biomarkers can afford us a better understanding of the mechanisms underlying disease development over time, and contribute to early detection/screening, diagnosis, therapy delivery/monitoring, and treatment follow-up in both research and clinical settings.
Collapse
Affiliation(s)
- Marie Beaurain
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Anne-Sophie Salabert
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Maria Joao Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Philippe Damier
- Inserm U913, Neurology Department, University Hospital, Nantes, France
| | | | - Jean-François Demonet
- Leenards Memory Centre, Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Payoux
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| |
Collapse
|
46
|
Al Rihani SB, Darakjian LI, Kaddoumi A. Oleocanthal-Rich Extra-Virgin Olive Oil Restores the Blood-Brain Barrier Function through NLRP3 Inflammasome Inhibition Simultaneously with Autophagy Induction in TgSwDI Mice. ACS Chem Neurosci 2019; 10:3543-3554. [PMID: 31244050 DOI: 10.1021/acschemneuro.9b00175] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by multiple hallmarks including extracellular amyloid (Aβ) plaques, neurofibrillary tangles, dysfunctional blood-brain barrier (BBB), neuroinflammation, and impaired autophagy. Thus, novel strategies that target multiple disease pathways would be essential to prevent, halt, or treat the disease. A growing body of evidence including our studies supports a protective effect of oleocanthal (OC) and extra-virgin olive oil (EVOO) at early AD stages before the onset of pathology. In addition, we reported previously that OC and EVOO exhibited such effect by restoring the BBB function; however, the mechanism(s) by which OC and EVOO exert such an effect and whether this effect extends to a later stage of AD remain unknown. In this work, we sought first to test the effect of OC-rich EVOO consumption at an advanced stage of the disease in TgSwDI mice, an AD mouse model, starting at the age of 6 months for 3 months treatment, and then to elucidate the mechanism(s) by which OC-rich EVOO exerts the observed beneficial effect. Overall findings demonstrated that OC-rich EVOO restored the BBB function and reduced AD-associated pathology by reducing neuroinflammation through inhibition of NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome and inducing autophagy through activation of AMP-activated protein kinase (AMPK)/Unc-51-like autophagy activating kinase 1 (ULK1) pathway. Thus, diet supplementation with OC-rich EVOO could provide beneficial effect to slow or halt the progression of AD.
Collapse
Affiliation(s)
- Sweilem B. Al Rihani
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
- Center for Neuroscience Initiative, Auburn University, Auburn, Alabama 36849, United States
| | - Lucy I. Darakjian
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
- Center for Neuroscience Initiative, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
47
|
The Interaction Between Neuroinflammation and β-Amyloid in Cognitive Decline in Parkinson's Disease. Mol Neurobiol 2019; 57:492-501. [PMID: 31385228 DOI: 10.1007/s12035-019-01714-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
Activated microglia have been reported to play an important role in Parkinson's disease (PD). A more rapid cognitive decline has been associated with deposits of β-amyloid. In this study, the aim was to evaluate the role of brain β-amyloid and its relationship with activated microglia in PD patients with normal and impaired cognition. We studied 17 PD patients with normal cognition (PDn), 12 PD patients with mild cognitive impairment (PD-MCI), and 12 healthy controls (HCs) with [11C] Pittsburgh compound B (PIB) to assess the impact of β-amyloid deposition in the brain on microglial activation evaluated using the translocator protein 18-kDa (TSPO) radioligand [18F]-FEPPA. [11C] PIB distribution volume ratio was measured in cortical and subcortical regions. [18F]-FEPPA total distribution volume values were compared for each brain region between groups to evaluate the effect of PIB positivity while adjusting for the TSPO rs6971 polymorphism. Factorial analysis of variance revealed a significant main effect of PIB positivity in the frontal lobe (F(1, 34) = 7.1, p = 0.012). Besides the frontal (p = 0.006) and temporal lobe (p = 0.001), the striatum (p = 0.018), the precuneus (p = 0.019), and the dorsolateral prefrontal cortex (p = 0.010) showed significant group × PIB positivity interaction effects. In these regions, PD-MCIs had significantly higher FEPPA VT if PIB-positive. Our results indicate an interaction between amyloid-β deposition and microglial activation in PD. Further investigations are necessary to evaluate if amyloid deposits cause neuroinflammation and further neurodegeneration or if increased microglia activation develops as a protective response.
Collapse
|
48
|
Dahoun T, Calcia MA, Veronese M, Bloomfield P, Reis Marques T, Turkheimer F, Howes OD. The association of psychosocial risk factors for mental health with a brain marker altered by inflammation: A translocator protein (TSPO) PET imaging study. Brain Behav Immun 2019; 80:742-750. [PMID: 31112791 DOI: 10.1016/j.bbi.2019.05.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022] Open
Abstract
Psychiatric disorders associated with psychosocial risk factors, including depression and psychosis, have been shown to demonstrate increased microglia activity. Whilst preclinical studies indicate that psychosocial stress leads to increased levels of microglia in the frontal cortex, no study has yet been performed in humans. This study aimed at investigating whether psychosocial risk factors for depression and/or psychosis would be associated with alterations in a brain marker expressed by microglia, the translocator specific protein (TSPO) in humans. We used [11C]-PBR28 Positron Emission Tomography on healthy subjects exposed to childhood and adulthood psychosocial risk factors (high-risk group, N = 12) and age- and sex-matched healthy controls not exposed to childhood and adulthood psychosocial risk factors (low-risk group, N = 12). The [11C]-PBR28 volume of distribution (VT) and Distribution Volume Ratio (DVR) were measured in the total gray matter, and frontal, parietal, temporal, occipital lobes. Levels of childhood trauma, anxiety and depression were measured using respectively the Childhood Trauma Questionnaire, State-anxiety questionnaire and Beck Depression Inventory. Compared to the low-risk group, the high-risk group did not exhibit significant differences in the mean [11C]-PBR28 VT (F(1,20) = 1.619, p = 0.218) or DVR (F(1,22) = 0.952, p = 0.340) on any region. There were no significant correlations between the [11C]-PBR28 VT and DVRs in total gray matter and frontal lobe and measures of childhood trauma, anxiety and depression. Psychosocial risk factors for depression and/or psychosis are unlikely to be associated with alterations in [11C]-PBR28 binding, indicating that alterations in TSPO expression reported in these disorders is unlikely to be caused by psychosocial risk factors alone.
Collapse
Affiliation(s)
- Tarik Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX37 JX, UK
| | - Marilia A Calcia
- Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Peter Bloomfield
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK
| | - Tiago Reis Marques
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Oliver D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK.
| |
Collapse
|
49
|
Recent Developments in TSPO PET Imaging as A Biomarker of Neuroinflammation in Neurodegenerative Disorders. Int J Mol Sci 2019; 20:ijms20133161. [PMID: 31261683 PMCID: PMC6650818 DOI: 10.3390/ijms20133161] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is an inflammatory response in the brain and spinal cord, which can involve the activation of microglia and astrocytes. It is a common feature of many central nervous system disorders, including a range of neurodegenerative disorders. An overlap between activated microglia, pro-inflammatory cytokines and translocator protein (TSPO) ligand binding was shown in early animal studies of neurodegeneration. These findings have been translated in clinical studies, where increases in TSPO positron emission tomography (PET) signal occur in disease-relevant areas across a broad spectrum of neurodegenerative diseases. While this supports the use of TSPO PET as a biomarker to monitor response in clinical trials of novel neurodegenerative therapeutics, the clinical utility of current TSPO PET radioligands has been hampered by the lack of high affinity binding to a prevalent form of polymorphic TSPO (A147T) compared to wild type TSPO. This review details recent developments in exploration of ligand-sensitivity to A147T TSPO that have yielded ligands with improved clinical utility. In addition to developing a non-discriminating TSPO ligand, the final frontier of TSPO biomarker research requires developing an understanding of the cellular and functional interpretation of the TSPO PET signal. Recent insights resulting from single cell analysis of microglial phenotypes are reviewed.
Collapse
|
50
|
Wang Q, Wang Y, Liu J, Sutphen CL, Cruchaga C, Blazey T, Gordon BA, Su Y, Chen C, Shimony JS, Ances BM, Cairns NJ, Fagan AM, Morris JC, Benzinger TLS. Quantification of white matter cellularity and damage in preclinical and early symptomatic Alzheimer's disease. Neuroimage Clin 2019; 22:101767. [PMID: 30901713 PMCID: PMC6428957 DOI: 10.1016/j.nicl.2019.101767] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/12/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023]
Abstract
Interest in understanding the roles of white matter (WM) inflammation and damage in the pathophysiology of Alzheimer disease (AD) has been growing significantly in recent years. However, in vivo magnetic resonance imaging (MRI) techniques for imaging inflammation are still lacking. An advanced diffusion-based MRI method, neuro-inflammation imaging (NII), has been developed to clinically image and quantify WM inflammation and damage in AD. Here, we employed NII measures in conjunction with cerebrospinal fluid (CSF) biomarker classification (for β-amyloid (Aβ) and neurodegeneration) to evaluate 200 participants in an ongoing study of memory and aging. Elevated NII-derived cellular diffusivity was observed in both preclinical and early symptomatic phases of AD, while disruption of WM integrity, as detected by decreased fractional anisotropy (FA) and increased radial diffusivity (RD), was only observed in the symptomatic phase of AD. This may suggest that WM inflammation occurs earlier than WM damage following abnormal Aβ accumulation in AD. The negative correlation between NII-derived cellular diffusivity and CSF Aβ42 level (a marker of amyloidosis) may indicate that WM inflammation is associated with increasing Aβ burden. NII-derived FA also negatively correlated with CSF t-tau level (a marker of neurodegeneration), suggesting that disruption of WM integrity is associated with increasing neurodegeneration. Our findings demonstrated the capability of NII to simultaneously image and quantify WM cellularity changes and damage in preclinical and early symptomatic AD. NII may serve as a clinically feasible imaging tool to study the individual and composite roles of WM inflammation and damage in AD.
Collapse
Affiliation(s)
- Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA
| | - Yong Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Engineering & Applied Science, St. Louis, MO 63015, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Jingxia Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Courtney L Sutphen
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tyler Blazey
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| | - Charlie Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M Ances
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M Fagan
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|