1
|
Quintanilla-Bordás C, Fernández-Patón M, Ten A, Ferrer-Pardo C, Carratala-Bosca S, Castillo-Villalba J, Cubas-Núñez L, Gasqué-Rubio R, Verdini-Martínez L, Pérez-Miralles F, Martí-Bonmatí L, Casanova B. Dynamic 18 F-FDG PET to detect differences among patients with progressive and relapsing multiple sclerosis: a pilot study. Neurol Sci 2024:10.1007/s10072-024-07921-x. [PMID: 39692831 DOI: 10.1007/s10072-024-07921-x] [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: 01/24/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Patients with multiple sclerosis (MS) may remain in a relapsing-remitting (RRMS) course despite long-standing disease, while others will develop secondary progression (SPMS). Chronic inflammation and changes in the blood-brain barrier resulting in perturbed glucose metabolism may account for these differences. PET-MRI with kinetic analysis of 2-deoxy-2(18 F)fluoro-d-glucose (18 F-FDG) provides insight into glucose metabolism and has proven useful in several chronic inflammatory diseases. However, to our knowledge, it has never been studied in MS. OBJECTIVE To explore potential differences in glucose distribution kinetics among individuals with long-standing SPMS and RRMS using dynamic 18-F-FDG PET-MRI. METHODS Dynamic 18-F-FDG PET-MRI scans were obtained in 11 patients with long-standing MS: 4 with RRMS and 7 with SPMS. Kinetic analysis of PET data was performed using a three-compartment model equation that represents plasma, tissue and 18 F-FDG phosphorylation. Individual rate constants of 18-F-FDG across the compartments were calculated. RESULTS Patients with SPMS exhibited a trend towards an increased net influx rate of glucose (p = 0.059) and an increased rate constant representing glucose phosphorylation. Together, the data suggest increased uptake of glucose and glycolysis in these patients. CONCLUSION Dynamic 18 F-FDG PET-MRI is a feasible technique that may show information in vivo of glucose metabolism in MS. Although preliminary data suggest a potential radiological marker of progression in MS, further studies are required to confirm this hypothesis.
Collapse
Affiliation(s)
- Carlos Quintanilla-Bordás
- Neuroimmunology Unit, La Fe University and Polytechnic Hospital, Avda. Fernando Abril Martorell, 106, Valencia, 46026, Spain.
| | - Matías Fernández-Patón
- Grupo de Investigación Biomédica de Imagen, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Amadeo Ten
- Grupo de Investigación Biomédica de Imagen, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Cristina Ferrer-Pardo
- Neuroimmunology Unit, La Fe University and Polytechnic Hospital, Avda. Fernando Abril Martorell, 106, Valencia, 46026, Spain
| | | | | | - Laura Cubas-Núñez
- Neuroimmunology Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Raquel Gasqué-Rubio
- Neuroimmunology Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | | | - Francisco Pérez-Miralles
- Neuroimmunology Unit, La Fe University and Polytechnic Hospital, Avda. Fernando Abril Martorell, 106, Valencia, 46026, Spain
| | - Luís Martí-Bonmatí
- Medical Imaging department, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Bonaventura Casanova
- Neuroimmunology Unit, La Fe University and Polytechnic Hospital, Avda. Fernando Abril Martorell, 106, Valencia, 46026, Spain
| |
Collapse
|
2
|
Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, Golabi B, Aletaha R, Motlagh Asghari K, Hamidi S, Mousavi SE, Jamalkhani S, Karamzad N, Shamekh A, Mohammadinasab R, Sullman MJM, Şahin F, Kolahi AA. Alzheimer's disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024; 11:1474043. [PMID: 39736972 PMCID: PMC11682909 DOI: 10.3389/fmed.2024.1474043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Background Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired reasoning. It is the leading cause of dementia in older adults, marked by the pathological accumulation of amyloid-beta plaques and neurofibrillary tangles. These pathological changes lead to widespread neuronal damage, significantly impacting daily functioning and quality of life. Objective This comprehensive review aims to explore various aspects of Alzheimer's disease, including its epidemiology, risk factors, clinical presentation, diagnostic advancements, management strategies, caregiving challenges, and emerging therapeutic interventions. Methods A systematic literature review was conducted across multiple electronic databases, including PubMed, MEDLINE, Cochrane Library, and Scopus, from their inception to May 2024. The search strategy incorporated a combination of keywords and Medical Subject Headings (MeSH) terms such as "Alzheimer's disease," "epidemiology," "risk factors," "symptoms," "diagnosis," "management," "caregiving," "treatment," and "novel therapies." Boolean operators (AND, OR) were used to refine the search, ensuring a comprehensive analysis of the existing literature on Alzheimer's disease. Results AD is significantly influenced by genetic predispositions, such as the apolipoprotein E (APOE) ε4 allele, along with modifiable environmental factors like diet, physical activity, and cognitive engagement. Diagnostic approaches have evolved with advances in neuroimaging techniques (MRI, PET), and biomarker analysis, allowing for earlier detection and intervention. The National Institute on Aging and the Alzheimer's Association have updated diagnostic criteria to include biomarker data, enhancing early diagnosis. Conclusion The management of AD includes pharmacological treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, which provide symptomatic relief but do not slow disease progression. Emerging therapies, including amyloid-beta and tau-targeting treatments, gene therapy, and immunotherapy, offer potential for disease modification. The critical role of caregivers is underscored, as they face considerable emotional, physical, and financial burdens. Support programs, communication strategies, and educational interventions are essential for improving caregiving outcomes. While significant advancements have been made in understanding and managing AD, ongoing research is necessary to identify new therapeutic targets and enhance diagnostic and treatment strategies. A holistic approach, integrating clinical, genetic, and environmental factors, is essential for addressing the multifaceted challenges of Alzheimer's disease and improving outcomes for both patients and caregivers.
Collapse
Affiliation(s)
- Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Morsali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Golabi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aletaha
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kimia Motlagh Asghari
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Hamidi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Jamalkhani
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional, Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J. M. Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Dinkel L, Hummel S, Zenatti V, Malara M, Tillmann Y, Colombo A, Monasor LS, Suh JH, Logan T, Roth S, Paeger L, Hoffelner P, Bludau O, Schmidt A, Müller SA, Schifferer M, Nuscher B, Njavro JR, Prestel M, Bartos LM, Wind-Mark K, Slemann L, Hoermann L, Kunte ST, Gnörich J, Lindner S, Simons M, Herms J, Paquet D, Lichtenthaler SF, Bartenstein P, Franzmeier N, Liesz A, Grosche A, Bremova-Ertl T, Catarino C, Beblo S, Bergner C, Schneider SA, Strupp M, Di Paolo G, Brendel M, Tahirovic S. Myeloid cell-specific loss of NPC1 in mice recapitulates microgliosis and neurodegeneration in patients with Niemann-Pick type C disease. Sci Transl Med 2024; 16:eadl4616. [PMID: 39630885 DOI: 10.1126/scitranslmed.adl4616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 07/12/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Niemann-Pick type C (NPC) disease is an inherited lysosomal storage disorder mainly driven by mutations in the NPC1 gene, causing lipid accumulation within late endosomes/lysosomes and resulting in progressive neurodegeneration. Although microglial activation precedes neuronal loss, it remains elusive whether loss of the membrane protein NPC1 in microglia actively contributes to NPC pathology. In a mouse model with depletion of NPC1 in myeloid cells, we report severe alterations in microglial lipidomic profiles, including the enrichment of bis(monoacylglycero)phosphate, increased cholesterol, and a decrease in cholesteryl esters. Lipid dyshomeostasis was associated with microglial hyperactivity, marked by an increase in translocator protein 18 kDa (TSPO). These hyperactive microglia initiated a pathological cascade resembling NPC-like phenotypes, including a shortened life span, motor impairments, astrogliosis, neuroaxonal pathology, and increased neurofilament light chain (NF-L), a neuronal injury biomarker. As observed in the mouse model, patients with NPC showed increased NF-L in the blood and microglial hyperactivity, as visualized by TSPO-PET imaging. Reduced TSPO expression in blood-derived macrophages of patients with NPC was measured after N-acetyl-l-leucine treatment, which has been recently shown to have beneficial effects in patients with NPC, suggesting that TSPO is a potential marker to monitor therapeutic interventions for NPC. Conclusively, these results demonstrate that myeloid dysfunction, driven by the loss of NPC1, contributes to NPC disease and should be further investigated for therapeutic targeting and disease monitoring.
Collapse
Affiliation(s)
- Lina Dinkel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Selina Hummel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Valerio Zenatti
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Mariagiovanna Malara
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Yannik Tillmann
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Alessio Colombo
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | | | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Todd Logan
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Patricia Hoffelner
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University, 82152 Planegg-Martinsried, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University, 82152 Planegg-Martinsried, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Brigitte Nuscher
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Jasenka Rudan Njavro
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Matthias Prestel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Karin Wind-Mark
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Luna Slemann
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sebastian T Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Institute of Neuronal Cell Biology (TUM-NZB), Technical University of Munich, 80802 Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Neuroproteomics School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Sahlgrenska Academy, Institute of Neuroscience and Physiology, SE-413 90 Mölndal and Gothenburg, Sweden
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Tatiana Bremova-Ertl
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Neurology, University Hospital Bern, 3010 Bern, Switzerland
| | - Claudia Catarino
- Friedrich Baur Institute, Department of Neurology, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Skadi Beblo
- Center for Pediatric Research Leipzig, Department of Women and Child Health, Hospital for Children and Adolescents, University Hospital Leipzig; Leipzig University Center for Rare Diseases, 04103 Leipzig, Germany
| | - Caroline Bergner
- Department of Neurology, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Susanne A Schneider
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Michael Strupp
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | | | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| |
Collapse
|
4
|
Li H, Fang Y, Wang D, Shi B, Thompson GJ. Impaired brain glucose metabolism in glucagon-like peptide-1 receptor knockout mice. Nutr Diabetes 2024; 14:86. [PMID: 39389952 PMCID: PMC11466955 DOI: 10.1038/s41387-024-00343-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Quantitative mapping of the brain's metabolism is a critical tool in studying and diagnosing many conditions, from obesity to neurodegenerative diseases. In particular, noninvasive approaches are urgently required. Recently, there have been promising drug development approaches for the treatment of disorders related to glucose metabolism in the brain and, therefore, against obesity-associated diseases. One of the most important drug targets to emerge has been the Glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R). GLP and GLP-1R play an important role in regulating blood sugar and maintaining energy homeostasis. However, the macroscopic effects on brain metabolism and function due to the presence of GLP-1R are unclear. METHODS To explore the physiological role of GLP-1R in mouse brain glucose metabolism, and its relationship to brain function, we used three methods. We used deuterium magnetic resonance spectroscopy (DMRS) to provide quantitative information about metabolic flux, fluorodeoxyglucose positron emission tomography (FDG-PET) to measure brain glucose metabolism, and resting state-functional MRI (rs-fMRI) to measure brain functional connectivity. We used these methods in both mice with complete GLP-1R knockout (GLP-1R KO) and wild-type C57BL/6N (WT) mice. RESULTS The metabolic rate of GLP-1R KO mice was significantly slower than that of WT mice (p = 0.0345, WT mice 0.02335 ± 0.057 mM/min, GLP-1R KO mice 0.01998 ± 0.07 mM/min). Quantification of the mean [18F]FDG signal in the whole brain also showed significantly reduced glucose uptake in GLP-1R KO mice versus control mice (p = 0.0314). Observing rs-fMRI, the functional brain connectivity in GLP-1R KO mice was significantly lower than that in the WT group (p = 0.0032 for gFCD, p = 0.0002 for whole-brain correlation, p < 0.0001 for ALFF). CONCLUSIONS GLP-1R KO mice exhibit impaired brain glucose metabolism to high doses of exogenous glucose, and they also have reduced functional connectivity. This suggests that the GLP-1R KO mouse model may serve as a model for correlated metabolic and functional connectivity loss.
Collapse
Affiliation(s)
- Hui Li
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Yujiao Fang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Da Wang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bowen Shi
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | | |
Collapse
|
5
|
Al-Onaizi M, Braysh K, Alkafeef SS, Altarrah D, Dannoon S, Alasousi D, Adel H, Al-Ajmi M, Kandari A, Najem R, Nizam R, Williams MR, John S, Thanaraj TA, Ahmad R, Al-Hussaini H, Al-Mulla F, Alzaid F. Glucose intolerance induces anxiety-like behaviors independent of obesity and insulin resistance in a novel model of nutritional metabolic stress. Nutr Neurosci 2024; 27:1143-1161. [PMID: 38319634 DOI: 10.1080/1028415x.2024.2310419] [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] [Indexed: 02/07/2024]
Abstract
OBJECTIVES Type 2 diabetes (T2D) is a metabolic disease of major public health concern. It impacts peripheral tissues and the central nervous system, leading to systemic dysmetabolism and neurocognitive impairments, including memory deficits, anxiety, and depression. The metabolic determinants of these neurocognitive impairments remain unidentified. Here, we sought to address this question by developing a proprietary (P-) high-fat diet (HFD), in which glucose intolerance precedes weight gain and insulin resistance. METHODS The P-HFD model was nutritionally characterized, and tested in vivo in mice that underwent behavioral and metabolic testing. The diet was benchmarked against reference models. . RESULTS P-HFD has 42% kcal from fat, high monounsaturated/polyunsaturated fatty acid ratio, and 10% (w/v) sucrose in drinking water. When administered, from the early stages of glucose intolerance alone, animals exhibit anxiety-like behavior, without depression nor recognition memory deficits. Long-term P-HFD feeding leads to weight gain, brain glucose hypometabolism as well as impaired recognition memory. Using an established genetic model of T2D (db/db) and of diet-induced obesity (60% kcal from fat) we show that additional insulin resistance and obesity are associated with depressive-like behaviors and recognition memory deficits. DISCUSSION Our findings demonstrate that glucose intolerance alone can elicit anxiety-like behavior. Through this study, we also provide a novel nutritional model (P-HFD) to characterize the discrete effects of glucose intolerance on cognition, behavior, and the physiology of metabolic disease.
Collapse
Affiliation(s)
- Mohammed Al-Onaizi
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Kawthar Braysh
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | - Selma S Alkafeef
- Faculty of Medicine, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Dana Altarrah
- Faculty of Public Health, Department of Social and Behavioral Science, Kuwait University, Kuwait City, Kuwait
| | - Shorouk Dannoon
- Faculty of Medicine, Department of Nuclear Medicine, Kuwait University, Kuwait City, Kuwait
| | - Dalal Alasousi
- Faculty of Science, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Hawraa Adel
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | - Mariam Al-Ajmi
- Faculty of Science, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Anwar Kandari
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | - Rawan Najem
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | | | - Sumi John
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | | | - Heba Al-Hussaini
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City, Kuwait
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| |
Collapse
|
6
|
Odusami M, Damaševičius R, Milieškaitė-Belousovienė E, Maskeliūnas R. Alzheimer's disease stage recognition from MRI and PET imaging data using Pareto-optimal quantum dynamic optimization. Heliyon 2024; 10:e34402. [PMID: 39145034 PMCID: PMC11320145 DOI: 10.1016/j.heliyon.2024.e34402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
The threat posed by Alzheimer's disease (AD) to human health has grown significantly. However, the precise diagnosis and classification of AD stages remain a challenge. Neuroimaging methods such as structural magnetic resonance imaging (sMRI) and fluorodeoxyglucose positron emission tomography (FDG-PET) have been used to diagnose and categorize AD. However, feature selection approaches that are frequently used to extract additional data from multimodal imaging are prone to errors. This paper suggests using a static pulse-coupled neural network and a Laplacian pyramid to combine sMRI and FDG-PET data. After that, the fused images are used to train the Mobile Vision Transformer (MViT), optimized with Pareto-Optimal Quantum Dynamic Optimization for Neural Architecture Search, while the fused images are augmented to avoid overfitting and then classify unfused MRI and FDG-PET images obtained from the AD Neuroimaging Initiative (ADNI) and Open Access Series of Imaging Studies (OASIS) datasets into various stages of AD. The architectural hyperparameters of MViT are optimized using Quantum Dynamic Optimization, which ensures a Pareto-optimal solution. The Peak Signal-to-Noise Ratio (PSNR), the Mean Squared Error (MSE), and the Structured Similarity Indexing Method (SSIM) are used to measure the quality of the fused image. We found that the fused image was consistent in all metrics, having 0.64 SIMM, 35.60 PSNR, and 0.21 MSE for the FDG-PET image. In the classification of AD vs. cognitive normal (CN), AD vs. mild cognitive impairment (MCI), and CN vs. MCI, the precision of the proposed method is 94.73%, 92.98% and 89.36%, respectively. The sensitivity is 90. 70%, 90. 70%, and 90. 91% while the specificity is 100%, 100%, and 85. 71%, respectively, in the ADNI MRI test data.
Collapse
Affiliation(s)
- Modupe Odusami
- Faculty of Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | | | | | - Rytis Maskeliūnas
- Faculty of Informatics, Kaunas University of Technology, Kaunas, Lithuania
| |
Collapse
|
7
|
Barzegar Behrooz A, Latifi‐Navid H, Lotfi J, Khodagholi F, Shojaei S, Ghavami S, Fahanik Babaei J. CSF amino acid profiles in ICV-streptozotocin-induced sporadic Alzheimer's disease in male Wistar rat: a metabolomics and systems biology perspective. FEBS Open Bio 2024; 14:1116-1132. [PMID: 38769074 PMCID: PMC11216934 DOI: 10.1002/2211-5463.13814] [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: 03/05/2024] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease (AD) is an increasingly important public health concern due to the increasing proportion of older individuals within the general population. The impairment of processes responsible for adequate brain energy supply primarily determines the early features of the aging process. Restricting brain energy supply results in brain hypometabolism prior to clinical symptoms and is anatomically and functionally associated with cognitive impairment. The present study investigated changes in metabolic profiles induced by intracerebroventricular-streptozotocin (ICV-STZ) in an AD-like animal model. To this end, male Wistar rats received a single injection of STZ (3 mg·kg-1) by ICV (2.5 μL into each ventricle for 5 min on each side). In the second week after receiving ICV-STZ, rats were tested for cognitive performance using the Morris Water Maze test and subsequently prepared for positron emission tomography (PET) to confirm AD-like symptoms. Tandem Mass Spectrometry (MS/MS) analysis was used to detect amino acid changes in cerebrospinal fluid (CFS) samples. Our metabolomics study revealed a reduction in the concentrations of various amino acids (alanine, arginine, aspartic acid, glutamic acid, glycine, isoleucine, methionine, phenylalanine, proline, serine, threonine, tryptophane, tyrosine, and valine) in CSF of ICV-STZ-treated animals as compared to controls rats. The results of the current study indicate amino acid levels could potentially be considered targets of nutritional and/or pharmacological interventions to interfere with AD progression.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
| | - Hamid Latifi‐Navid
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
- Department of Molecular MedicineNational Institute of Genetic Engineering and BiotechnologyTehranIran
- School of Biological SciencesInstitute for Research in Fundamental Sciences (IPM)TehranIran
| | - Jabar Lotfi
- Growth and Development Research CenterTehran University of Medical SciencesIran
| | - Fariba Khodagholi
- Neuroscience Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, College of MedicineUniversity of ManitobaWinnipegCanada
- Faculty of Medicine in ZabrzeUniversity of Technology in KatowiceZabrzePoland
- Research Institute of Oncology and HematologyCancer Care Manitoba‐University of ManitobaWinnipegCanada
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| |
Collapse
|
8
|
Malla A, Gupta S, Sur R. Glycolytic enzymes in non-glycolytic web: functional analysis of the key players. Cell Biochem Biophys 2024; 82:351-378. [PMID: 38196050 DOI: 10.1007/s12013-023-01213-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
To survive in the tumour microenvironment, cancer cells undergo rapid metabolic reprograming and adaptability. One of the key characteristics of cancer is increased glycolytic selectivity and decreased oxidative phosphorylation (OXPHOS). Apart from ATP synthesis, glycolysis is also responsible for NADH regeneration and macromolecular biosynthesis, such as amino acid biosynthesis and nucleotide biosynthesis. This allows cancer cells to survive and proliferate even in low-nutrient and oxygen conditions, making glycolytic enzymes a promising target for various anti-cancer agents. Oncogenic activation is also caused by the uncontrolled production and activity of glycolytic enzymes. Nevertheless, in addition to conventional glycolytic processes, some glycolytic enzymes are involved in non-canonical functions such as transcriptional regulation, autophagy, epigenetic changes, inflammation, various signaling cascades, redox regulation, oxidative stress, obesity and fatty acid metabolism, diabetes and neurodegenerative disorders, and hypoxia. The mechanisms underlying the non-canonical glycolytic enzyme activities are still not comprehensive. This review summarizes the current findings on the mechanisms fundamental to the non-glycolytic actions of glycolytic enzymes and their intermediates in maintaining the tumor microenvironment.
Collapse
Affiliation(s)
- Avirup Malla
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India
| | - Suvroma Gupta
- Department of Aquaculture Management, Khejuri college, West Bengal, Baratala, India.
| | - Runa Sur
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India.
| |
Collapse
|
9
|
Salcedo C, Pozo Garcia V, García-Adán B, Ameen AO, Gegelashvili G, Waagepetersen HS, Freude KK, Aldana BI. Increased glucose metabolism and impaired glutamate transport in human astrocytes are potential early triggers of abnormal extracellular glutamate accumulation in hiPSC-derived models of Alzheimer's disease. J Neurochem 2024; 168:822-840. [PMID: 38063257 DOI: 10.1111/jnc.16014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 05/19/2024]
Abstract
Glutamate recycling between neurons and astrocytes is essential to maintain neurotransmitter homeostasis. Disturbances in glutamate homeostasis, resulting in excitotoxicity and neuronal death, have been described as a potential mechanism in Alzheimer's disease (AD) pathophysiology. However, glutamate neurotransmitter metabolism in different human brain cells, particularly astrocytes, has been poorly investigated at the early stages of AD. We sought to investigate glucose and glutamate metabolism in AD by employing human induced pluripotent stem cell (hiPSC)-derived astrocytes and neurons carrying mutations in the amyloid precursor protein (APP) or presenilin-1 (PSEN-1) gene as found in familial types of AD (fAD). Methods such as live-cell bioenergetics and metabolic mapping using [13C]-enriched substrates were used to examine metabolism in the early stages of AD. Our results revealed greater glycolysis and glucose oxidative metabolism in astrocytes and neurons with APP or PSEN-1 mutations, accompanied by an elevated glutamate synthesis compared to control WT cells. Astrocytes with APP or PSEN-1 mutations exhibited reduced expression of the excitatory amino acid transporter 2 (EAAT2), and glutamine uptake increased in mutated neurons, with enhanced glutamate release specifically in neurons with a PSEN-1 mutation. These results demonstrate a hypermetabolic phenotype in astrocytes with fAD mutations possibly linked to toxic glutamate accumulation. Our findings further identify metabolic imbalances that may occur in the early phases of AD pathophysiology.
Collapse
Affiliation(s)
- Claudia Salcedo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Victoria Pozo Garcia
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bernat García-Adán
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aishat O Ameen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Georgi Gegelashvili
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Mravinacová S, Alanko V, Bergström S, Bridel C, Pijnenburg Y, Hagman G, Kivipelto M, Teunissen C, Nilsson P, Matton A, Månberg A. CSF protein ratios with enhanced potential to reflect Alzheimer's disease pathology and neurodegeneration. Mol Neurodegener 2024; 19:15. [PMID: 38350954 PMCID: PMC10863228 DOI: 10.1186/s13024-024-00705-z] [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: 08/31/2023] [Accepted: 01/23/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Amyloid and tau aggregates are considered to cause neurodegeneration and consequently cognitive decline in individuals with Alzheimer's disease (AD). Here, we explore the potential of cerebrospinal fluid (CSF) proteins to reflect AD pathology and cognitive decline, aiming to identify potential biomarkers for monitoring outcomes of disease-modifying therapies targeting these aggregates. METHOD We used a multiplex antibody-based suspension bead array to measure the levels of 49 proteins in CSF from the Swedish GEDOC memory clinic cohort at the Karolinska University Hospital. The cohort comprised 148 amyloid- and tau-negative individuals (A-T-) and 65 amyloid- and tau-positive individuals (A+T+). An independent sample set of 26 A-T- and 26 A+T+ individuals from the Amsterdam Dementia Cohort was used for validation. The measured proteins were clustered based on their correlation to CSF amyloid beta peptides, tau and NfL levels. Further, we used support vector machine modelling to identify protein pairs, matched based on their cluster origin, that reflect AD pathology and cognitive decline with improved performance compared to single proteins. RESULTS The protein-clustering revealed 11 proteins strongly correlated to t-tau and p-tau (tau-associated group), including mainly synaptic proteins previously found elevated in AD such as NRGN, GAP43 and SNCB. Another 16 proteins showed predominant correlation with Aβ42 (amyloid-associated group), including PTPRN2, NCAN and CHL1. Support vector machine modelling revealed that proteins from the two groups combined in pairs discriminated A-T- from A+T+ individuals with higher accuracy compared to single proteins, as well as compared to protein pairs composed of proteins originating from the same group. Moreover, combining the proteins from different groups in ratios (tau-associated protein/amyloid-associated protein) significantly increased their correlation to cognitive decline measured with cognitive scores. The results were validated in an independent cohort. CONCLUSIONS Combining brain-derived proteins in pairs largely enhanced their capacity to discriminate between AD pathology-affected and unaffected individuals and increased their correlation to cognitive decline, potentially due to adjustment of inter-individual variability. With these results, we highlight the potential of protein pairs to monitor neurodegeneration and thereby possibly the efficacy of AD disease-modifying therapies.
Collapse
Affiliation(s)
- Sára Mravinacová
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Vilma Alanko
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Claire Bridel
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Yolande Pijnenburg
- Department of Neurology, Alzheimer Centre, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Göran Hagman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology (AGE) Research Unit, Imperial College London, London, United Kingdom
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Charlotte Teunissen
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Anna Matton
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology (AGE) Research Unit, Imperial College London, London, United Kingdom
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.
| |
Collapse
|
11
|
Abou-El-Hassan H, Bernstock JD, Chalif JI, Yahya T, Rezende RM, Weiner HL, Izzy S. Elucidating the neuroimmunology of traumatic brain injury: methodological approaches to unravel intercellular communication and function. Front Cell Neurosci 2023; 17:1322325. [PMID: 38162004 PMCID: PMC10756680 DOI: 10.3389/fncel.2023.1322325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024] Open
Abstract
The neuroimmunology of traumatic brain injury (TBI) has recently gained recognition as a crucial element in the secondary pathophysiological consequences that occur following neurotrauma. Both immune cells residing within the central nervous system (CNS) and those migrating from the periphery play significant roles in the development of secondary brain injury. However, the precise mechanisms governing communication between innate and adaptive immune cells remain incompletely understood, partly due to a limited utilization of relevant experimental models and techniques. Therefore, in this discussion, we outline current methodologies that can aid in the exploration of TBI neuroimmunology, with a particular emphasis on the interactions between resident neuroglial cells and recruited lymphocytes. These techniques encompass adoptive cell transfer, intra-CNS injection(s), selective cellular depletion, genetic manipulation, molecular neuroimaging, as well as in vitro co-culture systems and the utilization of organoid models. By incorporating key elements of both innate and adaptive immunity, these methods facilitate the examination of clinically relevant interactions. In addition to these preclinical approaches, we also detail an emerging avenue of research that seeks to leverage human biofluids. This approach enables the investigation of how resident and infiltrating immune cells modulate neuroglial responses after TBI. Considering the growing significance of neuroinflammation in TBI, the introduction and application of advanced methodologies will be pivotal in advancing translational research in this field.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joshua I. Chalif
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Taha Yahya
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Saef Izzy
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Hao L, Wang L, Ju M, Feng W, Guo Z, Sun X, Xiao R. 27-Hydroxycholesterol impairs learning and memory ability via decreasing brain glucose uptake mediated by the gut microbiota. Biomed Pharmacother 2023; 168:115649. [PMID: 37806088 DOI: 10.1016/j.biopha.2023.115649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
Brain glucose hypometabolism is a significant manifestation of Alzheimer's disease (AD). 27-hydroxycholesterol (27-OHC) and the gut microbiota have been recognized as factors possibly influencing the pathogenesis of AD. This study aimed to investigate the link between 27-OHC, the gut microbiota, and brain glucose uptake in AD. Here, 6-month-old male C57BL/6 J mice were treated with sterile water or antibiotic cocktails, with or without 27-OHC and/or 27-OHC synthetic enzyme CYP27A1 inhibitor anastrozole (ANS). The gut microbiota, brain glucose uptake levels, and memory ability were measured. We observed that 27-OHC altered microbiota composition, damaged brain tissue structures, decreased the 2-deoxy-2-[18 F] fluorodeoxyglucose (18F-FDG) uptake value, downregulated the gene expression of glucose transporter type 4 (GLUT4), reduced the colocalization of GLUT1/glial fibrillary acidic protein (GFAP) in the hippocampus, and impaired spatial memory. ANS reversed the effects of 27-OHC. The antibiotic-treated mice did not exhibit similar results after 27-OHC treatment. This study reveals a potential molecular mechanism wherein 27-OHC-induced memory impairment might be linked to reduced brain glucose uptake, mediated by the gut microbiota.
Collapse
Affiliation(s)
- Ling Hao
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Lijing Wang
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Mengwei Ju
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Wenjing Feng
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Zhiting Guo
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Xuejing Sun
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Rong Xiao
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China.
| |
Collapse
|
13
|
Li X, Gao Y, Li B, Zhao W, Cai Q, Yin W, Zeng S, Li X, Gao H, Cheng M. Integrated proteomics and metabolomics analysis of D-pinitol function during hippocampal damage in streptozocin-induced aging-accelerated mice. Front Mol Neurosci 2023; 16:1251513. [PMID: 38025258 PMCID: PMC10664147 DOI: 10.3389/fnmol.2023.1251513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Diabetes can cause hippocampal damage and lead to cognitive impairment. Diabetic cognitive impairment (DCI) is a chronic complication of diabetes associated with a high disability rate; however, its pathogenesis and therapeutic targets are unclear. We aimed to explore the mechanism of hippocampal damage during diabetes and evaluate the potential role of D-pinitol (DP) in protecting hippocampal tissue and improving cognitive dysfunction. Methods DP (150 mg/kg/day) was administered intragastrically to streptozocin-induced aging-accelerated mice for 8 weeks. Hippocampal tissues were examined using tandem mass tag (TMT)-based proteomics and liquid chromatography-mass spectrometry (LC-MS)/MS-based non-targeted metabolomic analysis. Differentially expressed proteins (DEPs) and differentially regulated metabolites (DRMs) were screened for further analysis, and some DEPs were verified using western blotting. Results Our results showed that 329 proteins had significantly altered hippocampal expression in untreated diabetic mice (DM), which was restored to normal after DP treatment in 72 cases. In total, 207 DRMs were identified in the DM group, and the expression of 32 DRMs was restored to normal post-DP treatment. These proteins and metabolites are involved in metabolic pathways (purine metabolism, arginine and proline metabolism, and histidine metabolism), actin cytoskeleton regulation, oxidative phosphorylation, and Rap1-mediated signaling. Conclusions Our study may help to better understand the mechanism of diabetic hippocampal damage and cognitive impairment and suggest a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaoxia Li
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
- Department of Diabetes, The Third People's Hospital of Gansu Province, Lanzhou, China
| | - Yuan Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Baoying Li
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Health Management Center (East Area), Qilu Hospital of Shandong University, Jinan, China
| | - Wenqian Zhao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Qian Cai
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Wenbin Yin
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Shudong Zeng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Xiaoli Li
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiqing Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Mei Cheng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| |
Collapse
|
14
|
Abstract
Recently developed molecular imaging approaches can be used to visualize specific host responses and pathology in a quest to image infections where few microbe-specific tracers have been developed and in recognition that host responses contribute to morbidity and mortality in their own right. Here we highlight several recent examples of these imaging approaches adapted for imaging infections. The early successes and new avenues described here encompass diverse imaging modalities and leverage diverse aspects of the host response to infection-including inflammation, tissue injury and healing, and key nutrients during host-pathogen interactions. Clearly, these approaches merit further preclinical and clinical study as they are complementary and orthogonal to the pathogen-focused imaging modalities currently under investigation.
Collapse
Affiliation(s)
- Catherine A Foss
- Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adam R Renslo
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
15
|
Bertsch M, Franchi B, Tesi MC, Tora V. The role of A[Formula: see text] and Tau proteins in Alzheimer's disease: a mathematical model on graphs. J Math Biol 2023; 87:49. [PMID: 37646953 PMCID: PMC10468937 DOI: 10.1007/s00285-023-01985-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/25/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023]
Abstract
In this Note we study a mathematical model for the progression of Alzheimer's Disease in the human brain. The novelty of our approach consists in the representation of the brain as two superposed graphs where toxic proteins diffuse, the connectivity graph which represents the neural network, and the proximity graph which takes into account the extracellular space. Toxic proteins such as [Formula: see text] amyloid and Tau play in fact a crucial role in the development of Alzheimer's disease and, separately, have been targets of medical treatments. Recent biomedical literature stresses the potential impact of the synergetic action of these proteins. We numerically test various modelling hypotheses which confirm the relevance of this synergy.
Collapse
Affiliation(s)
- Michiel Bertsch
- Department of Mathematics, University of Roma “Tor Vergata”, Rome, Italy
- Istituto per le Applicazioni del Calcolo “M. Picone”, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Bruno Franchi
- Department of Mathematics, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Maria Carla Tesi
- Department of Mathematics, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Veronica Tora
- Department of Mathematics, University of Roma “Tor Vergata”, Rome, Italy
| |
Collapse
|
16
|
Palumbo G, Kunze LH, Oos R, Wind-Mark K, Lindner S, von Ungern-Sternberg B, Bartenstein P, Ziegler S, Brendel M. Longitudinal Studies on Alzheimer Disease Mouse Models with Multiple Tracer PET/CT: Application of Reduction and Refinement Principles in Daily Practice to Safeguard Animal Welfare during Progressive Aging. Animals (Basel) 2023; 13:1812. [PMID: 37531139 PMCID: PMC10251952 DOI: 10.3390/ani13111812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 08/03/2023] Open
Abstract
Longitudinal studies on mouse models related to Alzheimer disease (AD) pathology play an important role in the investigation of therapeutic targets to help pharmaceutical research in the development of new drugs and in the attempt of an early diagnosis that can contribute to improving people's quality of life. There are several advantages to enriching longitudinal studies in AD models with Positron Emission Tomography (PET); among these advantages, the possibility of following the principle of the 3Rs of animal welfare is fundamental. In this manuscript, good daily experimental practice focusing on animal welfare is described and commented upon, based on the experience attained from studies conducted in our Nuclear Medicine department.
Collapse
Affiliation(s)
- Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Lea Helena Kunze
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Rosel Oos
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital LMU Munich, Marchionini Strasse 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| |
Collapse
|
17
|
Lee RL, Funk KE. Imaging blood–brain barrier disruption in neuroinflammation and Alzheimer’s disease. Front Aging Neurosci 2023; 15:1144036. [PMID: 37009464 PMCID: PMC10063921 DOI: 10.3389/fnagi.2023.1144036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
The blood–brain barrier (BBB) is the neurovascular structure that regulates the passage of cells and molecules to and from the central nervous system (CNS). Alzheimer’s disease (AD) is a neurodegenerative disorder that is associated with gradual breakdown of the BBB, permitting entry of plasma-derived neurotoxins, inflammatory cells, and microbial pathogens into the CNS. BBB permeability can be visualized directly in AD patients using imaging technologies including dynamic contrast-enhanced and arterial spin labeling magnetic resonance imaging, and recent studies employing these techniques have shown that subtle changes in BBB stability occur prior to deposition of the pathological hallmarks of AD, senile plaques, and neurofibrillary tangles. These studies suggest that BBB disruption may be useful as an early diagnostic marker; however, AD is also accompanied by neuroinflammation, which can complicate these analyses. This review will outline the structural and functional changes to the BBB that occur during AD pathogenesis and highlight current imaging technologies that can detect these subtle changes. Advancing these technologies will improve both the diagnosis and treatment of AD and other neurodegenerative diseases.
Collapse
|
18
|
Duan J, Liu Y, Wu H, Wang J, Chen L, Chen CLP. Broad learning for early diagnosis of Alzheimer's disease using FDG-PET of the brain. Front Neurosci 2023; 17:1137567. [PMID: 36992851 PMCID: PMC10040750 DOI: 10.3389/fnins.2023.1137567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and the development of AD is irreversible. However, preventive measures in the presymptomatic stage of AD can effectively slow down deterioration. Fluorodeoxyglucose positron emission tomography (FDG-PET) can detect the metabolism of glucose in patients' brains, which can help to identify changes related to AD before brain damage occurs. Machine learning is useful for early diagnosis of patients with AD using FDG-PET, but it requires a sufficiently large dataset, and it is easy for overfitting to occur in small datasets. Previous studies using machine learning for early diagnosis with FDG-PET have either involved the extraction of elaborately handcrafted features or validation on a small dataset, and few studies have explored the refined classification of early mild cognitive impairment (EMCI) and late mild cognitive impairment (LMCI). This article presents a broad network-based model for early diagnosis of AD (BLADNet) through PET imaging of the brain; this method employs a novel broad neural network to enhance the features of FDG-PET extracted via 2D CNN. BLADNet can search for information over a broad space through the addition of new BLS blocks without retraining of the whole network, thus improving the accuracy of AD classification. Experiments conducted on a dataset containing 2,298 FDG-PET images of 1,045 subjects from the ADNI database demonstrate that our methods are superior to those used in previous studies on early diagnosis of AD with FDG-PET. In particular, our methods achieved state-of-the-art results in EMCI and LMCI classification with FDG-PET.
Collapse
Affiliation(s)
- Junwei Duan
- College of Information Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, China
- *Correspondence: Junwei Duan
| | - Yang Liu
- College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Huanhua Wu
- Department of Nuclear Medicine and PET/CT-MRI Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing Wang
- School of Computer Science, Guangdong Polytechnic Normal University, Guangzhou, China
- Jing Wang
| | - Long Chen
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Taipa, Macau SAR, China
| | - C. L. Philip Chen
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
19
|
Chronic exposure to a synthetic cannabinoid alters cerebral brain metabolism and causes long-lasting behavioral deficits in adult mice. J Neural Transm (Vienna) 2023:10.1007/s00702-023-02607-8. [PMID: 36853560 PMCID: PMC10374737 DOI: 10.1007/s00702-023-02607-8] [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: 11/14/2022] [Accepted: 02/17/2023] [Indexed: 03/01/2023]
Abstract
In recent years, there has been growing evidence that cannabinoids have promising medicinal and pharmacological effects. However, the growing interest in medical cannabis highlights the need to better understand brain alterations linking phytocannabinoids or synthetic cannabinoids to clinical and behavioral phenotypes. Therefore, the aim of this study was to investigate the effects of long-term WIN 55,212-2 treatment-with and without prolonged abstinence-on cerebral metabolism and memory function in healthy wildtype mice. Adult C57BI/6J mice were divided into two treatment groups to study the acute effects of WIN 55,212-2 treatment as well the effects of WIN 55,212-2 treatment after an extended washout phase. We could demonstrate that 3 mg/kg WIN 55,212-2 treatment in early adulthood leads to a hypometabolism in several brain regions including the hippocampus, cerebellum, amygdala and midbrain, even after prolonged abstinence. Furthermore, prolonged acute WIN 55,212-2 treatment in 6-months-old mice reduced the glucose metabolism in the hippocampus and midbrain. In addition, Win 55,212-2 treatment during adulthood lead to spatial memory and recognition memory deficits without affecting anxiety behavior. Overall we could demonstrate that treatment with the synthetic CB1/CB2 receptor aganist Win 55,212-2 during adulthood causes persistent memory deficits, especially when mice were treated in early adulthood. Our findings highlight the risks of prolonged WIN 55,212-2 use and provide new insights into the mechanisms underlying the effects of chronic cannabinoid exposure on the brain and behavior.
Collapse
|
20
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-specific effects of high-fat diet on rat brain glucose metabolism and early-onset dementia symptoms. Mech Ageing Dev 2023; 211:111795. [PMID: 36828273 DOI: 10.1016/j.mad.2023.111795] [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] [Received: 11/14/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Peripheral metabolic disturbances are associated with a variety of clinical health consequences and may contribute to the development of neurocognitive disorders. This study investigates whether long-term high-fat diet (HFD) consumption changes the brain glucose metabolism and impairs memory performance in a sex-dependent manner. Male and female rats, after weaning, were fed HFD or normal chow diet (NCD) for 16 weeks. Behavioral tests for spatial memory and an 18 F-FDG-PET scan were performed. Also, the expression of brain insulin resistance markers and Alzheimer's pathology-related genes was assessed by qPCR. The Morris water maze and Y-maze results showed, respectively, that memory retrieval and spatial working memory were impaired only in HFD male rats compared to NCD controls. In addition, measuring whole brain 18 F-FDG uptake indicated a significant reduction in glucose metabolism in male but not female HFD rats. Analysis of 15 genes related to glucose metabolism and Alzheimer's pathology, in the hippocampus, showed that expression of GLUT3, IRS2, and IDE is significantly reduced in HFD male rats. Our results suggest that sex affects the HFD-induced dysregulation of brain glucose metabolism and cognitive performance.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Sánchez CQ, Schmitt FW, Curdt N, Westhoff AC, Bänfer IWH, Bayer TA, Bouter Y. Search Strategy Analysis of 5xFAD Alzheimer Mice in the Morris Water Maze Reveals Sex- and Age-Specific Spatial Navigation Deficits. Biomedicines 2023; 11:biomedicines11020599. [PMID: 36831135 PMCID: PMC9953202 DOI: 10.3390/biomedicines11020599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Spatial disorientation and navigational impairments are not only some of the first memory deficits in Alzheimer's disease, but are also very disease-specific. In rodents, the Morris Water Maze is used to investigate spatial navigation and memory. Here, we examined the spatial memory in the commonly used 5xFAD Alzheimer mouse model in a sex- and age-dependent manner. Our findings show first spatial learning deficits in 7-month-old female 5xFAD and 12-month-old male 5xFAD mice, respectively. While the assessment of spatial working memory using escape latencies provides a global picture of memory performance, it does not explain how an animal solves a spatial task. Therefore, a detailed analysis of swimming strategies was performed to better understand the behavioral differences between 5xFAD and WT mice. 5xFAD mice used a qualitatively and quantitatively different search strategy pattern than wildtype animals that used more non-spatial strategies and showed allocentric-specific memory deficits. Furthermore, a detailed analysis of swimming strategies revealed allocentric memory deficits in the probe trial in female 3-month-old and male 7-month-old 5xFAD animals before the onset of severe reference memory deficits. Overall, we could demonstrate that spatial navigation deficits in 5xFAD mice are age- and sex-dependent, with female mice being more severely affected. In addition, the implementation of a search strategy classification system allowed an earlier detection of behavioral differences and therefore could be a powerful tool for preclinical drug testing in the 5xFAD model.
Collapse
Affiliation(s)
- Carolina Quintanilla Sánchez
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Franziska W. Schmitt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Nadine Curdt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Anna Celine Westhoff
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Irina Wanda Helene Bänfer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Thomas A. Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), 37075 Goettingen, Germany
- Correspondence:
| |
Collapse
|
22
|
Moyaert P, Padrela BE, Morgan CA, Petr J, Versijpt J, Barkhof F, Jurkiewicz MT, Shao X, Oyeniran O, Manson T, Wang DJJ, Günther M, Achten E, Mutsaerts HJMM, Anazodo UC. Imaging blood-brain barrier dysfunction: A state-of-the-art review from a clinical perspective. Front Aging Neurosci 2023; 15:1132077. [PMID: 37139088 PMCID: PMC10150073 DOI: 10.3389/fnagi.2023.1132077] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/15/2023] [Indexed: 05/05/2023] Open
Abstract
The blood-brain barrier (BBB) consists of specialized cells that tightly regulate the in- and outflow of molecules from the blood to brain parenchyma, protecting the brain's microenvironment. If one of the BBB components starts to fail, its dysfunction can lead to a cascade of neuroinflammatory events leading to neuronal dysfunction and degeneration. Preliminary imaging findings suggest that BBB dysfunction could serve as an early diagnostic and prognostic biomarker for a number of neurological diseases. This review aims to provide clinicians with an overview of the emerging field of BBB imaging in humans by answering three key questions: (1. Disease) In which diseases could BBB imaging be useful? (2. Device) What are currently available imaging methods for evaluating BBB integrity? And (3. Distribution) what is the potential of BBB imaging in different environments, particularly in resource limited settings? We conclude that further advances are needed, such as the validation, standardization and implementation of readily available, low-cost and non-contrast BBB imaging techniques, for BBB imaging to be a useful clinical biomarker in both resource-limited and well-resourced settings.
Collapse
Affiliation(s)
- Paulien Moyaert
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Lawson Health Research Institute, London, ON, Canada
- Department of Neurology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- *Correspondence: Paulien Moyaert,
| | - Beatriz E. Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Catherine A. Morgan
- School of Psychology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand
- Centre for Advanced MRI, Auckland UniServices Limited, Auckland, New Zealand
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jan Versijpt
- Department of Neurology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, United Kingdom
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Olujide Oyeniran
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Tabitha Manson
- Centre for Advanced MRI, Auckland UniServices Limited, Auckland, New Zealand
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Danny J. J. Wang
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine, University of Bremen, Bremen, Germany
| | - Eric Achten
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | - Henk J. M. M. Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Udunna C. Anazodo
- Lawson Health Research Institute, London, ON, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
Boyle AJ, Murrell E, Tong J, Schifani C, Narvaez A, Wuest M, West F, Wuest F, Vasdev N. PET Imaging of Fructose Metabolism in a Rodent Model of Neuroinflammation with 6-[ 18F]fluoro-6-deoxy-D-fructose. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238529. [PMID: 36500626 PMCID: PMC9736258 DOI: 10.3390/molecules27238529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Fluorine-18 labeled 6-fluoro-6-deoxy-D-fructose (6-[18F]FDF) targets the fructose-preferred facilitative hexose transporter GLUT5, which is expressed predominantly in brain microglia and activated in response to inflammatory stimuli. We hypothesize that 6-[18F]FDF will specifically image microglia following neuroinflammatory insult. 6-[18F]FDF and, for comparison, [18F]FDG were evaluated in unilateral intra-striatal lipopolysaccharide (LPS)-injected male and female rats (50 µg/animal) by longitudinal dynamic PET imaging in vivo. In LPS-injected rats, increased accumulation of 6-[18F]FDF was observed at 48 h post-LPS injection, with plateaued uptake (60-120 min) that was significantly higher in the ipsilateral vs. contralateral striatum (0.985 ± 0.047 and 0.819 ± 0.033 SUV, respectively; p = 0.002, n = 4M/3F). The ipsilateral-contralateral difference in striatal 6-[18F]FDF uptake expressed as binding potential (BPSRTM) peaked at 48 h (0.19 ± 0.11) and was significantly decreased at one and two weeks. In contrast, increased [18F]FDG uptake in the ipsilateral striatum was highest at one week post-LPS injection (BPSRTM = 0.25 ± 0.06, n = 4M). Iba-1 and GFAP immunohistochemistry confirmed LPS-induced activation of microglia and astrocytes, respectively, in ipsilateral striatum. This proof-of-concept study revealed an early response of 6-[18F]FDF to neuroinflammatory stimuli in rat brain. 6-[18F]FDF represents a potential PET radiotracer for imaging microglial GLUT5 density in brain with applications in neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Amanda J. Boyle
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Christin Schifani
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Andrea Narvaez
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Melinda Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
| | - Frederick West
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| |
Collapse
|
24
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
25
|
Lio CT, Kacprowski T, Klaedtke M, Jensen LR, Bouter Y, Bayer TA, Kuss AW. Small RNA Sequencing in the Tg4–42 Mouse Model Suggests the Involvement of snoRNAs in the Etiology of Alzheimer’s Disease. J Alzheimers Dis 2022; 87:1671-1681. [DOI: 10.3233/jad-220110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The Tg4-42 mouse model for sporadic Alzheimer’s disease (AD) has unique features, as the neuronal expression of wild type N-truncated Aβ4–42 induces an AD-typical neurological phenotype in the absence of plaques. It is one of the few models developing neuron death in the CA1 region of the hippocampus. As such, it could serve as a powerful tool for preclinical drug testing and identification of the underlying molecular pathways that drive the pathology of AD. Objective: The aim of this study was to use a differential co-expression analysis approach for analyzing a small RNA sequencing dataset from a well-established murine model in order to identify potentially new players in the etiology of AD. Methods: To investigate small nucleolar RNAs in the hippocampus of Tg4-42 mice, we used RNA-Seq data from this particular tissue and, instead of analyzing the data at single gene level, employed differential co-expression analysis, which takes the comparison to gene pair level and thus affords a new angle to the interpretation of these data. Results: We identified two clusters of differentially correlated small RNAs, including Snord55, Snord57, Snord49a, Snord12, Snord38a, Snord99, Snord87, Mir1981, Mir106b, Mir30d, Mir598, and Mir99b. Interestingly, some of them have been reported to be functionally relevant in AD pathogenesis, as AD biomarkers, regulating tau phosphorylation, TGF-β receptor function or Aβ metabolism. Conclusion: The majority of snoRNAs for which our results suggest a potential role in the etiology of AD were so far not conspicuously implicated in the context of AD pathogenesis and could thus point towards interesting new avenues of research in this field.
Collapse
Affiliation(s)
- Chit Tong Lio
- Chair of Experimental Bioinformatics, TechnicalUniversity of Munich, Freising, Germany
- Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Maik Klaedtke
- Department of Functional Genomics, Human Molecular Genetics Group, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Lars R. Jensen
- Department of Functional Genomics, Human Molecular Genetics Group, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Thomas A. Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Andreas W. Kuss
- Department of Functional Genomics, Human Molecular Genetics Group, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
26
|
Curdt N, Schmitt FW, Bouter C, Iseni T, Weile HC, Altunok B, Beindorff N, Bayer TA, Cooke MB, Bouter Y. Search strategy analysis of Tg4-42 Alzheimer Mice in the Morris Water Maze reveals early spatial navigation deficits. Sci Rep 2022; 12:5451. [PMID: 35361814 PMCID: PMC8971530 DOI: 10.1038/s41598-022-09270-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/18/2022] [Indexed: 12/20/2022] Open
Abstract
Spatial disorientation is one of the earliest symptoms in Alzheimer’s disease and allocentric deficits can already be detected in the asymptomatic preclinical stages of the disease. The Morris Water Maze (MWM) is used to study spatial learning in rodent models. Here we investigated the spatial memory of female 3, 7 and 12 month-old Alzheimer Tg4-42 mice in comparison to wild-type control animals. Conventional behavior analysis of escape latencies and quadrant preference revealed spatial memory and reference memory deficits in female 7 and 12 month-old Tg4-42 mice. In contrast, conventional analysis of the MWM indicated an intact spatial memory in 3 month-old Tg4-42 mice. However, a detailed analysis of the swimming strategies demonstrated allocentric-specific memory deficits in 3 month-old Tg4-42 mice before the onset of severe memory deficits. Furthermore, we could show that the spatial reference memory deficits in aged Tg4-42 animals are caused by the lack of allocentric and spatial strategies. Analyzing search strategies in the MWM allows to differentiate between hippocampus-dependent allocentric and hippocampus-independent egocentric search strategies. The spatial navigation impairments in young Tg4-42 mice are well in line with the hypometabolism and synaptic deficits in the hippocampus. Therefore, analyzing search strategies in the Tg4-42 model can be a powerful tool for preclinical drug testing and identifying early therapeutic successes.
Collapse
Affiliation(s)
- Nadine Curdt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Franziska W Schmitt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Trendelina Iseni
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Hanna C Weile
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Berfin Altunok
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-University Medicine Berlin, Berlin, Germany
| | - Thomas A Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany
| | - Matthew B Cooke
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Psychology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
27
|
Effect of Denoising and Deblurring 18F-Fluorodeoxyglucose Positron Emission Tomography Images on a Deep Learning Model’s Classification Performance for Alzheimer’s Disease. Metabolites 2022; 12:metabo12030231. [PMID: 35323674 PMCID: PMC8954205 DOI: 10.3390/metabo12030231] [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: 02/14/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common progressive neurodegenerative disease. 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) is widely used to predict AD using a deep learning model. However, the effects of noise and blurring on 18F-FDG PET images were not considered. The performance of a classification model trained using raw, deblurred (by the fast total variation deblurring method), or denoised (by the median modified Wiener filter) 18F-FDG PET images without or with cropping around the limbic system area using a 3D deep convolutional neural network was investigated. The classification model trained using denoised whole-brain 18F-FDG PET images achieved classification performance (0.75/0.65/0.79/0.39 for sensitivity/specificity/F1-score/Matthews correlation coefficient (MCC), respectively) higher than that with raw and deblurred 18F-FDG PET images. The classification model trained using cropped raw 18F-FDG PET images achieved higher performance (0.78/0.63/0.81/0.40 for sensitivity/specificity/F1-score/MCC) than the whole-brain 18F-FDG PET images (0.72/0.32/0.71/0.10 for sensitivity/specificity/F1-score/MCC, respectively). The 18F-FDG PET image deblurring and cropping (0.89/0.67/0.88/0.57 for sensitivity/specificity/F1-score/MCC) procedures were the most helpful for improving performance. For this model, the right middle frontal, middle temporal, insula, and hippocampus areas were the most predictive of AD using the class activation map. Our findings demonstrate that 18F-FDG PET image preprocessing and cropping improves the explainability and potential clinical applicability of deep learning models.
Collapse
|
28
|
Discovery of a novel pseudo β-hairpin structure of N-truncated amyloid-β for use as a vaccine against Alzheimer's disease. Mol Psychiatry 2022; 27:840-848. [PMID: 34776512 DOI: 10.1038/s41380-021-01385-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022]
Abstract
One of the hallmarks of Alzheimer's disease (AD) are deposits of amyloid-beta (Aβ) protein in amyloid plaques in the brain. The Aβ peptide exists in several forms, including full-length Aβ1-42 and Aβ1-40 - and the N-truncated species, pyroglutamate Aβ3-42 and Aβ4-42, which appear to play a major role in neurodegeneration. We previously identified a murine antibody (TAP01), which binds specifically to soluble, non-plaque N-truncated Aβ species. By solving crystal structures for TAP01 family antibodies bound to pyroglutamate Aβ3-14, we identified a novel pseudo β-hairpin structure in the N-terminal region of Aβ and show that this underpins its unique binding properties. We engineered a stabilised cyclic form of Aβ1-14 (N-Truncated Amyloid Peptide AntibodieS; the 'TAPAS' vaccine) and showed that this adopts the same 3-dimensional conformation as the native sequence when bound to TAP01. Active immunisation of two mouse models of AD with the TAPAS vaccine led to a striking reduction in amyloid-plaque formation, a rescue of brain glucose metabolism, a stabilisation in neuron loss, and a rescue of memory deficiencies. Treating both models with the humanised version of the TAP01 antibody had similar positive effects. Here we report the discovery of a unique conformational epitope in the N-terminal region of Aβ, which offers new routes for active and passive immunisation against AD.
Collapse
|
29
|
Effects of Chronic Arginase Inhibition with Norvaline on Tau Pathology and Brain Glucose Metabolism in Alzheimer's Disease Mice. Neurochem Res 2022; 47:1255-1268. [DOI: 10.1007/s11064-021-03519-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022]
|
30
|
Bouter C, Irwin C, Franke TN, Beindorff N, Bouter Y. Quantitative Brain Positron Emission Tomography in Female 5XFAD Alzheimer Mice: Pathological Features and Sex-Specific Alterations. Front Med (Lausanne) 2021; 8:745064. [PMID: 34901060 PMCID: PMC8661108 DOI: 10.3389/fmed.2021.745064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Successful back-translating clinical biomarkers and molecular imaging methods of Alzheimer's disease (AD), including positron emission tomography (PET), are very valuable for the evaluation of new therapeutic strategies and increase the quality of preclinical studies. 18F-Fluorodeoxyglucose (FDG)–PET and 18F-Florbetaben–PET are clinically established biomarkers capturing two key pathological features of AD. However, the suitability of 18F-FDG– and amyloid–PET in the widely used 5XFAD mouse model of AD is still unclear. Furthermore, only data on male 5XFAD mice have been published so far, whereas studies in female mice and possible sex differences in 18F-FDG and 18F-Florbetaben uptake are missing. The aim of this study was to evaluate the suitability of 18F-FDG– and 18F-Florbetaben–PET in 7-month-old female 5XFAD and to assess possible sex differences between male and female 5XFAD mice. We could demonstrate that female 5XFAD mice showed a significant reduction in brain glucose metabolism and increased cerebral amyloid deposition compared with wild type animals, in accordance with the pathology seen in AD patients. Furthermore, we showed for the first time that the hypometabolism in 5XFAD mice is gender-dependent and more pronounced in female mice. Therefore, these results support the feasibility of small animal PET imaging with 18F-FDG- and 18F-Florbetaben in 5XFAD mice in both, male and female animals. Moreover, our findings highlight the need to account for sex differences in studies working with 5XFAD mice.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Timon N Franke
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
31
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
32
|
Pursuit of precision medicine: Systems biology approaches in Alzheimer's disease mouse models. Neurobiol Dis 2021; 161:105558. [PMID: 34767943 PMCID: PMC10112395 DOI: 10.1016/j.nbd.2021.105558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a complex disease that is mediated by numerous factors and manifests in various forms. A systems biology approach to studying AD involves analyses of various body systems, biological scales, environmental elements, and clinical outcomes to understand the genotype to phenotype relationship that potentially drives AD development. Currently, there are many research investigations probing how modifiable and nonmodifiable factors impact AD symptom presentation. This review specifically focuses on how imaging modalities can be integrated into systems biology approaches using model mouse populations to link brain level functional and structural changes to disease onset and progression. Combining imaging and omics data promotes the classification of AD into subtypes and paves the way for precision medicine solutions to prevent and treat AD.
Collapse
|
33
|
Bayer TA. N-Truncated Aβ Starting at Position Four-Biochemical Features, Preclinical Models, and Potential as Drug Target in Alzheimer's Disease. Front Aging Neurosci 2021; 13:710579. [PMID: 34489680 PMCID: PMC8417877 DOI: 10.3389/fnagi.2021.710579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/29/2021] [Indexed: 12/21/2022] Open
Abstract
The discussion of whether amyloid plaque Aβ is a valid drug target to fight Alzheimer’s disease (AD) has been a matter of scientific dispute for decades. This question can only be settled by successful clinical trials and the approval of disease-modifying drugs. However, many clinical trials with antibodies against different regions of the amyloid Aβ peptide have been discontinued, as they did not meet the clinical endpoints required. Recently, passive immunization of AD patients with Donanemab, an antibody directed against the N-terminus of pyroglutamate Aβ, showed beneficial effects in a phase II trial, supporting the concept that N-truncated Aβ is a relevant target for AD therapy. There is long-standing evidence that N-truncated Aβ variants are the main variants found in amyloid plaques besides full-length Aβ1–42, t, therefore their role in triggering AD pathology and as targets for drug development are of interest. While the contribution of pyroglutamate Aβ3–42 to AD pathology has been well studied in the past, the potential role of Aβ4–42 has been largely neglected. The present review will therefore focus on Aβ4–42 as a possible drug target based on human and mouse pathology, in vitro and in vivo toxicity, and anti-Aβ4-X therapeutic effects in preclinical models.
Collapse
Affiliation(s)
- Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
34
|
Liu Y, Hong H, Xue J, Luo J, Liu Q, Chen X, Pan Y, Zhou J, Liu Z, Chen T. Near-Infrared Radiation-Assisted Drug Delivery Nanoplatform to Realize Blood-Brain Barrier Crossing and Protection for Parkinsonian Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:37746-37760. [PMID: 34318658 DOI: 10.1021/acsami.1c12675] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Mitochondrial dysfunction, which is directly involved in Parkinson's disease (PD), is characterized by the production of reactive oxygen species (ROS) and aberrant energy metabolism. Thus, regulating mitochondrial function might be an effective strategy to treat PD. However, the blood-brain barrier (BBB) presents a significant challenge for the intracerebral delivery of drugs. Here, we synthesized a zeolitic imidazolate framework 8-coated Prussian blue nanocomposite (ZIF-8@PB), which was encapsulated with quercetin (QCT), a natural antioxidant, to treat PD. ZIF-8@PB-QCT exhibited superior near-infrared radiation (NIR) response and penetrated through the BBB to the site of mitochondrial damage guided by the photothermal effect. In the mice model of PD, the QCT released from ZIF-8@PB-QCT significantly increased the adenosine triphosphate levels, reduced the oxidative stress levels, and reversed dopaminergic neuronal damage as well as PD-related behavioral deficits without any damage to the normal tissues. Furthermore, we explored the underlying neuroprotective mechanism of ZIF-8@PB-QCT that was mediated by activating the PI3K/Akt signaling pathway. Thus, combined with noninvasive NIR radiation, the biocompatible ZIF-8@PB-QCT nanocomposite could be used to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yao Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Honghai Hong
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jincheng Xue
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Jingshan Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qiao Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingwei Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zeming Liu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
35
|
Oh KT, Kim D, Ye BS, Lee S, Yun M, Yoo SK. Segmentation of white matter hyperintensities on 18F-FDG PET/CT images with a generative adversarial network. Eur J Nucl Med Mol Imaging 2021; 48:3422-3431. [PMID: 33693968 DOI: 10.1007/s00259-021-05285-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/25/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE White matter hyperintensities (WMH) are typically segmented using MRI because WMH are hardly visible on 18F-FDG PET/CT. This retrospective study was conducted to segment WMH and estimate their volumes from 18F-FDG PET with a generative adversarial network (WhyperGAN). METHODS We selected patients whose interval between MRI and FDG PET/CT scans was within 3 months, from January 2017 to December 2018, and classified them into mild, moderate, and severe groups by following the semiquantitative rating method of Fazekas. For each group, 50 patients were selected, and of them, we randomly selected 35 patients for training and 15 for testing. WMH were automatically segmented from FLAIR MRI with manual adjustment. Patches of WMH were extracted from 18F-FDG PET and segmented MRI. WhyperGAN was compared with H-DenseUnet, a deep learning method widely used for segmentation tasks, for segmentation performance based on the dice similarity coefficient (DSC), recall, and average volume differences (AVD). For volume estimation, the predicted WMH volumes from PET were compared with ground truth volumes. RESULTS The DSC values were associated with WMH volumes on MRI. For volumes >60 mL, the DSC values were 0.751 for WhyperGAN and 0.564 for H-DenseUnet. For volumes ≤60 mL, the DSC values rapidly decreased as the volume decreased (0.362 for WhyperGAN vs. 0.237 for H-DenseUnet). For recall, WhyperGAN achieved the highest value in the severe group (0.579 for WhyperGAN vs. 0.509 for H-DenseUnet). For AVD, WhyperGAN achieved the lowest score in the severe group (0.494 for WhyperGAN vs. 0.941 for H-DenseUnet). For the WMH volume estimation, WhyperGAN performed better than H-DenseUnet and yielded excellent correlation coefficients (r = 0.998, 0.983, and 0.908 in the severe, moderate, and mild group). CONCLUSIONS Although limited by visual analysis, the WhyperGAN based can be used to automatically segment and estimate volumes of WMH from 18F-FDG PET/CT. This would increase the usefulness of 18F-FDG PET/CT for the evaluation of WMH in patients with cognitive impairment.
Collapse
Affiliation(s)
- Kyeong Taek Oh
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dongwoo Kim
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangwon Lee
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Sun Kook Yoo
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Hinteregger B, Loeffler T, Flunkert S, Neddens J, Bayer TA, Madl T, Hutter-Paier B. Metabolic, Phenotypic, and Neuropathological Characterization of the Tg4-42 Mouse Model for Alzheimer's Disease. J Alzheimers Dis 2021; 80:1151-1168. [PMID: 33646155 PMCID: PMC8150512 DOI: 10.3233/jad-201204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Preclinical Alzheimer's disease (AD) research strongly depends on transgenic mouse models that display major symptoms of the disease. Although several AD mouse models have been developed representing relevant pathologies, only a fraction of available mouse models, like the Tg4-42 mouse model, display hippocampal atrophy caused by the death of neurons as the key feature of AD. The Tg4-42 mouse model is therefore very valuable for use in preclinical research. Furthermore, metabolic biomarkers which have the potential to detect biochemical changes, are crucial to gain deeper insights into the pathways, the underlying pathological mechanisms and disease progression. OBJECTIVE We thus performed an in-depth characterization of Tg4-42 mice by using an integrated approach to analyze alterations of complex biological networks in this AD in vivo model. METHODS Therefore, untargeted NMR-based metabolomic phenotyping was combined with behavioral tests and immunohistological and biochemical analyses. RESULTS Our in vivo experiments demonstrate a loss of body weight increase in homozygous Tg4-42 mice over time as well as severe impaired learning behavior and memory deficits in the Morris water maze behavioral test. Furthermore, we found significantly altered metabolites in two different brain regions and metabolic changes of the glutamate/4-aminobutyrate-glutamine axis. Based on these results, downstream effects were analyzed showing increased Aβ42 levels, increased neuroinflammation as indicated by increased astro- and microgliosis as well as neuronal degeneration and neuronal loss in homozygous Tg4-42 mice. CONCLUSION Our study provides a comprehensive characterization of the Tg4-42 mouse model which could lead to a deeper understanding of pathological features of AD. Additionally this study reveals changes in metabolic biomarker which set the base for future preclinical studies or drug development.
Collapse
Affiliation(s)
- Barbara Hinteregger
- QPS Austria GmbH, Grambach, Austria
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | | | | | - Thomas A. Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center, Göttingen (UMG), Göttingen, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | |
Collapse
|
37
|
Miceli A, Cossu V, Marini C, Castellani P, Raffa S, Donegani MI, Bruno S, Ravera S, Emionite L, Orengo AM, Grillo F, Nobili F, Morbelli S, Uccelli A, Sambuceti G, Bauckneht M. 18F-Fluorodeoxyglucose Positron Emission Tomography Tracks the Heterogeneous Brain Susceptibility to the Hyperglycemia-Related Redox Stress. Int J Mol Sci 2020; 21:ijms21218154. [PMID: 33142766 PMCID: PMC7672601 DOI: 10.3390/ijms21218154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022] Open
Abstract
In cognitively normal patients, mild hyperglycemia selectively decreases 18F-Fluorodeoxyglucose (FDG) uptake in the posterior brain, reproducing Alzheimer disease pattern, hampering the diagnostic accuracy of this widely used tool. This phenomenon might involve either a heterogeneous response of glucose metabolism or a different sensitivity to hyperglycemia-related redox stress. Indeed, previous studies reported a close link between FDG uptake and activation of a specific pentose phosphate pathway (PPP), triggered by hexose-6P-dehydrogenase (H6PD) and contributing to fuel NADPH-dependent antioxidant responses in the endoplasmic reticulum (ER). To clarify this issue, dynamic positron emission tomography was performed in 40 BALB/c mice four weeks after administration of saline (n = 17) or 150 mg/kg streptozotocin (n = 23, STZ). Imaging data were compared with biochemical and histological indexes of glucose metabolism and redox balance. Cortical FDG uptake was homogeneous in controls, while it was selectively decreased in the posterior brain of STZ mice. This difference was independent of the activity of enzymes regulating glycolysis and cytosolic PPP, while it was paralleled by a decreased H6PD catalytic function and enhanced indexes of oxidative damage. Thus, the relative decrease in FDG uptake of the posterior brain reflects a lower activation of ER-PPP in response to hyperglycemia-related redox stress in these areas.
Collapse
Affiliation(s)
- Alberto Miceli
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
| | - Vanessa Cossu
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
| | - Cecilia Marini
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (A.M.O.)
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), 20090 Milano, Italy
| | | | - Stefano Raffa
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
| | - Maria Isabella Donegani
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova 16132, Italy; (S.B.); (S.R.)
| | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova 16132, Italy; (S.B.); (S.R.)
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (A.M.O.)
| | - Federica Grillo
- Department of Surgical Sciences and Integrated Diagnostics, Pathology Unit, University of Genoa, 16132 Genova, Italy;
| | - Flavio Nobili
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Center of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy; (F.N.); (A.U.)
- Clinical Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Morbelli
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (A.M.O.)
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Center of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy; (F.N.); (A.U.)
- Clinical Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Gianmario Sambuceti
- Department of Health Sciences, University of Genoa, 16132 Genova, Italy; (A.M.); (V.C.); (S.R.); (M.I.D.); (S.M.); (G.S.)
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (A.M.O.)
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (A.M.O.)
- Correspondence:
| |
Collapse
|
38
|
Kuhla A, Meuth L, Stenzel J, Lindner T, Lappe C, Kurth J, Krause BJ, Teipel S, Glass Ä, Kundt G, Vollmar B. Longitudinal [ 18F]FDG-PET/CT analysis of the glucose metabolism in ApoE-deficient mice. EJNMMI Res 2020; 10:119. [PMID: 33029684 PMCID: PMC7541807 DOI: 10.1186/s13550-020-00711-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/24/2020] [Indexed: 11/15/2022] Open
Abstract
Background Strong line of evidence suggests that the increased risk to develop AD may at least be partly mediated by cholesterol metabolism. A key regulator of cholesterol transport is the Apolipoprotein E4 (ApoE4), which plays a fundamental role in neuronal maintenance and repair. Impaired function of ApoE4 may contribute to altered cerebral metabolism leading to higher susceptibility to neurodegeneration. Methods To determine a possible link between ApoE function and alterations in AD in the brain of Apolipoprotein E-deficient mice (ApoE−/−) in a longitudinal manner metabolic and neurochemical parameters were analyzed. Cortical metabolism was measured by 2-deoxy-2-[18F]fluoroglucose ([18F]FDG)-PET/CT and proton magnetic resonance spectroscopy (1H-MRS) served to record neurochemical status. Results By using [18F]FDG-PET/CT, we showed that brain metabolism declined significantly stronger with age in ApoE−/− versus wild type (wt) mice. This difference was particularly evident at the age of 41 weeks in almost each analyzed brain region. In contrast, the 1H-MRS-measured N-acetylaspartate to creatine ratio, a marker of neuronal viability, did not decline with age and did not differ between ApoE−/− and wt mice. Conclusion In summary, this longitudinal in vivo study shows for the first time that ApoE−/− mice depict cerebral hypometabolism without neurochemical alterations.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany.
| | - Lou Meuth
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - Chris Lappe
- Institute of Diagnostic and Interventional Radiology, Pediatric and Neuroradiology, Rostock University Medical Center, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock, Greifswald, Germany
| | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd J Krause
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany.,Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Greifswald, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Änne Glass
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Guenther Kundt
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany.,Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
39
|
Franke TN, Irwin C, Bayer TA, Brenner W, Beindorff N, Bouter C, Bouter Y. In vivo Imaging With 18F-FDG- and 18F-Florbetaben-PET/MRI Detects Pathological Changes in the Brain of the Commonly Used 5XFAD Mouse Model of Alzheimer's Disease. Front Med (Lausanne) 2020; 7:529. [PMID: 33043029 PMCID: PMC7522218 DOI: 10.3389/fmed.2020.00529] [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: 04/20/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Imaging biomarkers of Alzheimer's disease (AD) that are able to detect molecular changes in vivo and transgenic animal models mimicking AD pathologies are essential for the evaluation of new therapeutic strategies. Positron-emission tomography (PET) using either 18F-Fluorodeoxyglucose (18F-FDG) or amyloid-tracers is a well-established, non-invasive tool in the clinical diagnostics of AD assessing two major pathological hallmarks. 18F-FDG-PET is able to detect early changes in cerebral glucose metabolism and amyloid-PET shows cerebral amyloid load. However, the suitability of 18F-FDG- and amyloid-PET in the widely used 5XFAD mouse model of AD is unclear as only a few studies on the use of PET biomarkers are available showing some conflicting results. The aim of this study was the evaluation of 18F-FDG-PET and amyloid-PET in 5XFAD mice in comparison to neurological deficits and neuropathological changes. Seven- and 12-month-old male 5XFAD mice showed a significant reduction in brain glucose metabolism in 18F-FDG-PET and amyloid-PET with 18F-Florbetaben demonstrated an increased cerebral amyloid deposition (n = 4-6 per group). Deficits in spatial reference memory were detected in 12-month-old 5XFAD mice in the Morris Water Maze (n = 10-12 per group). Furthermore, an increased plaque load and gliosis could be proven immunohistochemically in 5XFAD mice (n = 4-6 per group). PET biomarkers 18F-FDG and 18F-Florbetaben detected cerebral hypometabolism and increased plaque load even before the onset of severe memory deficits. Therefore, the 5XFAD mouse model of AD is well-suited for in vivo monitoring of AD pathologies and longitudinal testing of new therapeutic approaches.
Collapse
Affiliation(s)
- Timon N Franke
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Thomas A Bayer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
40
|
Bouter Y, Kacprowski T, Rößler F, Jensen LR, Kuss AW, Bayer TA. miRNA Alterations Elicit Pathways Involved in Memory Decline and Synaptic Function in the Hippocampus of Aged Tg4-42 Mice. Front Neurosci 2020; 14:580524. [PMID: 33013313 PMCID: PMC7511553 DOI: 10.3389/fnins.2020.580524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/18/2020] [Indexed: 12/30/2022] Open
Abstract
The transcriptome of non-coding RNA (ncRNA) species is increasingly focused in Alzheimer’s disease (AD) research. NcRNAs comprise, among others, transfer RNAs, long non-coding RNAs and microRNAs (miRs), each with their own specific biological function. We used smallRNASeq to assess miR expression in the hippocampus of young (3 month old) and aged (8 month old) Tg4-42 mice, a model system for sporadic AD, as well as age-matched wildtype controls. Tg4-42 mice express N-truncated Aβ4–42, develop age-related neuron loss, reduced neurogenesis and behavioral deficits. Our results do not only confirm known miR-AD associations in Tg4-42 mice, but more importantly pinpoint 22 additional miRs associated to the disease. Twenty-five miRs were differentially expressed in both aged Tg4-42 and aged wildtype mice while eight miRs were differentially expressed only in aged wildtype mice, and 33 only in aged Tg4-42 mice. No significant alteration in the miRNome was detected in young mice, which indicates that the changes observed in aged mice are down-stream effects of Aβ-induced pathology in the Tg4-42 mouse model for AD. Targets of those miRs were predicted using miRWalk. For miRs that were differentially expressed only in the Tg4-42 model, 128 targets could be identified, whereas 18 genes were targeted by miRs only differentially expressed in wildtype mice and 85 genes were targeted by miRs differentially expressed in both mouse models. Genes targeted by differentially expressed miRs in the Tg4-42 model were enriched for negative regulation of long-term synaptic potentiation, learning or memory, regulation of trans-synaptic signaling and modulation of chemical synaptic transmission obtained. This untargeted miR sequencing approach supports previous reports on the Tg4-42 mice as a valuable model for AD. Furthermore, it revealed miRs involved in AD, which can serve as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Tim Kacprowski
- Research Group Computational Systems Medicine, Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan (WZW), Technical University of Munich (TUM), Weihenstephan, Germany
| | - Fanny Rößler
- Research Group Computational Systems Medicine, Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan (WZW), Technical University of Munich (TUM), Weihenstephan, Germany
| | - Lars R Jensen
- Human Molecular Genetics Group, Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Andreas W Kuss
- Human Molecular Genetics Group, Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
41
|
Weerasekera A, Crabbé M, Tomé SO, Gsell W, Sima D, Casteels C, Dresselaers T, Deroose C, Van Huffel S, Rudolf Thal D, Van Damme P, Himmelreich U. Non-invasive characterization of amyotrophic lateral sclerosis in a hTDP-43 A315T mouse model: A PET-MR study. NEUROIMAGE-CLINICAL 2020; 27:102327. [PMID: 32653817 PMCID: PMC7352080 DOI: 10.1016/j.nicl.2020.102327] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 05/02/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
Abstract
Currently TAR DNA binding protein 43 (TDP-43) pathology, underlying Amyotrophic Lateral Sclerosis (ALS), remains poorly understood which hinders both clinical diagnosis and drug discovery efforts. To better comprehend the disease pathophysiology, positron emission tomography (PET) and multi-parametric magnetic resonance imaging (mp-MRI) provide a non-invasive mode to investigate molecular, structural, and neurochemical abnormalities in vivo. For the first time, we report the findings of a longitudinal PET-MR study in the TDP-43A315T ALS mouse model, investigating disease-related changes in the mouse brain. 2-deoxy-2-[18F]fluoro-D-glucose [18F]FDG PET showed significantly lowered glucose metabolism in the motor and somatosensory cortices of TDP-43A315T mice whereas metabolism was elevated in the region covering the bilateral substantia nigra, reticular and amygdaloid nucleus between 3 and 7 months of age, as compared to non-transgenic controls. MR spectroscopy data showed significant changes in glutamate + glutamine (Glx) and choline levels in the motor cortex and hindbrain of TDP-43A315T mice compared to controls. Cerebral blood flow (CBF) measurements, using an arterial spin labelling approach, showed no significant age- or group-dependent changes in brain perfusion. Diffusion MRI indices demonstrated transient changes in different motor areas of the brain in TDP-43A315T mice around 14 months of age. Cytoplasmic TDP-43 proteinaceous inclusions were observed in the brains of symptomatic, 18-month-old mice, but not in non-symptomatic transgenic or wild-type mice. Our results reveal that disease- and age-related functional and neurochemical alterations, together with limited structural changes, occur in specific brain regions of transgenic TDP-43A315T mice, as compared to their healthy counterparts. Altogether these findings shed new light on TDP-43A315T disease pathogenesis and may prove useful for clinical management of ALS.
Collapse
Affiliation(s)
- Akila Weerasekera
- Biomedical MRI Unit/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium; A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School (MGH/HMS), Boston, MA, USA
| | - Melissa Crabbé
- Division of Nuclear Medicine, Department of Imaging and Pathology, KU Leuven, Belgium; MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium.
| | - Sandra O Tomé
- Laboratory for Neuropathology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI Unit/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Diana Sima
- Icometrix, R&D department, Leuven, Belgium; Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, KU Leuven, Leuven, Belgium
| | - Cindy Casteels
- Division of Nuclear Medicine, Department of Imaging and Pathology, KU Leuven, Belgium; MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium
| | - Tom Dresselaers
- Division of Radiology, Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Christophe Deroose
- Division of Nuclear Medicine, Department of Imaging and Pathology, KU Leuven, Belgium; MoSAIC - Molecular Small Animal Imaging Centre, KU Leuven, Leuven, Belgium
| | - Sabine Van Huffel
- Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, KU Leuven, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Philip Van Damme
- Laboratory of Neurobiology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Center for Brain & Disease Research, VIB, Leuven, Belgium; Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI Unit/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Hinteregger B, Loeffler T, Flunkert S, Neddens J, Birner-Gruenberger R, Bayer TA, Madl T, Hutter-Paier B. Transgene integration causes RARB downregulation in homozygous Tg4-42 mice. Sci Rep 2020; 10:6377. [PMID: 32286473 PMCID: PMC7156671 DOI: 10.1038/s41598-020-63512-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease can be modelled by different transgenic mouse strains. To gain deeper insight into disease model mechanisms, the previously described Tg4-42 mouse was analysed for transgene integration. On RNA/DNA level the transgene integration resulted in more than 20 copy numbers and further caused a deletion of exon 2 of the retinoic acid receptor beta. These findings were also confirmed on protein level with highly decreased retinoic acid receptor beta protein levels in homozygous Tg4-42 mice and may have an impact on the previously described phenotype of homozygous Tg4-42 mice to be solely dependent on amyloid-ß 4-42 expression. Since hemizygous mice show no changes in RARB protein levels it can be concluded that the previously described phenotype of these mice should not be affected by the retinoic acid receptor beta gene knockout. In order to fully understand the results of transgenesis, it is extremely advisable to determine the genome integration site and the basic structure of the inserted transgenes. This can be carried out for instance by next-generation sequencing techniques. Our results thus suggest that a detailed characterization of new disease models using the latest genomics technologies prior to functional studies could be a valuable tool to avoid an unexpected genetic influence on the animals' phenotype that is not only based on the inserted transgene. This would also significantly improve the selection of mouse models that are best suited for therapeutic development and basic research.
Collapse
Affiliation(s)
- Barbara Hinteregger
- QPS Austria GmbH, Parkring 12, 8074, Grambach, Austria
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging) Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Tina Loeffler
- QPS Austria GmbH, Parkring 12, 8074, Grambach, Austria
| | | | - Joerg Neddens
- QPS Austria GmbH, Parkring 12, 8074, Grambach, Austria
| | - Ruth Birner-Gruenberger
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging) Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- Diagnostic and Research Institute of Pathology & Omics Center Graz, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Vienna University of Technology, Institute of Chemical Technologies and Analytics, Vienna, Austria
| | - Thomas A Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging) Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | |
Collapse
|
43
|
Wirths O, Zampar S. Emerging roles of N- and C-terminally truncated Aβ species in Alzheimer’s disease. Expert Opin Ther Targets 2019; 23:991-1004. [DOI: 10.1080/14728222.2019.1702972] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Oliver Wirths
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Silvia Zampar
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
44
|
Sichler ME, Löw MJ, Schleicher EM, Bayer TA, Bouter Y. Reduced Acoustic Startle Response and Prepulse Inhibition in the Tg4-42 Model of Alzheimer's Disease. J Alzheimers Dis Rep 2019; 3:269-278. [PMID: 31867566 PMCID: PMC6918877 DOI: 10.3233/adr-190132] [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] [Indexed: 12/19/2022] Open
Abstract
Sensorimotor deficits have been described in several neuropsychiatric disorders including Alzheimer’s disease. The aim of the present study was to evaluate possible sensorimotor gating deficits in the Tg4-42 mouse model of Alzheimer’s disease using the prepulse inhibition task (PPI). Previous studies indicated that the hippocampus is essentially involved in the regulation of PPI. We analyzed 7-month-old homozygous Tg4-42 mice as mice at this age display severe neuron loss especially in the CA1 region of the hippocampus. Our results revealed a reduced startle response and PPI in Tg4-42 mice. The observed deficits in startle response and PPI are likely due to altered sensory processing abilities rather than hearing deficits as Tg4-42 displayed intact hearing in the fear conditioning task. The present study demonstrates for the first time that sensorimotor gating is impaired in Tg4-42 mice. Analyzing startle response as well as the PPI may offer valuable measurements to assess the efficacy of therapeutic strategies in the future in this Alzheimer’s disease model.
Collapse
Affiliation(s)
- Marius E Sichler
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Maximilian J Löw
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Eva M Schleicher
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Goettingen, Germany
| |
Collapse
|
45
|
Iozzo P, Guzzardi MA. Imaging of brain glucose uptake by PET in obesity and cognitive dysfunction: life-course perspective. Endocr Connect 2019; 8:R169-R183. [PMID: 31590145 PMCID: PMC6865363 DOI: 10.1530/ec-19-0348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The prevalence of obesity has reached epidemic proportions and keeps growing. Obesity seems implicated in the pathogenesis of cognitive dysfunction, Alzheimer's disease and dementia, and vice versa. Growing scientific efforts are being devoted to the identification of central mechanisms underlying the frequent association between obesity and cognitive dysfunction. Glucose brain handling undergoes dynamic changes during the life-course, suggesting that its alterations might precede and contribute to degenerative changes or signaling abnormalities. Imaging of the glucose analog 18F-labeled fluorodeoxyglucose (18FDG) by positron emission tomography (PET) is the gold-standard for the assessment of cerebral glucose metabolism in vivo. This review summarizes the current literature addressing brain glucose uptake measured by PET imaging, and the effect of insulin on brain metabolism, trying to embrace a life-course vision in the identification of patterns that may explain (and contribute to) the frequent association between obesity and cognitive dysfunction. The current evidence supports that brain hypermetabolism and brain insulin resistance occur in selected high-risk conditions as a transient phenomenon, eventually evolving toward normal or low values during life or disease progression. Associative studies suggest that brain hypermetabolism predicts low BDNF levels, hepatic and whole body insulin resistance, food desire and an unfavorable balance between anticipated reward from food and cognitive inhibitory control. Emerging mechanistic links involve the microbiota and the metabolome, which correlate with brain metabolism and cognition, deserving attention as potential future prevention targets.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
- Correspondence should be addressed to P Iozzo:
| | | |
Collapse
|
46
|
Wagner JM, Sichler ME, Schleicher EM, Franke TN, Irwin C, Löw MJ, Beindorff N, Bouter C, Bayer TA, Bouter Y. Analysis of Motor Function in the Tg4-42 Mouse Model of Alzheimer's Disease. Front Behav Neurosci 2019; 13:107. [PMID: 31156407 PMCID: PMC6533559 DOI: 10.3389/fnbeh.2019.00107] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia. Hallmarks of AD are memory impairments and cognitive deficits, but non-cognitive impairments, especially motor dysfunctions are also associated with the disease and may even precede classic clinical symptoms. With an aging society and increasing hospitalization of the elderly, motor deficits are of major interest to improve independent activities in daily living. Consistent with clinical findings, a variety of AD mouse models develop motor deficits as well. We investigated the motor function of 3- and 7-month-old Tg4-42 mice in comparison to wild-type controls and 5XFAD mice and discuss the results in context with several other AD mouse model. Our study shows impaired balance and motor coordination in aged Tg4-42 mice in the balance beam and rotarod test, while general locomotor activity and muscle strength is not impaired at 7 months. The cerebellum is a major player in the regulation and coordination of balance and locomotion through practice. Particularly, the rotarod test is able to detect cerebellar deficits. Furthermore, supposed cerebellar impairment was verified by 18F-FDG PET/MRI. Aged Tg4-42 mice showed reduced cerebellar glucose metabolism in the 18F-FDG PET. Suggesting that, deficits in coordination and balance are most likely due to cerebellar impairment. In conclusion, Tg4-42 mice develop motor deficits before memory deficits, without confounding memory test. Thus, making the Tg4-42 mouse model a good model to study the effects on cognitive decline of therapies targeting motor impairments.
Collapse
Affiliation(s)
- Jannek M. Wagner
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Marius E. Sichler
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Eva M. Schleicher
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Timon N. Franke
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Maximilian Johannes Löw
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center, Charité – University Medicine Berlin, Berlin, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Thomas A. Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
47
|
Bouter C, Bouter Y. 18F-FDG-PET in Mouse Models of Alzheimer's Disease. Front Med (Lausanne) 2019; 6:71. [PMID: 31058151 PMCID: PMC6482246 DOI: 10.3389/fmed.2019.00071] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/21/2019] [Indexed: 01/08/2023] Open
Abstract
Suitable animal models and in vivo biomarkers are essential for development and evaluation of new therapeutic strategies in Alzheimer's disease (AD). 18F-Fluorodeoxyglucose (18F-FDG)-positron-emission tomography (PET) is an imaging biomarker that allows the assessment of cerebral glucose metabolism in vivo. While 18F-FDG-PET/CT is an established tool in the evaluation of AD patients, its role in preclinical studies with AD mouse models remains unclear. Here, we want to review available studies on 18F-FDG-PET/CT in AD mouse models in order to evaluate the method and its impact in preclinical AD research. Only a limited number of studies using 18F-FDG-PET in AD mice were carried out so far showing contradictory findings in cerebral FDG uptake. Methodological differences as well as underlying pathological features of used mouse models seem to be accountable for those varying results. However, 18F-FDG-PET can be a valuable tool in longitudinal in vivo therapy monitoring with a lot of potential for future studies.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
48
|
Nanowired delivery of cerebrolysin with neprilysin and p-Tau antibodies induces superior neuroprotection in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2019; 245:145-200. [DOI: 10.1016/bs.pbr.2019.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|