1
|
Vockert N, Machts J, Kleineidam L, Nemali A, Incesoy EI, Bernal J, Schütze H, Yakupov R, Peters O, Gref D, Schneider LS, Preis L, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Rostamzadeh A, Glanz W, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Spottke A, Roy N, Heneka MT, Brosseron F, Wagner M, Wolfsgruber S, Dobisch L, Dechent P, Hetzer S, Scheffler K, Zeidman P, Stern Y, Schott BH, Jessen F, Düzel E, Maass A, Ziegler G. Cognitive reserve against Alzheimer's pathology is linked to brain activity during memory formation. Nat Commun 2024; 15:9815. [PMID: 39537609 DOI: 10.1038/s41467-024-53360-9] [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: 10/30/2023] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The cognitive reserve (CR) hypothesis posits that individuals can differ in how their brain function is disrupted by pathology associated with aging and neurodegeneration. Here, we test this hypothesis in the continuum from cognitively normal to at-risk stages for Alzheimer's Disease (AD) to AD dementia using longitudinal data from 490 participants of the DELCODE multicentric observational study. Brain function is measured using task fMRI of visual memory encoding. Using a multivariate moderation analysis, we identify a CR-related activity pattern underlying successful memory encoding that moderates the detrimental effect of AD pathological load on cognitive performance. CR is mainly represented by a more pronounced expression of the task-active network encompassing deactivation of the default mode network (DMN) and activation of inferior temporal regions including the fusiform gyrus. We devise personalized fMRI-based CR scores that moderate the impact of AD pathology on cognitive performance and are positively associated with years of education. Furthermore, higher CR scores attenuate the effect of AD pathology on cognitive decline over time. Our findings primarily provide evidence for the maintenance of core cognitive circuits including the DMN as the neural basis of CR. Individual brain activity levels of these areas during memory encoding have prognostic value for future cognitive decline.
Collapse
Affiliation(s)
- Niklas Vockert
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Judith Machts
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Aditya Nemali
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Enise I Incesoy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Hartmut Schütze
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Daria Gref
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Luisa Sophie Schneider
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, ECRC Experimental and Clinical Research Center, Berlin, Germany
| | - Lukas Preis
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
- School of Medicine, Technical University of Munich, Department of Psychiatry and Psychotherapy, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Ayda Rostamzadeh
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg, Luxembourg
| | | | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Goettingen, Goettingen, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tuebingen, Tuebingen, Germany
| | - Peter Zeidman
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, London, UK
| | - Yaakov Stern
- Cognitive Neuroscience Division, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, University of Cologne, Koeln, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Koeln, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
2
|
Studart- A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [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: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
3
|
Vaiasicca S, Balietti M, Bevilacqua L, Giorgetti B, Casoli T. Convergence between brain aging and Alzheimer's disease: focus on mitochondria. Mech Ageing Dev 2024:112001. [PMID: 39490933 DOI: 10.1016/j.mad.2024.112001] [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: 09/03/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) accounts for the majority of dementia cases, with aging being the primary risk factor for developing this neurodegenerative condition. Aging and AD share several characteristics, including the formation of amyloid plaques and neurofibrillary tangles, synaptic loss, and neuroinflammation. This overlap suggests that mechanisms driving the aging process might also promote AD; however, the underlying processes are not yet fully understood. In this narrative review, we will focus on the role of mitochondria, not only as the "powerhouse of the cell", but also in programmed cell death, immune response, macromolecular synthesis, and calcium regulation. We will explore both the common changes between aging and AD and the differences between them. Additionally, we will provide an overview of interventions aimed at maintaining mitochondrial function in an attempt to slow the progression of AD. This will include a discussion of antioxidant molecules, factors that trigger mitochondrial biogenesis, compounds capable of restoring the fission/fusion balance, and a particular focus on recent techniques for mitochondrial DNA gene therapy.
Collapse
Affiliation(s)
| | - Marta Balietti
- Center for Neurobiology of Aging, IRCCS INRCA, 60121 Ancona, Italy.
| | - Lisa Bevilacqua
- Center for Neurobiology of Aging, IRCCS INRCA, 60121 Ancona, Italy
| | | | - Tiziana Casoli
- Center for Neurobiology of Aging, IRCCS INRCA, 60121 Ancona, Italy
| |
Collapse
|
4
|
Arbizu J, Morbelli S, Minoshima S, Barthel H, Kuo P, Van Weehaeghe D, Horner N, Colletti PM, Guedj E. SNMMI Procedure Standard/EANM Practice Guideline for Brain [ 18F]FDG PET Imaging, Version 2.0. J Nucl Med 2024:jnumed.124.268754. [PMID: 39419552 DOI: 10.2967/jnumed.124.268754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
PREAMBLEThe Society of Nuclear Medicine and Molecular Imaging (SNMMI) is an international scientific and professional organization founded in 1954 to promote the science, technology, and practical application of nuclear medicine. The European Association of Nuclear Medicine (EANM) is a professional nonprofit medical association that facilitates communication worldwide between individuals pursuing clinical and research excellence in nuclear medicine. The EANM was founded in 1985. The EANM was founded in 1985. SNMMI and EANM members are physicians, technologists, and scientists specializing in the research and practice of nuclear medicine.The SNMMI and EANM will periodically define new guidelines for nuclear medicine practice to help advance the science of nuclear medicine and to improve the quality of service to patients throughout the world. Existing practice guidelines will be reviewed for revision or renewal, as appropriate, on their fifth anniversary or sooner, if indicated.Each practice guideline, representing a policy statement by the SNMMI/EANM, has undergone a thorough consensus process in which it has been subjected to extensive review. The SNMMI and EANM recognize that the safe and effective use of diagnostic nuclear medicine imaging requires specific training, skills, and techniques, as described in each document. Reproduction or modification of the published practice guideline by those entities not providing these services is not authorized.These guidelines are an educational tool designed to assist practitioners in providing appropriate care for patients. They are not inflexible rules or requirements of practice and are not intended, nor should they be used, to establish a legal standard of care. For these reasons and those set forth below, both the SNMMI and the EANM caution against the use of these guidelines in litigation in which the clinical decisions of a practitioner are called into question.The ultimate judgment regarding the propriety of any specific procedure or course of action must be made by the physician or medical physicist in light of all the circumstances presented. Thus, there is no implication that an approach differing from the guidelines, standing alone, is below the standard of care. To the contrary, a conscientious practitioner may responsibly adopt a course of action different from that set forth in the guidelines when, in the reasonable judgment of the practitioner, such course of action is indicated by the condition of the patient, limitations of available resources, or advances in knowledge or technology subsequent to publication of the guidelines.The practice of medicine includes both the art and the science of the prevention, diagnosis, alleviation, and treatment of disease. The variety and complexity of human conditions make it impossible to always reach the most appropriate diagnosis or to predict with certainty a particular response to treatment.Therefore, it should be recognized that adherence to these guidelines will not ensure an accurate diagnosis or a successful outcome. All that should be expected is that the practitioner will follow a reasonable course of action based on current knowledge, available resources, and the needs of the patient to deliver effective and safe medical care. The sole purpose of these guidelines is to assist practitioners in achieving this objective.
Collapse
Affiliation(s)
- Javier Arbizu
- Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain;
| | - Silvia Morbelli
- Nuclear Medicine Unit, Citta'della Scenza e della Salute di Torino, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University Medical Centre, Leipzig, Germany
| | | | | | - Neil Horner
- Atlantic Health System, Morristown, New Jersey, and Icahn School of Medicine at Mount Sinai, New York, New York
| | - Patrick M Colletti
- Department of Radiology and Nuclear Medicine, University of Southern California, Los Angeles, California; and
| | - Eric Guedj
- APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix Marseille University, Marseille, France
| |
Collapse
|
5
|
Jagust WJ, Koeppe RA, Rabinovici GD, Villemagne VL, Harrison TM, Landau SM. The ADNI PET Core at 20. Alzheimers Dement 2024; 20:7340-7349. [PMID: 39108002 PMCID: PMC11485322 DOI: 10.1002/alz.14165] [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: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) PET Core has evolved over time, beginning with positron emission tomography (PET) imaging of a subsample of participants with [18F]fluorodeoxyglucose (FDG)-PET, adding tracers for measurement of β-amyloid, followed by tau tracers. This review examines the evolution of the ADNI PET Core, the novel aspects of PET imaging in each stage of ADNI, and gives an accounting of PET images available in the ADNI database. The ADNI PET Core has been and continues to be a rich resource that provides quantitative PET data and preprocessed PET images to the scientific community, allowing interrogation of both basic and clinically relevant questions. By standardizing methods across different PET scanners and multiple PET tracers, the Core has demonstrated the feasibility of large-scale, multi-center PET studies. Data managed and disseminated by the PET Core has been critical to defining pathophysiological models of Alzheimer's disease (AD) and helped to drive methods used in modern therapeutic trials. HIGHLIGHTS: The ADNI PET Core began with FDG-PET and now includes three amyloid and three tau PET ligands. The PET Core has standardized acquisition and analysis of multitracer PET images. The ADNI PET Core helped to develop methods that have facilitated clinical trials in AD.
Collapse
Affiliation(s)
- William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Robert A. Koeppe
- Department of RadiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | | | - Susan M. Landau
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | |
Collapse
|
6
|
De Keersmaecker S, De Meyer S, Vandenberghe R. Non-Alzheimer's amnestic mild cognitive impairment with medial temporal hypometabolism. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70018. [PMID: 39445341 PMCID: PMC11497174 DOI: 10.1002/dad2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION The increasing use of Alzheimer's disease (AD) biomarkers has led to the recognition of a subgroup of non-AD amnestic mild cognitive impairment (aMCI) patients who have medial temporal hypometabolism on fluorodeoxyglucose-positron emission tomography (FDG-PET). METHODS In this academic memory-clinic-based consecutive series, 16 non-AD aMCI patients and 28 AD controls matched for sex, age, and baseline Mini-Mental State Examination (MMSE) were followed for a median duration of 4.5 years. Our primary outcome was the MMSE decline rate over the subsequent years. We also determined the final diagnosis over time. RESULTS FDG-PET showed more pronounced medial temporal hypometabolism in non-AD cases and more inferior parietal lobule hypometabolism in AD controls. MMSE decline was slower in non-AD (β = -0.51) than in AD (β = -2.00) patients. Five non-AD cases developed frontotemporal dementia years after symptom onset, and one developed dementia with Lewy bodies. DISCUSSION Non-AD aMCI patients with medial temporal hypometabolism show slower cognitive decline. Highlights Non-AD aMCI with medial temporal hypometabolism shows slower cognitive decline than AD.FDG-PET revealed distinct metabolic patterns between non-AD aMCI and AD patients.Approximately one-third of non-AD aMCI cases developed frontotemporal dementia.Comprehensive diagnostic biomarkers are crucial for non-AD aMCI characterization.
Collapse
|
7
|
Salmon E, Collette F, Bastin C. Cerebral glucose metabolism in Alzheimer's disease. Cortex 2024; 179:50-61. [PMID: 39141935 DOI: 10.1016/j.cortex.2024.07.004] [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: 05/01/2024] [Revised: 07/05/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
18F-fluoro-deoxy-glucose positron emission tomography (FDG-PET) is a useful paraclinical exam for the diagnosis of Alzheimer's disease (AD). In this narrative review, we report seminal studies in clinically probable AD that have shown the importance of posterior brain metabolic decrease and the paradoxical variability of the hippocampal metabolism. The FDG-PET pattern was a sensitive indicator of AD in pathologically confirmed cases and it was used for differential diagnosis of dementia conditions. In prodromal AD, the AD FDG-PET pattern was observed in converters and predicted conversion. Automated data analysis techniques provided variable accuracy according to the reported indices and machine learning methods showed variable reliability of results. FDG-PET could confirm AD clinical heterogeneity and image data driven analyses identified hypometabolic subtypes with variable involvement of the hippocampus, reminiscent if the paradoxical FDG uptake. In studies dedicated to clinical and metabolic correlations, episodic memory was related to metabolism in the default mode network (and Papez's circuit) in prodromal and mild AD stages, and specific cognitive processes were associated to precisely distributed brain metabolism. Cerebral metabolic correlates of anosognosia could also be related to current neuropsychological models. AD FDG-PET pattern was reported in preclinical AD stages and related to cognition or to conversion to mild cognitive impairment (MCI). Using other biomarkers, the AD FDG-PET pattern was confirmed in AD participants with positive PET-amyloid. Intriguing observations reported increased metabolism related to brain amyloid and/or tau deposition. Preserved glucose metabolism sometimes appear as a compensation, but it was frequently detrimental and the nature of such a preservation of glucose metabolism remains an open question. Limbic metabolic involvement was frequently related to non-AD biomarkers profile and clinical stability, and it was reported in non-AD pathologies, such as the limbic predominant age-related encephalopathy (LATE). FDG-PET abnormalities observed in the absence of classical AD proteinopathies can be useful to search for pathological mechanisms and differential diagnosis of AD.
Collapse
Affiliation(s)
- Eric Salmon
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| | - Fabienne Collette
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| | - Christine Bastin
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| |
Collapse
|
8
|
Heyer S, Simon M, Doyen M, Mortada A, Roch V, Jeanbert E, Thilly N, Malaplate C, Kearney-Schwartz A, Jonveaux T, Bannay A, Verger A. 18F-FDG PET can effectively rule out conversion to dementia and the presence of CSF biomarker of neurodegeneration: a real-world data analysis. Alzheimers Res Ther 2024; 16:182. [PMID: 39135067 PMCID: PMC11320856 DOI: 10.1186/s13195-024-01535-3] [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: 03/05/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Precisely defining the delay in onset of dementia is a particular challenge for early diagnosis. Brain [18F] fluoro-2-deoxy-2-D-glucose (18F-FDG) Positron Emission Tomography (PET) is a particularly interesting tool for the early diagnosis of neurodegenerative diseases, through the measurement of the cerebral glucose metabolic rate. There is currently a lack of longitudinal studies under real-life conditions, with sufficient patients, to accurately evaluate the predictive values of brain 18F-FDG PET scans. Here, we aimed to estimate the value of brain 18F-FDG PET for predicting the risk of dementia conversion and the risk of occurrence of a neurodegenerative pathology. METHODS Longitudinal data for a cohort of patients with no diagnosis of dementia at the time of recruitment referred by a tertiary memory clinic for brain 18F-FDG PET were matched with (Prince M, Wimo A, Guerchet Maëlenn, Ali G-C, Wu Y-T et al. World Alzheimer Report 2015. The Global Impact of Dementia: An analysis of prevalence, incidence, cost and trends. [Research Report] Alzheimer's Disease International. 2015. 2015.) data from the French National Health Data System (NHDS), (Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14(4):535-62.) data from the National Alzheimer Bank (NAB), and (Davis M, O`Connell T, Johnson S, Cline S, Merikle E, Martenyi F, et al. Estimating Alzheimer's Disease Progression Rates from Normal Cognition Through Mild Cognitive Impairment and Stages of Dementia. CAR. 2018;15(8):777-88.) lumbar puncture (LP) biomarker data. The criteria for dementia conversion were the designation, within the three years after the brain 18F-FDG PET scan, of a long-term condition for dementia in the NHDS and a dementia stage of cognitive impairment in the NAB. The criterion for the identification of a neurodegenerative disease in the medical records was the determination of LP biomarker levels. RESULTS Among the 403 patients (69.9 ± 11.4 years old, 177 women) from the initial cohort with data matched with the NHDS data, 137 were matched with the NAB data, and 61 were matched with LP biomarker data. Within three years of the scan, a 18F-FDG PET had negative predictive values of 85% for dementia conversion (according to the NHDS and NAB datasets) and 95% for the presence of LP neurodegeneration biomarkers. CONCLUSION A normal brain 18F-FDG PET scan can help rule out the risk of dementia conversion and the presence of cerebrospinal fluid (CSF) biomarker of neurodegeneration early with high certainty, allowing modifications to patient management regimens in the short term. TRIAL REGISTRATION Clinical Trials database (NCT04804722). March 18, 2021. Retrospectively registered.
Collapse
Affiliation(s)
- Sébastien Heyer
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, Université de Lorraine, CHRU Nancy, Nancy, F-54000, France
| | - Maïa Simon
- Department of Methodology, Promotion and Investigation, Université de Lorraine, CHRU-Nancy, Nancy, F-54000, France
| | - Matthieu Doyen
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, Université de Lorraine, CHRU Nancy, Nancy, F-54000, France
- Université de Lorraine, IADI, INSERM U1254, Nancy, F-54000, France
| | - Ali Mortada
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, Université de Lorraine, CHRU Nancy, Nancy, F-54000, France
| | - Véronique Roch
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, Université de Lorraine, CHRU Nancy, Nancy, F-54000, France
| | - Elodie Jeanbert
- Department of Methodology, Promotion and Investigation, Université de Lorraine, CHRU-Nancy, Nancy, F-54000, France
| | - Nathalie Thilly
- Department of Methodology, Promotion and Investigation, Université de Lorraine, CHRU-Nancy, Nancy, F-54000, France
| | - Catherine Malaplate
- Department of Biochemistry, Université de Lorraine, CHRU-Nancy, Nancy, F-54000, France
| | - Anna Kearney-Schwartz
- Department of Geriatrics, Université de Lorraine, CHRU-Nancy, Nancy, F-54000, France
- CMRR, University Hospital Nancy, Nancy, F-54000, France
| | - Thérèse Jonveaux
- CMRR, University Hospital Nancy, Nancy, F-54000, France
- Department of Neurology, University Hospital Nancy, Nancy, F-54000, France
| | - Aurélie Bannay
- Medical Assessment and Information Department, Université de Lorraine, CHRU-Nancy, Nancy, 54000, France
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, Université de Lorraine, CHRU Nancy, Nancy, F-54000, France.
- Université de Lorraine, IADI, INSERM U1254, Nancy, F-54000, France.
| |
Collapse
|
9
|
Bougea A, Gourzis P. Biomarker-Based Precision Therapy for Alzheimer's Disease: Multidimensional Evidence Leading a New Breakthrough in Personalized Medicine. J Clin Med 2024; 13:4661. [PMID: 39200803 PMCID: PMC11355840 DOI: 10.3390/jcm13164661] [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: 06/22/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
(1) Background: Alzheimer's disease (AD) is a worldwide neurodegenerative disorder characterized by the buildup of abnormal proteins in the central nervous system and cognitive decline. Since no radical therapy exists, only symptomatic treatments alleviate symptoms temporarily. In this review, we will explore the latest advancements in precision medicine and biomarkers for AD, including their potential to revolutionize the way we diagnose and treat this devastating condition. (2) Methods: A literature search was performed combining the following Medical Subject Heading (MeSH) terms on PubMed: "Alzheimer's disease", "biomarkers", "APOE", "APP", "GWAS", "cerebrospinal fluid", "polygenic risk score", "Aβ42", "τP-181", " p-tau217", "ptau231", "proteomics", "total tau protein", and "precision medicine" using Boolean operators. (3) Results: Genome-wide association studies (GWAS) have identified numerous genetic variants associated with AD risk, while a transcriptomic analysis has revealed dysregulated gene expression patterns in the brains of individuals with AD. The proteomic and metabolomic profiling of biological fluids, such as blood, urine, and CSF, and neuroimaging biomarkers have also yielded potential biomarkers of AD that could be used for the early diagnosis and monitoring of disease progression. (4) Conclusion: By leveraging a combination of the above biomarkers, novel ultrasensitive immunoassays, mass spectrometry methods, and metabolomics, researchers are making significant strides towards personalized healthcare for individuals with AD.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Philippos Gourzis
- 1st Department of Psychiatry, University of Patras, 26504 Rio, Greece;
| |
Collapse
|
10
|
Womack CL, Perkins A, Arnold JM. Cognitive Impairment in the Primary Care Clinic. Prim Care 2024; 51:233-251. [PMID: 38692772 DOI: 10.1016/j.pop.2024.02.010] [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: 05/03/2024]
Abstract
Cognitive impairment is a common problem in the geriatric population and is characterized by variable symptoms of memory difficulties, executive dysfunction, language or visuospatial problems, and behavioral changes. It is imperative that primary care clinicians recognize and differentiate the variable symptoms associated with cognitive impairment from changes attributable to normal aging or secondary to other medical conditions. A thorough evaluation for potentially reversible causes of dementia is required before diagnosis with a neurodegenerative dementia. Other abnormal neurologic findings, rapid progression, or early age of onset are red flags that merit referral to neurology for more specialized evaluation and treatment.
Collapse
Affiliation(s)
- Cindy L Womack
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA
| | - Andrea Perkins
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA
| | - Jennifer M Arnold
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA.
| |
Collapse
|
11
|
Duman Sastim D, Elboga G, Elboga U, Gungor K. Evaluation of the relationship between FDG-PET hypometabolism and retinal layer thickness in patients with Alzheimer's disease. Acta Neurol Belg 2024; 124:987-993. [PMID: 38546932 DOI: 10.1007/s13760-024-02511-2] [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: 04/10/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024]
Abstract
We aimed to investigate the diagnostic value of Optical coherence tomography (OCT) in Alzheimer's disease (AD) and to assess the correlation between OCT and fluorodeoxyglucose (FDG)-positron emission tomography (PET) which shows high diagnostic agreement with findings from postmortem histopathology-the gold standard method. Patients who were diagnosed with AD-related dementia were selected for the study. Patients with a mini mental test (MMT) score between 18 and 23 were included in the study (n = 31). Volunteers with MMT ≥ 28 and no cognitive impairment were included in the study as the control group (n = 31). OCT imaging was performed in the patient and control groups after detailed ophthalmological examinations including visual acuity and intraocular pressure measurements. Brain glucose metabolism measurement was performed using 18 F-FDG PET/computed tomography. When adjusted for age and sex, mean retinal nerve fiber layer thickness (RNFL) thickness showed a significant difference between groups and the RNFL thickness in the superior temporal and superior nasal quadrants in AD-related mild dementia group showed a significant difference (p < 0.05). Furthermore, only the RNFL thickness in the inferior nasal quadrant of the right eye showed a significant difference between the groups (p = 0.016). It is thought that OCT is a promising imaging method in the elderly population due to its low-cost, non-invasive and easily applicability, and therefore, it may contribute in the future as a tool in the periodic follow-up of patients diagnosed with AD.
Collapse
Affiliation(s)
- Demet Duman Sastim
- Department of Psychiatry, Tunceli State Hospital, Merkez, 62000, Merkez/Tunceli, Turkey.
| | - Gulcin Elboga
- Faculty of Medicine, Department of Psychiatry, Gaziantep University, Gaziantep, Turkey
| | - Umut Elboga
- Faculty of Medicine, Department of Nuclear Medicine, Gaziantep University, Gaziantep, Turkey
| | - Kivanc Gungor
- Faculty of Medicine, Department of Ophthalmology, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
12
|
Lopez-Lee C, Torres ERS, Carling G, Gan L. Mechanisms of sex differences in Alzheimer's disease. Neuron 2024; 112:1208-1221. [PMID: 38402606 PMCID: PMC11076015 DOI: 10.1016/j.neuron.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Alzheimer's disease (AD) and the mechanisms underlying its etiology and progression are complex and multifactorial. The higher AD risk in women may serve as a clue to better understand these complicated processes. In this review, we examine aspects of AD that demonstrate sex-dependent effects and delve into the potential biological mechanisms responsible, compiling findings from advanced technologies such as single-cell RNA sequencing, metabolomics, and multi-omics analyses. We review evidence that sex hormones and sex chromosomes interact with various disease mechanisms during aging, encompassing inflammation, metabolism, and autophagy, leading to unique characteristics in disease progression between men and women.
Collapse
Affiliation(s)
- Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Eileen Ruth S Torres
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Kikuchi M, Miyashita A, Hara N, Kasuga K, Saito Y, Murayama S, Kakita A, Akatsu H, Ozaki K, Niida S, Kuwano R, Iwatsubo T, Nakaya A, Ikeuchi T. Polygenic effects on the risk of Alzheimer's disease in the Japanese population. Alzheimers Res Ther 2024; 16:45. [PMID: 38414085 PMCID: PMC10898021 DOI: 10.1186/s13195-024-01414-x] [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: 08/10/2023] [Accepted: 02/11/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Polygenic effects have been proposed to account for some disease phenotypes; these effects are calculated as a polygenic risk score (PRS). This score is correlated with Alzheimer's disease (AD)-related phenotypes, such as biomarker abnormalities and brain atrophy, and is associated with conversion from mild cognitive impairment (MCI) to AD. However, the AD PRS has been examined mainly in Europeans, and owing to differences in genetic structure and lifestyle, it is unclear whether the same relationships between the PRS and AD-related phenotypes exist in non-European populations. In this study, we calculated and evaluated the AD PRS in Japanese individuals using genome-wide association study (GWAS) statistics from Europeans. METHODS In this study, we calculated the AD PRS in 504 Japanese participants (145 cognitively unimpaired (CU) participants, 220 participants with late mild cognitive impairment (MCI), and 139 patients with mild AD dementia) enrolled in the Japanese Alzheimer's Disease Neuroimaging Initiative (J-ADNI) project. In order to evaluate the clinical value of this score, we (1) determined the polygenic effects on AD in the J-ADNI and validated it using two independent cohorts (a Japanese neuropathology (NP) cohort (n = 565) and the North American ADNI (NA-ADNI) cohort (n = 617)), (2) examined the AD-related phenotypes associated with the PRS, and (3) tested whether the PRS helps predict the conversion of MCI to AD. RESULTS The PRS using 131 SNPs had an effect independent of APOE. The PRS differentiated between CU participants and AD patients with an area under the curve (AUC) of 0.755 when combined with the APOE variants. Similar AUC was obtained when PRS calculated by the NP and NA-ADNI cohorts was applied. In MCI patients, the PRS was associated with cerebrospinal fluid phosphorylated-tau levels (β estimate = 0.235, p value = 0.026). MCI with a high PRS showed a significantly increased conversion to AD in APOE ε4 noncarriers with a hazard rate of 2.22. In addition, we also developed a PRS model adjusted for LD and observed similar results. CONCLUSIONS We showed that the AD PRS is useful in the Japanese population, whose genetic structure is different from that of the European population. These findings suggest that the polygenicity of AD is partially common across ethnic differences.
Collapse
Affiliation(s)
- Masataka Kikuchi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, 6-2-3 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan.
- Department of Medical Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Yuko Saito
- Brain Bank for Aging Research (Department of Neuropathology), Tokyo Metropolitan Institute of Geriatrics and Gerontology, Tokyo, Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research (Department of Neuropathology), Tokyo Metropolitan Institute of Geriatrics and Gerontology, Tokyo, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyasu Akatsu
- Department of General Medicine & General Internal Medicine, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Kouichi Ozaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Shumpei Niida
- Core Facility Administration, National Center for Geriatrics and Gerontology, Research Institute, Aichi, Japan
| | - Ryozo Kuwano
- Social Welfare Corporation Asahigawaso, Asahigawaso Research Institute, Okayama, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Nakaya
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, 6-2-3 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan.
| |
Collapse
|
14
|
Bi S, Yan S, Chen Z, Cui B, Shan Y, Yang H, Qi Z, Zhao Z, Han Y, Lu J. Comparison of 18F-FDG PET and arterial spin labeling MRI in evaluating Alzheimer's disease and amnestic mild cognitive impairment using integrated PET/MR. EJNMMI Res 2024; 14:9. [PMID: 38270821 PMCID: PMC10811308 DOI: 10.1186/s13550-024-01068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Developing biomarkers for early stage AD patients is crucial. Glucose metabolism measured by 18F-FDG PET is the most common biomarker for evaluating cellular energy metabolism to diagnose AD. Arterial spin labeling (ASL) MRI can potentially provide comparable diagnostic information to 18F-FDG PET in patients with neurodegenerative disorders. However, the conclusions about the diagnostic performance of AD are still controversial between 18F-FDG PET and ASL. This study aims to compare quantitative cerebral blood flow (CBF) and glucose metabolism measured by 18F-FDG PET diagnostic values in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) using integrated PET/MR. RESULTS Analyses revealed overlapping between decreased regional rCBF and 18F-FDG PET SUVR in patients with AD compared with NC participants in the bilateral parietotemporal regions, frontal cortex, and cingulate cortex. Compared with NC participants, patients with aMCI exclusively demonstrated lower 18F-FDG PET SUVR in the bilateral temporal cortex, insula cortex, and inferior frontal cortex. Comparison of the rCBF in patients with aMCI and NC participants revealed no significant difference (P > 0.05). The ROC analysis of rCBF in the meta-ROI could diagnose patients with AD (AUC, 0.87) but not aMCI (AUC, 0.61). The specificity of diagnosing aMCI has been improved to 75.56% when combining rCBF and 18F-FDG PET SUVR. CONCLUSION ASL could detect similar aberrant patterns of abnormalities compared to 18F-FDG PET in patients with AD compared with NC participants but not in aMCI. The diagnostic efficiency of 18F-FDG-PET for AD and aMCI patients remained higher to ASL. Our findings support that applying 18F-FDG PET may be preferable for diagnosing AD and aMCI.
Collapse
Affiliation(s)
- Sheng Bi
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Shaozhen Yan
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigeng Chen
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Bixiao Cui
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yi Shan
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Hongwei Yang
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigang Qi
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhilian Zhao
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| |
Collapse
|
15
|
Lang M, Colby S, Ashby-Padial C, Bapna M, Jaimes C, Rincon SP, Buch K. An imaging review of the hippocampus and its common pathologies. J Neuroimaging 2024; 34:5-25. [PMID: 37872430 DOI: 10.1111/jon.13165] [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: 06/29/2023] [Revised: 10/07/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023] Open
Abstract
The hippocampus is a complex structure located in the mesial temporal lobe that plays a critical role in cognitive and memory-related processes. The hippocampal formation consists of the dentate gyrus, hippocampus proper, and subiculum, and its importance in the neural circuitry makes it a key anatomic structure to evaluate in neuroimaging studies. Advancements in imaging techniques now allow detailed assessment of hippocampus internal architecture and signal features that has improved identification and characterization of hippocampal abnormalities. This review aims to summarize the neuroimaging features of the hippocampus and its common pathologies. It provides an overview of the hippocampal anatomy on magnetic resonance imaging and discusses how various imaging techniques can be used to assess the hippocampus. The review explores neuroimaging findings related to hippocampal variants (incomplete hippocampal inversion, sulcal remnant and choroidal fissure cysts), and pathologies of neoplastic (astrocytoma and glioma, ganglioglioma, dysembryoplastic neuroepithelial tumor, multinodular and vacuolating neuronal tumor, and metastasis), epileptic (mesial temporal sclerosis and focal cortical dysplasia), neurodegenerative (Alzheimer's disease, progressive primary aphasia, and frontotemporal dementia), infectious (Herpes simplex virus and limbic encephalitis), vascular (ischemic stroke, arteriovenous malformation, and cerebral cavernous malformations), and toxic-metabolic (transient global amnesia and opioid-associated amnestic syndrome) etiologies.
Collapse
Affiliation(s)
- Min Lang
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha Colby
- Department of Neurosurgery, University of Utah Health, Salt Lake City, Utah, USA
| | | | - Monika Bapna
- School of Medicine, Georgetown University, Washington, DC, USA
| | - Camilo Jaimes
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Sandra P Rincon
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Karen Buch
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Triumbari EKA, Chiaravalloti A, Schillaci O, Mercuri NB, Liguori C. Positron Emission Tomography/Computed Tomography Imaging in Therapeutic Clinical Trials in Alzheimer's Disease: An Overview of the Current State of the Art of Research. J Alzheimers Dis 2024; 101:S603-S628. [PMID: 39422956 DOI: 10.3233/jad-240349] [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: 10/19/2024]
Abstract
The integration of positron emission tomography/computed tomography (PET/CT) has revolutionized the landscape of Alzheimer's disease (AD) research and therapeutic interventions. By combining structural and functional imaging, PET/CT provides a comprehensive understanding of disease pathology and response to treatment assessment. PET/CT, particularly with 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG), facilitates the visualization of glucose metabolism in the brain, enabling early diagnosis, staging, and monitoring of neurodegenerative disease progression. The advent of amyloid and tau PET imaging has further propelled the field forward, offering invaluable tools for tracking pathological hallmarks, assessing treatment response, and predicting clinical outcomes. While some therapeutic interventions targeting amyloid plaque load showed promising results with the reduction of cerebral amyloid accumulation over time, others failed to demonstrate a significant impact of anti-amyloid agents for reducing the amyloid plaques burden in AD brains. Tau PET imaging has conversely fueled the advent of disease-modifying therapeutic strategies in AD by supporting the assessment of neurofibrillary tangles of tau pathology deposition over time. Looking ahead, PET imaging holds immense promise for studying additional targets such as neuroinflammation, cholinergic deficit, and synaptic dysfunction. Advances in radiotracer development, dedicated brain PET/CT scanners, and Artificial Intelligence-powered software are poised to enhance the quality, sensitivity, and diagnostic power of molecular neuroimaging. Consequently, PET/CT remains at the forefront of AD research, offering unparalleled opportunities for unravelling the complexities of the disease and advancing therapeutic interventions, although it is not yet enough alone to allow patients' recruitment in therapeutic clinical trials.
Collapse
Affiliation(s)
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
17
|
Su Z, Zhang G, Li X, Zhang H. Inverse correlation between Alzheimer's disease and cancer from the perspective of hypoxia. Neurobiol Aging 2023; 131:59-73. [PMID: 37572528 DOI: 10.1016/j.neurobiolaging.2023.07.002] [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: 01/08/2023] [Revised: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 08/14/2023]
Abstract
Sporadic Alzheimer's disease and cancer remain epidemiologically inversely related, and exploring the reverse pathogenesis is important for our understanding of both. Cognitive dysfunctions in Alzheimer's disease (AD) might result from the depletion of adaptive reserves in the brain. Energy storage in the brain is limited and is dynamically regulated by neurovascular and neurometabolic coupling. The research on neurodegenerative diseases has been dominated by the neurocentric view that neuronal defects cause the diseases. However, the proposal of the 2-hit vascular hypothesis in AD led us to focus on alterations in the vasculature, especially hypoperfusion. Chronic hypoxia is a feature shared by AD and cancer. It is interesting how contradicting chronic hypoxia's effects on both cancer and AD are. In this article, we discuss the potential links between the 2 diseases' etiology, from comparable upstream circumstances to diametrically opposed downstream effects. We suggest opposing potential mechanisms, including upregulation and downregulation of hypoxia-inducible factor-1α, the Warburg and reverse-Warburg effects, lactate-mediated intracellular acidic and alkaline conditions, and VDAC1-mediated apoptosis and antiapoptosis, and search for regulators that may be identified as the crossroads between cancer and AD.
Collapse
Affiliation(s)
- Zhan Su
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiangting Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
18
|
Cao E, Ma D, Nayak S, Duong TQ. Deep learning combining FDG-PET and neurocognitive data accurately predicts MCI conversion to Alzheimer's dementia 3-year post MCI diagnosis. Neurobiol Dis 2023; 187:106310. [PMID: 37769746 DOI: 10.1016/j.nbd.2023.106310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023] Open
Abstract
INTRODUCTION This study reports a novel deep learning approach to predict mild cognitive impairment (MCI) conversion to Alzheimer's dementia (AD) within three years using whole-brain fluorodeoxyglucose (FDG) positron emission tomography (PET) and cognitive scores (CS). METHODS This analysis consisted of 150 normal controls (CN), 257 MCI, and 205 AD subjects from ADNI. FDG-PET and CS were obtained at MCI diagnosis to predict AD conversion within three years of MCI diagnosis using convolutional neural networks. RESULTS Neurocognitive scores predicted better than FDG-PET per se, but the best model was a combination of FDG-PET, age, and neurocognitive data, yielding an AUC of 0.785 ± 0.096 and a balanced accuracy of 0.733 ± 0.098. Saliency maps highlighted putamen, thalamus, inferior frontal gyrus, parietal operculum, precuneus cortices, calcarine cortices, temporal gyrus, and planum temporale to be important for prediction. DISCUSSION Deep learning accurately predicts MCI conversion to AD and provides neural correlates of brain regions associated with AD conversion.
Collapse
Affiliation(s)
- Eric Cao
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10467, United States
| | - Da Ma
- Department of Internal Medicine Section of Gerontology and Geriatric Medicine, Wake Forest, University School of Medicine, Winston-Salam, NC 27109, United States
| | - Siddharth Nayak
- Department of Radiology, Weill Cornell Medicine, New York, 10065, United States
| | - Tim Q Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10467, United States.
| |
Collapse
|
19
|
Lima-Filho RAS, Benedet AL, De Bastiani MA, Povala G, Cozachenco D, Ferreira ST, De Felice FG, Rosa-Neto P, Zimmer ER, Lourenco MV. Association of the fibronectin type III domain-containing protein 5 rs1746661 single nucleotide polymorphism with reduced brain glucose metabolism in elderly humans. Brain Commun 2023; 5:fcad216. [PMID: 37601408 PMCID: PMC10438215 DOI: 10.1093/braincomms/fcad216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/23/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
Fibronectin type III domain-containing protein 5 (FNDC5) and its derived hormone, irisin, have been associated with metabolic control in humans, with described FNDC5 single nucleotide polymorphisms being linked to obesity and metabolic syndrome. Decreased brain FNDC5/irisin has been reported in subjects with dementia due to Alzheimer's disease. Since impaired brain glucose metabolism develops in ageing and is prominent in Alzheimer's disease, here, we examined associations of a single nucleotide polymorphism in the FNDC5 gene (rs1746661) with brain glucose metabolism and amyloid-β deposition in a cohort of 240 cognitively unimpaired and 485 cognitively impaired elderly individuals from the Alzheimer's Disease Neuroimaging Initiative. In cognitively unimpaired elderly individuals harbouring the FNDC5 rs1746661(T) allele, we observed a regional reduction in low glucose metabolism in memory-linked brain regions and increased brain amyloid-β PET load. No differences in cognition or levels of cerebrospinal fluid amyloid-β42, phosphorylated tau and total tau were observed between FNDC5 rs1746661(T) allele carriers and non-carriers. Our results indicate that a genetic variant of FNDC5 is associated with low brain glucose metabolism in elderly individuals and suggest that FNDC5 may participate in the regulation of brain metabolism in brain regions vulnerable to Alzheimer's disease pathophysiology. Understanding the associations between genetic variants in metabolism-linked genes and metabolic brain signatures may contribute to elucidating genetic modulators of brain metabolism in humans.
Collapse
Affiliation(s)
- Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 413 45, Sweden
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Guilherme Povala
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ 22281-100, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ 22281-100, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health University Institute, Departments of Neurology and Neurosurgery, Psychiatry, and Pharmacology, McGill University, Montreal, QC H4H 1R3, Canada
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | | |
Collapse
|
20
|
Zhang NK, Zhang SK, Zhang LI, Tao HW, Zhang GW. Sensory processing deficits and related cortical pathological changes in Alzheimer's disease. Front Aging Neurosci 2023; 15:1213379. [PMID: 37649717 PMCID: PMC10464619 DOI: 10.3389/fnagi.2023.1213379] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder primarily affecting cognitive functions. However, sensory deficits in AD start to draw attention due to their high prevalence and early onsets which suggest that they could potentially serve as diagnostic biomarkers and even contribute to the disease progression. This literature review examines the sensory deficits and cortical pathological changes observed in visual, auditory, olfactory, and somatosensory systems in AD patients, as well as in various AD animal models. Sensory deficits may emerge at the early stages of AD, or even precede the cognitive decline, which is accompanied by cortical pathological changes including amyloid-beta deposition, tauopathy, gliosis, and alterations in neuronal excitability, synaptic inputs, and functional plasticity. Notably, these changes are more pronounced in sensory association areas and superficial cortical layers, which may explain the relative preservation of basic sensory functions but early display of deficits of higher sensory functions. We propose that sensory impairment and the progression of AD may establish a cyclical relationship that mutually perpetuates each condition. This review highlights the significance of sensory deficits with or without cortical pathological changes in AD and emphasizes the need for further research to develop reliable early detection and intervention through sensory systems.
Collapse
Affiliation(s)
- Nicole K. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Selena K. Zhang
- Biomedical Engineering Program, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Huizhong W. Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
21
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
22
|
Gan J, Shi Z, Zuo C, Zhao X, Liu S, Chen Y, Zhang N, Cai L, Cui R, Ai L, Guan YH, Ji Y. Analysis of positron emission tomography hypometabolic patterns and neuropsychiatric symptoms in patients with dementia syndromes. CNS Neurosci Ther 2023. [PMID: 36924296 DOI: 10.1111/cns.14169] [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: 11/02/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
AIMS To estimate the proportions of specific hypometabolic patterns and their association with neuropsychiatric symptoms (NPS) in patients with cognitive impairment (CI). METHODS This multicenter study with 1037 consecutive patients was conducted from December 2012 to December 2019. 18 F-FDG PET and clinical/demographic information, NPS assessments were recorded and analyzed to explore the associations between hypometabolic patterns and clinical features by correlation analysis and multivariable logistic regression models. RESULTS Patients with clinical Alzheimer's disease (AD, 81.6%, 605/741) and dementia with Lewy bodies (67.9%, 19/28) mostly had AD-pattern hypometabolism, and 76/137 (55.5%) of patients with frontotemporal lobar degeneration showed frontal and anterior temporal pattern (FT-P) hypometabolism. Besides corticobasal degeneration, patients with behavioral variant frontotemporal dementia (36/58), semantic dementia (7/10), progressive non-fluent aphasia (6/9), frontotemporal lobar degeneration and amyotrophic lateral sclerosis (3/5), and progressive supranuclear palsy (21/37) also mostly showed FT-P hypometabolism. The proportion of FT-P hypometabolism was associated with the presence of hallucinations (R = 0.171, p = 0.04), anxiety (R = 0.182, p = 0.03), and appetite and eating abnormalities (R = 0.200, p = 0.01) in AD. CONCLUSION Specific hypometabolic patterns in FDG-PET are associated with NPS and beneficial for the early identification and management of NPS in patients with CI.
Collapse
Affiliation(s)
- Jinghuan Gan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhihong Shi
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaobin Zhao
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Yongjie Chen
- Department of Epidemiology and Statistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Nan Zhang
- Department of Neurology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Li Cai
- Department of PET-CT Diagnostics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruixue Cui
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi-Hui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Ji
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
23
|
Gnörich J, Reifschneider A, Wind K, Zatcepin A, Kunte ST, Beumers P, Bartos LM, Wiedemann T, Grosch M, Xiang X, Fard MK, Ruch F, Werner G, Koehler M, Slemann L, Hummel S, Briel N, Blume T, Shi Y, Biechele G, Beyer L, Eckenweber F, Scheifele M, Bartenstein P, Albert NL, Herms J, Tahirovic S, Haass C, Capell A, Ziegler S, Brendel M. Depletion and activation of microglia impact metabolic connectivity of the mouse brain. J Neuroinflammation 2023; 20:47. [PMID: 36829182 PMCID: PMC9951492 DOI: 10.1186/s12974-023-02735-8] [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: 07/03/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
AIM We aimed to investigate the impact of microglial activity and microglial FDG uptake on metabolic connectivity, since microglial activation states determine FDG-PET alterations. Metabolic connectivity refers to a concept of interacting metabolic brain regions and receives growing interest in approaching complex cerebral metabolic networks in neurodegenerative diseases. However, underlying sources of metabolic connectivity remain to be elucidated. MATERIALS AND METHODS We analyzed metabolic networks measured by interregional correlation coefficients (ICCs) of FDG-PET scans in WT mice and in mice with mutations in progranulin (Grn) or triggering receptor expressed on myeloid cells 2 (Trem2) knockouts (-/-) as well as in double mutant Grn-/-/Trem2-/- mice. We selected those rodent models as they represent opposite microglial signatures with disease associated microglia in Grn-/- mice and microglia locked in a homeostatic state in Trem2-/- mice; however, both resulting in lower glucose uptake of the brain. The direct influence of microglia on metabolic networks was further determined by microglia depletion using a CSF1R inhibitor in WT mice at two different ages. Within maps of global mean scaled regional FDG uptake, 24 pre-established volumes of interest were applied and assigned to either cortical or subcortical networks. ICCs of all region pairs were calculated and z-transformed prior to group comparisons. FDG uptake of neurons, microglia, and astrocytes was determined in Grn-/- and WT mice via assessment of single cell tracer uptake (scRadiotracing). RESULTS Microglia depletion by CSF1R inhibition resulted in a strong decrease of metabolic connectivity defined by decrease of mean cortical ICCs in WT mice at both ages studied (6-7 m; p = 0.0148, 9-10 m; p = 0.0191), when compared to vehicle-treated age-matched WT mice. Grn-/-, Trem2-/- and Grn-/-/Trem2-/- mice all displayed reduced FDG-PET signals when compared to WT mice. However, when analyzing metabolic networks, a distinct increase of ICCs was observed in Grn-/- mice when compared to WT mice in cortical (p < 0.0001) and hippocampal (p < 0.0001) networks. In contrast, Trem2-/- mice did not show significant alterations in metabolic connectivity when compared to WT. Furthermore, the increased metabolic connectivity in Grn-/- mice was completely suppressed in Grn-/-/Trem2-/- mice. Grn-/- mice exhibited a severe loss of neuronal FDG uptake (- 61%, p < 0.0001) which shifted allocation of cellular brain FDG uptake to microglia (42% in Grn-/- vs. 22% in WT). CONCLUSIONS Presence, absence, and activation of microglia have a strong impact on metabolic connectivity of the mouse brain. Enhanced metabolic connectivity is associated with increased microglial FDG allocation.
Collapse
Affiliation(s)
- Johannes Gnörich
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Anika Reifschneider
- grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Karin Wind
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Artem Zatcepin
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Sebastian T. Kunte
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Philipp Beumers
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Laura M. Bartos
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Thomas Wiedemann
- grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maximilian Grosch
- grid.5252.00000 0004 1936 973XGerman Center for Vertigo and Balance Disorders, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Xianyuan Xiang
- grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany ,grid.9227.e0000000119573309CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055 China
| | - Maryam K. Fard
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Francois Ruch
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Georg Werner
- grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mara Koehler
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Luna Slemann
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Selina Hummel
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Nils Briel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XCenter for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tanja Blume
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XCenter for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yuan Shi
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XCenter for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gloria Biechele
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Leonie Beyer
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Florian Eckenweber
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Maximilian Scheifele
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Peter Bartenstein
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nathalie L. Albert
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Jochen Herms
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XCenter for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sabina Tahirovic
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Christian Haass
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anja Capell
- grid.5252.00000 0004 1936 973XMetabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sibylle Ziegler
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377, Munich, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
24
|
Parra MA, Orellana P, Leon T, Victoria CG, Henriquez F, Gomez R, Avalos C, Damian A, Slachevsky A, Ibañez A, Zetterberg H, Tijms BM, Yokoyama JS, Piña-Escudero SD, Cochran JN, Matallana DL, Acosta D, Allegri R, Arias-Suárez BP, Barra B, Behrens MI, Brucki SMD, Busatto G, Caramelli P, Castro-Suarez S, Contreras V, Custodio N, Dansilio S, De la Cruz-Puebla M, de Souza LC, Diaz MM, Duque L, Farías GA, Ferreira ST, Guimet NM, Kmaid A, Lira D, Lopera F, Meza BM, Miotto EC, Nitrini R, Nuñez A, O'neill S, Ochoa J, Pintado-Caipa M, de Paula França Resende E, Risacher S, Rojas LA, Sabaj V, Schilling L, Sellek AF, Sosa A, Takada LT, Teixeira AL, Unaucho-Pilalumbo M, Duran-Aniotz C. Biomarkers for dementia in Latin American countries: Gaps and opportunities. Alzheimers Dement 2023; 19:721-735. [PMID: 36098676 PMCID: PMC10906502 DOI: 10.1002/alz.12757] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/29/2022] [Accepted: 06/14/2022] [Indexed: 12/13/2022]
Abstract
Limited knowledge on dementia biomarkers in Latin American and Caribbean (LAC) countries remains a serious barrier. Here, we reported a survey to explore the ongoing work, needs, interests, potential barriers, and opportunities for future studies related to biomarkers. The results show that neuroimaging is the most used biomarker (73%), followed by genetic studies (40%), peripheral fluids biomarkers (31%), and cerebrospinal fluid biomarkers (29%). Regarding barriers in LAC, lack of funding appears to undermine the implementation of biomarkers in clinical or research settings, followed by insufficient infrastructure and training. The survey revealed that despite the above barriers, the region holds a great potential to advance dementia biomarkers research. Considering the unique contributions that LAC could make to this growing field, we highlight the urgent need to expand biomarker research. These insights allowed us to propose an action plan that addresses the recommendations for a biomarker framework recently proposed by regional experts.
Collapse
Affiliation(s)
- Mario A. Parra
- School of Psychological Sciences and Health, University of Strathclyde. Glasgow, United Kingdom
| | - Paulina Orellana
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez. Santiago, Chile
| | - Tomas Leon
- Global Brain Health Institute, Trinity College. Dublin, Ireland
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador y Facultad de Medicina, Universidad de Chile. Santiago, Chile
| | - Cabello G. Victoria
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, Universidad de Chile. Santiago, Chile
- Unit of Brain Health, Department of Neurology and Neurosurgery, Faculty of Medicine, Universidad de Chile. Santiago, Chile
| | - Fernando Henriquez
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, Universidad de Chile. Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO). Santiago, Chile
- Laboratory for Cognitive and Evolutionary Neuroscience (LaNCE), Department of Psychiatry, Faculty of Medicine, Pontificia Universidad Católica de Chile. Santiago, Chile
| | - Rodrigo Gomez
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador y Facultad de Medicina, Universidad de Chile. Santiago, Chile
- Graduate School, Faculty of Medicine, Universidad Mayor, Chile - Centro de Apoyo Comunitario a personas con Demencia Kintun. Santiago, Chile
| | - Constanza Avalos
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez. Santiago, Chile
| | - Andres Damian
- Centro Uruguayo de Imagenología Molecular (CUDIM) - Centro de Medicina Nuclear e Imagenología Molecular, Hospital de Clínicas, Universidad de la República. Montevideo, Uruguay
| | - Andrea Slachevsky
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador y Facultad de Medicina, Universidad de Chile. Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, Universidad de Chile. Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO). Santiago, Chile
- Department of Neurology and Psyquiatry, Clínica Alemana-Universidad del Desarrollo. Santiago, Chile
| | - Agustin Ibañez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez. Santiago, Chile
- Global Brain Health Institute, Trinity College. Dublin, Ireland
- Global Brain Health Institute and the Memory and Aging Center, Weill Institute for Neurosciences, Departments of Neurology and Radiology & Biomedical Imaging, University of California, San Francisco (UCSF). San Francisco, USA
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, & National Scientific and Technical Research Council (CONICET). Buenos Aires, Argentina
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg. Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital. Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology. Queen Square, London, UK
- UK Dementia Research Institute at UCL. London, UK
- Hong Kong Center for Neurodegenerative Diseases. Clear Water Bay, Hong Kong, China
| | - Betty M. Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience. Amsterdam UMC, The Netherlands
| | - Jennifer S. Yokoyama
- Global Brain Health Institute and the Memory and Aging Center, Weill Institute for Neurosciences, Departments of Neurology and Radiology & Biomedical Imaging, University of California, San Francisco (UCSF). San Francisco, USA
- Department of Neurology, Memory and Aging Center, UCSF. San Francisco, USA
| | - Stefanie D. Piña-Escudero
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
| | | | - Diana L Matallana
- Medical School, Aging Institute and Psychiatry Department, Neuroscience PhD Program, Pontificia Universidad Javeriana. Bogotá,Colombia
- Memory and Cognition Center, Intellectus, Hospital Universitario San Ignacio. Bogotá, Colombia
- Psychiatry Department, Hospital Universitario Santa Fe de Bogotá. Bogotá, Colombia
| | - Daisy Acosta
- Universidad Nacional Pedro Henriquez Urena (UNPHU). Santo Domingo, República Dominicana
| | - Ricardo Allegri
- Department of Cognitive Neurology, Neuropsychiatry and Neuropsychology, Instituto Neurológico Fleni. Buenos Aires, Argentina
- Department of Neurosciences, Universidad de la Costa. Barranquilla, Colombia
| | - Bianca P. Arias-Suárez
- Faculty of Human Medicine, Postgraduate Section, National University of San Marcos. Lima, Perú
| | - Bernardo Barra
- Mental Health Service, Clínica Universidad de los Andes. Santiago, Chile
- Department of Psychiatry, Medicine School, Andrés Bello University of Santiago (UNAB). Santiago, Chile
| | - Maria Isabel Behrens
- Department of Neurology and Psyquiatry, Clínica Alemana-Universidad del Desarrollo. Santiago, Chile
- Center for Advanced Clinical Research (CICA). Department of Neurology & Neurosurgery and Neuroscience Department, Faculty of Medicine, Universidad de Chile. Santiago, Chile
- Department of Neurology and Neurosurgery, Hospital Clínico Universidad de Chile. Santiago, Chile
- Department of Neurocience, Faculty of Medicine, Universidad de Chile. Santiago, Chile
| | - Sonia M. D. Brucki
- Cognitive and Behavioral Neurology Unit, Department of Neurology, University of São Paulo Medical School, University of São Paulo. São Paulo, Brazil
| | - Geraldo Busatto
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP. São Paulo, Brazil
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais. Belo Horizonte, Brazil
| | - Sheila Castro-Suarez
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
- Instituto Nacional de Ciencias Neurológicas. Lima, Perú
| | | | - Nilton Custodio
- Unit of diagnosis of cognitive impairment and dementia prevention, Instituto Peruano de Neurociencias.Lima, Perú
| | - Sergio Dansilio
- Department of Neuropsychology, Institut of Neurology, Hospital de Clínicas, Faculty of Medicine,Universidad de la República. Montevideo, Uruguay
| | - Myriam De la Cruz-Puebla
- Global Brain Health Institute and the Memory and Aging Center, Weill Institute for Neurosciences, Departments of Neurology and Radiology & Biomedical Imaging, University of California, San Francisco (UCSF). San Francisco, USA
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute. Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Neuroscience Institute, Autonomous University of Barcelona. Barcelona, Spain
- Department of Internal Medicine, Health Sciences Faculty, Technical University of Ambato. Tungurahua, Ecuador
| | - Leonardo Cruz de Souza
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP. São Paulo, Brazil
- Neurology Service, School of Medicine, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
| | - Monica M. Diaz
- Department of Neurology, University of North Carolina at Chapel Hill. North Carolina, USA
- School of Public Health, Universidad Peruana Cayetano Heredia. Lima, Peru
| | - Lissette Duque
- Unit of Cognitive diseases, Neuromedicenter. Quito, Ecuador
| | - Gonzalo A. Farías
- Center for Advanced Clinical Research (CICA). Department of Neurology & Neurosurgery and Neuroscience Department, Faculty of Medicine, Universidad de Chile. Santiago, Chile
| | - Sergio T. Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro. Rio de Janeiro, Brazil
| | - Nahuel Magrath Guimet
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
- Department of Cognitive Neurology, Neuropsychiatry and Neuropsychology, Instituto Neurológico Fleni. Buenos Aires, Argentina
| | - Ana Kmaid
- Unit of Cognitive evaluation. Department of Geriatry ang Gerentology. Hospital de Clínicas. Faculty of Medicine. Universidad de la República. Montevideo, Uruguay
| | - David Lira
- Unit of diagnosis of cognitive impairment and dementia prevention, Instituto Peruano de Neurociencias.Lima, Perú
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, School of Medicine. Medellín, Colombia
| | - Beatriz Mar Meza
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
- Department of Geriatry ang Gerentology, Hospital Central de la Fuerza Aérea del Perú. Lima, Perú
| | - Eliane C Miotto
- Cognitive and Behavioral Neurology Unit, Department of Neurology, University of São Paulo Medical School, University of São Paulo. São Paulo, Brazil
| | - Ricardo Nitrini
- Cognitive and Behavioral Neurology Unit, Department of Neurology, University of São Paulo Medical School, University of São Paulo. São Paulo, Brazil
| | - Alberto Nuñez
- Unit of Cognitive diseases, Neuromedicenter. Quito, Ecuador
| | - Santiago O'neill
- Neurosciences Institute, Favaloro Foundation University Hospital. Buenos Aires, Argentina
| | - John Ochoa
- Group of Neuropsychology and behavior, Universidad de Antioquia, School of Medicine. Medellín, Colombia
| | - Maritza Pintado-Caipa
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
- Unit of diagnosis of cognitive impairment and dementia prevention, Instituto Peruano de Neurociencias.Lima, Perú
| | - Elisa de Paula França Resende
- Global Brain Health Institute and the Memory and Aging Center, Weill Institute for Neurosciences, Departments of Neurology and Radiology & Biomedical Imaging, University of California, San Francisco (UCSF). San Francisco, USA
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais. Belo Horizonte, Brazil
- Neurology Service, School of Medicine, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
- Faculdade de Ciências Médicas de Minas Gerais. Belo Horizonte, Brazil
| | - Shannon Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer’s Disease Research Center, Department of Neurology, Indiana University School of Medicine. Indianapolis, USA
| | - Luz Angela Rojas
- Research Group, MI Dneuropsy, Universidad Surcolombiana. Neiva, Colombia
| | - Valentina Sabaj
- Unit of Neuropsychogeriatry, Instituto Nacional de Geriatría. Santiago, Chile
| | - Lucas Schilling
- Neurology Service, School of Medicine, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
- Graduate Program in Biomedical Gerontology, Pontifical University of Rio Grande do Sul (PUCRS). Porto Alegre, Brazil
| | | | - Ana Sosa
- Instituto Nacional de Neurología y Neurocirugía (INNN), Manuel Velasco Suarez. Ciudad de México, México
| | - Leonel T. Takada
- Cognitive and Behavioral Neurology Unit, Department of Neurology, University of São Paulo Medical School, University of São Paulo. São Paulo, Brazil
| | - Antonio L. Teixeira
- Faculdade Santa Casa BH. Belo Horizonte, Brazil
- Neuropsychiatry Program, University of Texas Health Science Center at Houston. Houston, USA
| | - Martha Unaucho-Pilalumbo
- Atlantic Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), University of California San Francisco. San Francisco, USA
- Departamento de Neurología, Hospital Universidad Técnica Particular de Loja. Loja, Ecuador
| | - Claudia Duran-Aniotz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez. Santiago, Chile
| |
Collapse
|
25
|
Loftus JR, Puri S, Meyers SP. Multimodality imaging of neurodegenerative disorders with a focus on multiparametric magnetic resonance and molecular imaging. Insights Imaging 2023; 14:8. [PMID: 36645560 PMCID: PMC9842851 DOI: 10.1186/s13244-022-01358-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Neurodegenerative diseases afflict a large number of persons worldwide, with the prevalence and incidence of dementia rapidly increasing. Despite their prevalence, clinical diagnosis of dementia syndromes remains imperfect with limited specificity. Conventional structural-based imaging techniques also lack the accuracy necessary for confident diagnosis. Multiparametric magnetic resonance imaging and molecular imaging provide the promise of improving specificity and sensitivity in the diagnosis of neurodegenerative disease as well as therapeutic monitoring of monoclonal antibody therapy. This educational review will briefly focus on the epidemiology, clinical presentation, and pathologic findings of common and uncommon neurodegenerative diseases. Imaging features of each disease spanning from conventional magnetic resonance sequences to advanced multiparametric methods such as resting-state functional magnetic resonance imaging and arterial spin labeling imaging will be described in detail. Additionally, the review will explore the findings of each diagnosis on molecular imaging including single-photon emission computed tomography and positron emission tomography with a variety of clinically used and experimental radiotracers. The literature and clinical cases provided demonstrate the power of advanced magnetic resonance imaging and molecular techniques in the diagnosis of neurodegenerative diseases and areas of future and ongoing research. With the advent of combined positron emission tomography/magnetic resonance imaging scanners, hybrid protocols utilizing both techniques are an attractive option for improving the evaluation of neurodegenerative diseases.
Collapse
Affiliation(s)
- James Ryan Loftus
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Savita Puri
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| | - Steven P. Meyers
- grid.412750.50000 0004 1936 9166Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642 USA
| |
Collapse
|
26
|
Milano C, Hoxhaj D, Del Chicca M, Pascazio A, Paoli D, Tommasini L, Vergallo A, Pizzanelli C, Tognoni G, Nuti A, Ceravolo R, Siciliano G, Hampel H, Baldacci F. Alzheimer's Disease and Neurosyphilis: Meaningful Commonalities and Differences of Clinical Phenotype and Pathophysiological Biomarkers. J Alzheimers Dis 2023; 94:611-625. [PMID: 37334599 DOI: 10.3233/jad-230170] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Neurosyphilis-associated cognitive and behavioral impairment- historically coined as "general paralysis of the insane"- share clinical and neuroradiological features with the neurodegenerative disease spectrum, in particular Alzheimer's disease (AD). Anatomopathological similarities have been extensively documented, i.e., neuronal loss, fibrillary alterations, and local amyloid-β deposition. Consequently, accurate classification and timely differential diagnosis may be challenging. OBJECTIVE To describe clinical, bio-humoral, brain MRI, FDG-PET, and amyloid-PET features in cases of neurosyphilis with an AD-like phenotypical presentation, as well as clinical outcome in terms of response to antibiotic therapy. METHODS We selected the studies comparing patients with AD and with neurosyphilis associated cognitive impairment, to investigate candidate biomarkers classifying the two neurological diseases. RESULTS The neuropsychological phenotype of general paralysis, characterized by episodic memory impairment and executive disfunction, substantially mimics clinical AD features. Neuroimaging often shows diffuse or medial temporal cortical atrophy, thus contributing to a high rate of misdiagnosis. Cerebrospinal fluid (CSF)-based analysis may provide supportive diagnostic value, since increased proteins or cells are often found in neurosyphilis, while published data on pathophysiological AD candidate biomarkers are controversial. Finally, psychometric testing using cross-domain cognitive tests, may highlight a wider range of compromised functions in neurosyphilis, involving language, attention, executive function, and spatial ability, which are atypical for AD. CONCLUSION Neurosyphilis should be considered a potential etiological differential diagnosis of cognitive impairment whenever imaging, neuropsychological or CSF features are atypical for AD, in order to promptly start antibiotic therapy and delay or halt cognitive decline and disease progression.
Collapse
Affiliation(s)
- Chiara Milano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Domeniko Hoxhaj
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marta Del Chicca
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessia Pascazio
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Davide Paoli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Tommasini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Chiara Pizzanelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Angelo Nuti
- Division of Neurology, Versilia Hospital, Lido di Camaiore, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
27
|
He DL, Fan YG, Wang ZY. Energy Crisis Links to Autophagy and Ferroptosis in Alzheimer's Disease: Current Evidence and Future Avenues. Curr Neuropharmacol 2023; 21:67-86. [PMID: 35980072 PMCID: PMC10193753 DOI: 10.2174/1570159x20666220817140737] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/14/2022] [Accepted: 08/11/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases worldwide. The occult nature of the onset and the uncertainty of the etiology largely impede the development of therapeutic strategies for AD. Previous studies revealed that the disorder of energy metabolism in the brains of AD patients appears far earlier than the typical pathological features of AD, suggesting a tight association between energy crisis and the onset of AD. Energy crisis in the brain is known to be induced by the reductions in glucose uptake and utilization, which may be ascribed to the diminished expressions of cerebral glucose transporters (GLUTs), insulin resistance, mitochondrial dysfunctions, and lactate dysmetabolism. Notably, the energy sensors such as peroxisome proliferators-activated receptor (PPAR), transcription factor EB (TFEB), and AMP-activated protein kinase (AMPK) were shown to be the critical regulators of autophagy, which play important roles in regulating beta-amyloid (Aβ) metabolism, tau phosphorylation, neuroinflammation, iron dynamics, as well as ferroptosis. In this study, we summarized the current knowledge on the molecular mechanisms involved in the energy dysmetabolism of AD and discussed the interplays existing between energy crisis, autophagy, and ferroptosis. In addition, we highlighted the potential network in which autophagy may serve as a bridge between energy crisis and ferroptosis in the progression of AD. A deeper understanding of the relationship between energy dysmetabolism and AD may provide new insight into developing strategies for treating AD; meanwhile, the energy crisis in the progression of AD should gain more attention.
Collapse
Affiliation(s)
- Da-Long He
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Zhan-You Wang
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| |
Collapse
|
28
|
Functional Correlates of Striatal Dopamine Transporter Cerebrospinal Fluid Levels in Alzheimer's Disease: A Preliminary 18F-FDG PET/CT Study. Int J Mol Sci 2023; 24:ijms24010751. [PMID: 36614193 PMCID: PMC9820963 DOI: 10.3390/ijms24010751] [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: 12/09/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
The aim of our study was to investigate regional glucose metabolism with 18F-FDG positron emission tomography/computed tomography in a population of patients with Alzheimer's disease (AD) in relation to cerebrospinal (CSF) levels of striatal dopamine transporter (DAT). All patients underwent lumbar puncture and received a biomarker-based diagnosis of AD. Differences in regional brain glucose metabolism were assessed by Statistical Parametric Mapping version 12 with the use of age, gender, and MMSE as covariates in the analysis. A positive correlation between CSF DAT levels and glucose metabolism at the level of two brain areas involved in the pathophysiological process of Alzheimer's disease, the substantia nigra and the posterior cingulate gyrus, has been highlighted. Results indicate that patients with higher CSF DAT levels have a better metabolic pattern in two key zones, suggesting less advanced disease status in patients with more conserved dopaminergic systems.
Collapse
|
29
|
Morimoto-Ishikawa D, Hanaoka K, Watanabe S, Yamada T, Yamakawa Y, Minagawa S, Takenouchi S, Ohtani A, Mizuta T, Kaida H, Ishii K. Evaluation of the performance of a high-resolution time-of-flight PET system dedicated to the head and breast according to NEMA NU 2-2012 standard. EJNMMI Phys 2022; 9:88. [PMID: 36525103 PMCID: PMC9758266 DOI: 10.1186/s40658-022-00518-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND This study evaluated the physical performance of a positron emission tomography (PET) system dedicated to the head and breast according to the National Electrical Manufacturers Association (NEMA) NU2-2012 standard. METHODS The spatial resolution, sensitivity, scatter fraction, count rate characteristics, corrections for count losses and randoms, and image quality of the system were determined. All measurements were performed according to the NEMA NU2-2012 acquisition protocols, but image quality was assessed using a brain-sized phantom. Furthermore, scans of the three-dimensional (3D) Hoffmann brain phantom and mini-Derenzo phantom were acquired to allow visual evaluation of the imaging performance for small structures. RESULTS The tangential, radial, and axial full width at half maximum (FWHM) at a 10-mm offset in half the axial field of view were measured as 2.3, 2.5, and 2.9 mm, respectively. The average system sensitivity at the center of the field of view and at a 10-cm radial offset was 7.18 and 8.65 cps/kBq, respectively. The peak noise-equivalent counting rate was 35.2 kcps at 4.8 kBq/ml. The corresponding scatter fraction at the peak noise-equivalent counting rate was 46.8%. The peak true rate and scatter fraction at 8.6 kBq/ml were 127.8 kcps and 54.3%, respectively. The percent contrast value for a 10-mm sphere was approximately 50%. On the 3D Hoffman brain phantom image, the structures of the thin layers composing the phantom were visualized on the sagittal and coronal images. On the mini-Derenzo phantom, each of the 1.6-mm rods was clearly visualized. CONCLUSION Taken together, these results indicate that the head- and breast-dedicated PET system has high resolution and is well suited for clinical PET imaging.
Collapse
Affiliation(s)
- Daisuke Morimoto-Ishikawa
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan
| | - Kohei Hanaoka
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan
| | - Shota Watanabe
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan
| | - Takahiro Yamada
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan
| | - Yoshiyuki Yamakawa
- grid.274249.e0000 0004 0571 0853Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Suzuka Minagawa
- grid.274249.e0000 0004 0571 0853Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Shiho Takenouchi
- grid.274249.e0000 0004 0571 0853Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Atsushi Ohtani
- grid.274249.e0000 0004 0571 0853Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Tetsuro Mizuta
- grid.274249.e0000 0004 0571 0853Medical Systems Division, Shimadzu Corporation, Kyoto, Japan
| | - Hayato Kaida
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan ,grid.258622.90000 0004 1936 9967Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kazunari Ishii
- grid.413111.70000 0004 0466 7515Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University Hospital, 377-2 Ohno-Higashi, Osakasayama, Osaka 589-8511 Japan ,grid.258622.90000 0004 1936 9967Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
30
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, Souza LCD, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer’s disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022. [DOI: 10.1590/1980-5764-dn-2022-s102en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
ABSTRACT This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer’s disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil
| | | | | | | | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Brasil; Universidade de São Paulo, Brasil
| | | | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Brasil; Universidade Federal de Pernambuco, Brasil; Instituto de Medicina Integral Prof. Fernando Figueira, Brasil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, de Souza LC, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer's disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022; 16:25-39. [PMID: 36533157 PMCID: PMC9745995 DOI: 10.1590/1980-5764-dn-2022-s102pt] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/22/2021] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer's disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brasil
| | | | - Márcia Radanovic
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brasil
| | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Departamento de Fonoaudiologia, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Jerusa Smid
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
- Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brasil
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brasil
| | | | - Leonardo Cruz de Souza
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | - Francisco Assis Carvalho Vale
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brasil
| | - Paulo Caramelli
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | | | - Márcia Lorena Fagundes Chaves
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brasil
| | - Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Benito Pereira Damasceno
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brasil
| | - Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| |
Collapse
|
32
|
Exploring the brain metabolic correlates of process-specific CSF biomarkers in patients with MCI due to Alzheimer's disease: preliminary data. Neurobiol Aging 2022; 117:212-221. [DOI: 10.1016/j.neurobiolaging.2022.03.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 12/30/2022]
|
33
|
Hadjihambi A, Cudalbu C, Pierzchala K, Simicic D, Donnelly C, Konstantinou C, Davies N, Habtesion A, Gourine AV, Jalan R, Hosford PS. Abnormal brain oxygen homeostasis in an animal model of liver disease. JHEP Rep 2022; 4:100509. [PMID: 35865351 PMCID: PMC9293761 DOI: 10.1016/j.jhepr.2022.100509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/23/2022] [Accepted: 05/10/2022] [Indexed: 01/15/2023] Open
Abstract
Background & Aims Increased plasma ammonia concentration and consequent disruption of brain energy metabolism could underpin the pathogenesis of hepatic encephalopathy (HE). Brain energy homeostasis relies on effective maintenance of brain oxygenation, and dysregulation impairs neuronal function leading to cognitive impairment. We hypothesised that HE is associated with reduced brain oxygenation and we explored the potential role of ammonia as an underlying pathophysiological factor. Methods In a rat model of chronic liver disease with minimal HE (mHE; bile duct ligation [BDL]), brain tissue oxygen measurement, and proton magnetic resonance spectroscopy were used to investigate how hyperammonaemia impacts oxygenation and metabolic substrate availability in the central nervous system. Ornithine phenylacetate (OP, OCR-002; Ocera Therapeutics, CA, USA) was used as an experimental treatment to reduce plasma ammonia concentration. Results In BDL animals, glucose, lactate, and tissue oxygen concentration in the cerebral cortex were significantly lower than those in sham-operated controls. OP treatment corrected the hyperammonaemia and restored brain tissue oxygen. Although BDL animals were hypotensive, cortical tissue oxygen concentration was significantly improved by treatments that increased arterial blood pressure. Cerebrovascular reactivity to exogenously applied CO2 was found to be normal in BDL animals. Conclusions These data suggest that hyperammonaemia significantly decreases cortical oxygenation, potentially compromising brain energy metabolism. These findings have potential clinical implications for the treatment of patients with mHE. Lay summary Brain dysfunction is a serious complication of cirrhosis and affects approximately 30% of these patients; however, its treatment continues to be an unmet clinical need. This study shows that oxygen concentration in the brain of an animal model of cirrhosis is markedly reduced. Low arterial blood pressure and increased ammonia (a neurotoxin that accumulates in patients with liver failure) are shown to be the main underlying causes. Experimental correction of these abnormalities restored oxygen concentration in the brain, suggesting potential therapeutic avenues to explore.
Collapse
Key Words
- 1H-MRS, proton magnetic resonance spectroscopy
- AIT, Animal Imaging and Technology
- ALT, alanine transaminase
- ATZ, acetazolamide
- Ala, alanine
- Asc, ascorbate
- Asp, aspartate
- BDL, bile duct ligation
- BOLD, blood oxygen level dependent
- BP, blood pressure
- CBF, cerebral blood flow
- CIBM, Center for Biomedical Imaging
- CLD, chronic liver disease
- CMRO2, cerebral metabolic rate of oxygen
- CNS, central nervous system
- Chronic liver disease
- Cr, creatine
- EPFL, Ecole Polytechnique Fédérale de Lausanne
- GABA, γ-aminobutyric acid
- GPC, glycerophosphocholine
- GSH, glutathione
- Glc, glucose
- Gln, glutamine
- Glu, glutamate
- HE, hepatic encephalopathy
- Hyperammonaemia
- Ins, myo-inositol
- Lac, lactate
- MAP, mean arterial pressure
- NAA, N acetylaspartate
- NO, nitric oxide
- OP, ornithine phenylacetate
- Ornithine phenylacetate
- Oxygen
- PCho, phosphocholine
- PCr, phosphocreatine
- PE, phenylephrine
- Phenylephrine
- SPECIAL, spin echo full intensity acquired localised
- TE, echo time
- Tau, taurine
- VOI, volume of interest
- [18F]-FDG PET, [18F]-fluorodeoxyglucose positron emission tomography
- eNOS, endothelial nitric oxide synthase
- fMRI, functional magnetic resonance imaging
- hepatic encephalopathy
- mHE, minimal HE
- pCO2, partial pressure of carbon dioxide
- pO2, partial pressure of oxygen
- tCho, total choline
- tCr, total creatine
Collapse
Affiliation(s)
- Anna Hadjihambi
- UCL Institute for Liver and Digestive Health, Division of Medicine, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London, UK
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
- The Roger Williams Institute of Hepatology London, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Cristina Cudalbu
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Katarzyna Pierzchala
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Laboratory of Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Dunja Simicic
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Chris Donnelly
- Institute of Sports Science and Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Christos Konstantinou
- The Roger Williams Institute of Hepatology London, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Nathan Davies
- UCL Institute for Liver and Digestive Health, Division of Medicine, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London, UK
| | - Abeba Habtesion
- UCL Institute for Liver and Digestive Health, Division of Medicine, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London, UK
| | - Alexander V. Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Rajiv Jalan
- UCL Institute for Liver and Digestive Health, Division of Medicine, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London, UK
- European Foundation for the Study of Chronic Liver Failure
| | - Patrick S. Hosford
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
34
|
Drehmer E, Navarro-Moreno MÁ, Carrera-Juliá S, Moreno ML. A comparative study between olive oil and corn oil on oxidative metabolism. Food Funct 2022; 13:7157-7167. [PMID: 35699154 DOI: 10.1039/d2fo00919f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fats are an important part of diet, but not all lipids have the same structure and chemical properties. Unsaturated fatty acids have one or more double bonds in their structure and can be monounsaturated or polyunsaturated, respectively. Most vegetable oils, such as olive oil and corn oil, contain significant amounts of these fatty acids. The presence of double bonds in the molecule of a fatty acid constitutes vulnerable sites for oxidation reactions generating lipid peroxides, potentially toxic compounds that can cause cellular damage. In response to this oxidative damage, aerobic organisms have intracellular enzymatic antioxidant defense mechanisms. The aim of the present investigation was to study comparatively the effects of control liquid diets, of a defined composition, containing olive oil or corn oil as a lipid source respectively of monounsaturated and polyunsaturated fatty acids, on the oxidative metabolism of rats. Rats were divided into three groups which received a control animal feed diet (A.F.), olive oil liquid diet (O.O) and corn oil liquid diet (C.O) for 30 days. It was observed that the activity of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), increased in the liver and white fat tissue of rats fed with olive oil when compared to the corn oil group. However, in brown fat tissue and blood cells, the enzyme activities showed a tendency to decrease in the olive oil group. In addition, the effect of olive oil and corn oil on several glucose metabolism parameters (pyruvate, lactate, LDH, acetoacetate and beta-hydroxybutyrate) showed that corn oil impairs to a greater extent the cellular metabolism. All these results helped in concluding that some body tissues are more adversely affected than others by the administration of corn oil or olive oil, and their antioxidant defenses and cellular metabolism respond differently too.
Collapse
Affiliation(s)
- Eraci Drehmer
- Department of Health Sciences, Universidad Católica de Valencia "San Vicente Mártir", Valencia, Spain
| | | | - Sandra Carrera-Juliá
- Department of Nutrition and Dietetics, Universidad Católica de Valencia "San Vicente Mártir", Valencia, Spain
| | - Mari Luz Moreno
- Department of Human Physiology and Anatomy, Universidad Católica de Valencia "San Vicente Mártir", C/Ramiro de Maeztu, 14., 46900 Torrente, Valencia, Spain.
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW This article discusses neuroimaging in dementia diagnosis, with a focus on new applications of MRI and positron emission tomography (PET). RECENT FINDINGS Although the historical use of MRI in dementia diagnosis has been supportive to exclude structural etiologies, recent innovations allow for quantification of atrophy patterns that improve sensitivity for supporting the diagnosis of dementia causes. Neuronuclear approaches allow for localization of specific amyloid and tau neuropathology on PET and are available for clinical use, in addition to dopamine transporter scans in dementia with Lewy bodies and metabolic studies with fludeoxyglucose PET (FDG-PET). SUMMARY Using computerized software programs for MRI analysis and cross-sectional and longitudinal evaluations of hippocampal, ventricular, and lobar volumes improves sensitivity in support of the diagnosis of Alzheimer disease and frontotemporal dementia. MRI protocol requirements for such quantification are three-dimensional T1-weighted volumetric imaging protocols, which may need to be specifically requested. Fluid-attenuated inversion recovery (FLAIR) and 3.0T susceptibility-weighted imaging (SWI) sequences are useful for the detection of white matter hyperintensities as well as microhemorrhages in vascular dementia and cerebral amyloid angiopathy. PET studies for amyloid and/or tau pathology can add additional specificity to the diagnosis but currently remain largely inaccessible outside of research settings because of prohibitive cost constraints in most of the world. Dopamine transporter PET scans can help identify Lewy body dementia and are thus of potential clinical value.
Collapse
Affiliation(s)
- Cyrus A. Raji
- Washington University in St. Louis Mallinckrodt Institute of Radiology, Division of Neuroradiology
- Washington University in St. Louis Department of Neurology
- Washington University in St. Louis Neuroimaging Laboratories
- Knight Alzheimer Disease Research Center, Washington University in St. Louis
| | - Tammie L. S. Benzinger
- Washington University in St. Louis Mallinckrodt Institute of Radiology, Division of Neuroradiology
- Washington University in St. Louis Neuroimaging Laboratories
- Knight Alzheimer Disease Research Center, Washington University in St. Louis
- Washington University in St. Louis Department of Neurosurgery
| |
Collapse
|
36
|
Functional Imaging for Neurodegenerative Diseases. Presse Med 2022; 51:104121. [PMID: 35490910 DOI: 10.1016/j.lpm.2022.104121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/13/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
Diagnosis and monitoring of neurodegenerative diseases has changed profoundly over the past twenty years. Biomarkers are now included in most diagnostic procedures as well as in clinical trials. Neuroimaging biomarkers provide access to brain structure and function over the course of neurodegenerative diseases. They have brought new insights into a wide range of neurodegenerative diseases and have made it possible to describe some of the imaging challenges in clinical populations. MRI mainly explores brain structure while molecular imaging, functional MRI and electro- and magnetoencephalography examine brain function. In this paper, we describe and analyse the current and potential contribution of MRI and molecular imaging in the field of neurodegenerative diseases.
Collapse
|
37
|
Senthilkumar T, Kumarganesh S, Sivakumar P, Periyarselvam K. Primitive detection of Alzheimer’s disease using neuroimaging: A progression model for Alzheimer’s disease: Their applications, benefits, and drawbacks. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2022. [DOI: 10.3233/jifs-220628] [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
Alzheimer’s disease (A.D.) is the most widespread type of Dementia, and it is not a curable neurodegenerative disease that affects millions of older people. Researchers were able to use their understanding of Alzheimer’s disease risk variables to develop enrichment processes for longitudinal imaging studies. Using this method, they reduced their sample size and study time. This paper describes the primitive detective of Alzheimer’s diseases using Neuroimaging techniques. Several preprocessing methods were used to ensure that the dataset was ready for subsequent feature extraction and categorization. The noise was reduced by converting and averaging many scan frames from real to DCT space. Both sides of the averaged image were filtered and combined into a single shot after being converted to real space. InceptionV3 and DenseNet201 are two pre-trained models used in the suggested model. The PCA approach was used to select the traits, and the resulting explained variance ratio was 0.99The Simons Foundation Autism Research Initiative (SFARI)—Simon’s Simplex Collection (SSC)—and UCI machine learning datasets showed that our method is faster and more successful at identifying complete long-risk patterns when compared to existing methods.
Collapse
Affiliation(s)
- T. Senthilkumar
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| | | | - P. Sivakumar
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| | - K. Periyarselvam
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| |
Collapse
|
38
|
Goyzueta-Mamani LD, Chávez-Fumagalli MA, Alvarez-Fernandez K, Aguilar-Pineda JA, Nieto-Montesinos R, Davila Del-Carpio G, Vera-Lopez KJ, Lino Cardenas CL. Alzheimer's Disease: A Silent Pandemic - A Systematic Review on the Situation and Patent Landscape of the Diagnosis. Recent Pat Biotechnol 2022; 16:355-378. [PMID: 35400333 DOI: 10.2174/1872208316666220408114129] [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: 07/16/2021] [Revised: 01/13/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive impairment, tau protein deposits, and amyloid beta plaques. AD impacted 44 million people in 2016, and it is estimated to affect 100 million people by 2050. AD is disregarded as a pandemic compared with other diseases. To date, there is no effective treatment or diagnosis. OBJECTIVE We aimed to discuss the current tools used to diagnose COVID-19, to point out their potential to be adapted for AD diagnosis, and to review the landscape of existing patents in the AD field and future perspectives for AD diagnosis. METHOD We carried out a scientific screening following a research strategy in PubMed; Web of Science; the Derwent Innovation Index; the KCI-Korean Journal Database; SciELO; the Russian Science Citation index; and the CDerwent, EDerwent, and MDerwent index databases. RESULTS A total of 326 from 6,446 articles about AD and 376 from 4,595 articles about COVID-19 were analyzed. Of these, AD patents were focused on biomarkers and neuroimaging with no accurate, validated diagnostic methods, and only 7% of kit development patents were found. In comparison, COVID-19 patents were 60% about kit development for diagnosis; they are highly accurate and are now commercialized. CONCLUSION AD is still neglected and not recognized as a pandemic that affects the people and economies of all nations. There is a gap in the development of AD diagnostic tools that could be filled if the interest and effort that has been invested to tackle the COVID-19 emergency could also be applied for innovation.
Collapse
Affiliation(s)
- Luis Daniel Goyzueta-Mamani
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Miguel Angel Chávez-Fumagalli
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Karla Alvarez-Fernandez
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Jorge A Aguilar-Pineda
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Rita Nieto-Montesinos
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Gonzalo Davila Del-Carpio
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Karin J Vera-Lopez
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigacion, Universidad Catolica de Santa Maria, Arequipa, Peru
| | - Christian L Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
39
|
HMTM-Mediated Enhancement of Brain Bioenergetics in a Mouse Tauopathy Model Is Blocked by Chronic Administration of Rivastigmine. Biomedicines 2022; 10:biomedicines10040867. [PMID: 35453617 PMCID: PMC9029156 DOI: 10.3390/biomedicines10040867] [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: 03/21/2022] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 01/25/2023] Open
Abstract
The tau protein aggregation inhibitor hydromethylthionine mesylate (HMTM) was shown recently to have concentration-dependent pharmacological activity in delaying cognitive decline and brain atrophy in phase 3 Alzheimer’s disease (AD) clinical trials; the activity was reduced in patients receiving symptomatic therapies. The methylthionine (MT) moiety has been reported to increase the clearance of pathological tau and to enhance mitochondrial activity, which is impaired in AD patients. In line 1 (L1) mice (a model of AD), HMTM (5/15 mg/kg) was administered either as a monotherapy or as an add-on to a chronic administration with the cholinesterase inhibitor rivastigmine (0.1/0.5 mg/kg) to explore mitochondrial function and energy substrate utilization as potential targets of drug interference. Compared with wild-type NMRI mice, the L1 mice accumulated greater levels of l-lactate and of the LDH-A subunit responsible for the conversion of pyruvate into l-lactate. In contrast, the levels of LDH-B and mitochondrial ETC subunits and the activity of complexes I and IV was not altered in the L1 mice. The activity of complex I and complex IV tended to increase with the HMTM dosing, in turn decreasing l-lactate accumulation in the brains of the L1 mice, despite increasing the levels of LDH-A. The chronic pre-dosing of the L1 mice with rivastigmine partially prevented the enhancement of the activity of complexes I and IV by HMTM and the increase in the levels of LDH-A while further reducing the levels of l-lactate. Thus, HMTM in combination with rivastigmine leads to a depletion in the energy substrate l-lactate, despite bioenergetic production not being favoured. In this study, the changes in l-lactate appear to be regulated by LDH-A, since neither of the experimental conditions affected the levels of LDH-B. The data show that HMTM monotherapy facilitates the use of substrates for energy production, particularly l-lactate, which is provided by astrocytes, additionally demonstrating that a chronic pre-treatment with rivastigmine prevented most of the HMTM-associated effects.
Collapse
|
40
|
Lazarou I, Georgiadis K, Nikolopoulos S, Oikonomou VP, Stavropoulos TG, Tsolaki A, Kompatsiaris I, Tsolaki M. Exploring Network Properties Across Preclinical Stages of Alzheimer’s Disease Using a Visual Short-Term Memory and Attention Task with High-Density Electroencephalography: A Brain-Connectome Neurophysiological Study. J Alzheimers Dis 2022; 87:643-664. [DOI: 10.3233/jad-215421] [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: Visual short-term memory (VSTMT) and visual attention (VAT) exhibit decline in the Alzheimer’s disease (AD) continuum; however, network disruption in preclinical stages is scarcely explored. Objective: To advance our knowledge about brain networks in AD and discover connectivity alterations during VSTMT and VAT. Methods: Twelve participants with AD, 23 with mild cognitive impairment (MCI), 17 with subjective cognitive decline (SCD), and 21 healthy controls (HC) were examined using a neuropsychological battery at baseline and follow-up (three years). At baseline, the subjects were examined using high density electroencephalography while performing a VSTMT and VAT. For exploring network organization, we constructed weighted undirected networks and examined clustering coefficient, strength, and betweenness centrality from occipito-parietal regions. Results: One-way ANOVA and pair-wise t-test comparisons showed statistically significant differences in HC compared to SCD (t (36) = 2.43, p = 0.026), MCI (t (42) = 2.34, p = 0.024), and AD group (t (31) = 3.58, p = 0.001) in Clustering Coefficient. Also with regards to Strength, higher values for HC compared to SCD (t (36) = 2.45, p = 0.019), MCI (t (42) = 2.41, p = 0.020), and AD group (t (31) = 3.58, p = 0.001) were found. Follow-up neuropsychological assessment revealed converge of 65% of the SCD group to MCI. Moreover, SCD who were converted to MCI showed significant lower values in all network metrics compared to the SCD that remained stable. Conclusion: The present findings reveal that SCD exhibits network disorganization during visual encoding and retrieval with intermediate values between MCI and HC.
Collapse
Affiliation(s)
- Ioulietta Lazarou
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
- 1 Department of Neurology, G.H. “AHEPA”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Makedonia, Greece
| | - Kostas Georgiadis
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
- Informatics Department, Aristotle University of Thessaloniki, Makedonia, Greece
| | - Spiros Nikolopoulos
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
| | - Vangelis P. Oikonomou
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
| | - Thanos G. Stavropoulos
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
| | - Anthoula Tsolaki
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
- Greek Association of Alzheimer’s Disease and Related Disorders, Thessaloniki, Makedonia, Greece
| | - Ioannis Kompatsiaris
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
| | - Magda Tsolaki
- Information Technologies Institute, Centre for Research and Technology Hellas (CERTH-ITI), Thessaloniki, Makedonia, Greece
- 1 Department of Neurology, G.H. “AHEPA”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Makedonia, Greece
- Greek Association of Alzheimer’s Disease and Related Disorders, Thessaloniki, Makedonia, Greece
| | | |
Collapse
|
41
|
Boots A, Thomason ME, Espinoza-Heredia C, Pruitt PJ, Damoiseaux JS, Roseboom TJ, de Rooij SR. Sex-specific effects of prenatal undernutrition on resting-state functional connectivity in the human brain at age 68. Neurobiol Aging 2022; 112:129-138. [PMID: 35151035 PMCID: PMC9459445 DOI: 10.1016/j.neurobiolaging.2022.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022]
Abstract
Prenatal nutrition may significantly impact brain aging. Results from the Dutch Famine Birth Cohort indicated that prenatal undernutrition is negatively associated with cognition, brain volumes, perfusion and structural brain aging in late life, predominantly in men. This study investigates the association between prenatal undernutrition and late-life functional brain network connectivity. In an exploratory resting-state functional magnetic resonance imaging study of 112 participants from the Dutch Famine Birth Cohort, we investigated whether the within- and between-network functional connectivity of the default mode network, salience network and central executive network differ at age 68 in men (N = 49) and women (N = 63) either exposed or unexposed to undernutrition in early gestation. Additionally, we explored sex-specific effects. Compared to unexposed participants, exposed participants revealed multiple clusters of different functional connectivity within and between the three networks studied. Sex-specific analyses suggested a pattern of network desegregation fitting with brain aging in men and a more diffuse pattern of group differences in women. This study demonstrates that associations between prenatal undernutrition and brain network functional connectivity extend late into life.
Collapse
Affiliation(s)
- Amber Boots
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
| | - Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University Langone Health, New York, NY, USA; Department of Population Health, New York University Langone Health, New York, NY, USA; Neuroscience Institute, New York University Langone Health, New York, NY, USA
| | - Claudia Espinoza-Heredia
- Department of Child and Adolescent Psychiatry, New York University Langone Health, New York, NY, USA
| | - Patrick J Pruitt
- Institute of Gerontology, Wayne State University, Detroit, MI, USA
| | - Jessica S Damoiseaux
- Institute of Gerontology, Wayne State University, Detroit, MI, USA; Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Tessa J Roseboom
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands; Department of Obstetrics and Gynaecology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Isella V, Crivellaro C, Formenti A, Musarra M, Pacella S, Morzenti S, Ferri F, Mapelli C, Gallivanone F, Guerra L, Appollonio I, Ferrarese C. Validity of cingulate–precuneus–temporo-parietal hypometabolism for single-subject diagnosis of biomarker-proven atypical variants of Alzheimer’s Disease. J Neurol 2022; 269:4440-4451. [PMID: 35347453 PMCID: PMC9293827 DOI: 10.1007/s00415-022-11086-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/14/2022]
Abstract
The aim of our study was to establish empirically to what extent reduced glucose uptake in the precuneus, posterior cingulate and/or temporo-parietal cortex (PCTP), which is thought to indicate brain amyloidosis in patients with dementia or MCI due to Alzheimer’s Disease (AD), permits to distinguish amyloid-positive from amyloid-negative patients with non-classical AD phenotypes at the single-case level. We enrolled 127 neurodegenerative patients with cognitive impairment and a positive (n. 63) or negative (n. 64) amyloid marker (cerebrospinal fluid or amy-PET). Three rating methods of FDG-PET scan were applied: purely qualitative visual interpretation of uptake images (VIUI), and visual reading assisted by a semi-automated and semi-quantitative tool: INLAB, provided by the Italian National Research Council, or Cortex ID Suite, marketed by GE Healthcare. Fourteen scans (11.0%) patients remained unclassified by VIUI or INLAB procedures, therefore, validity values were computed on the remaining 113 cases. The three rating approaches showed good total accuracy (77–78%), good to optimal sensitivity (81–93%), but poorer specificity (62–75%). VIUI showed the highest sensitivity and the lowest specificity, and also the highest proportion of unclassified cases. Cases with asymmetric temporo-parietal hypometabolism and a progressive aphasia or corticobasal clinical profile, in particular, tended to be rated as AD-like, even if biomarkers indicated non-amyloid pathology. Our findings provide formal support to the value of PCTP hypometabolism for single-level diagnosis of amyloid pathophysiology in atypical AD, but also highlight the risk of qualitative assessment to misclassify patients with non-AD PPA or CBS underpinned by asymmetric temporo-parietal hypometabolism.
Collapse
|
43
|
Moran C, Than S, Callisaya M, Beare R, Srikanth V. New Horizons-Cognitive Dysfunction Associated With Type 2 Diabetes. J Clin Endocrinol Metab 2022; 107:929-942. [PMID: 34788847 DOI: 10.1210/clinem/dgab797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 11/19/2022]
Abstract
The prevalence of type 2 diabetes (T2D) and cognitive dysfunction increases with age. As society ages, clinicians will be increasingly tasked with managing older people who have both T2D and cognitive dysfunction. T2D is associated with an increased risk of cognitive dysfunction and hence there is increasing interest in whether T2D is a causal factor in the pathogenesis of cognitive decline and dementia. Recent advances in the use of sensitive measures of in vivo brain dysfunction in life-course studies can help understand potential mechanistic pathways and also help guide recommendations for clinical practice. In this article we will describe new horizons in the understanding of cognitive dysfunction associated with T2D. Coming from a clinical perspective, we discuss potential mechanisms and pathways linking the 2 conditions and the contribution of multimodal neuroimaging and study designs to advancing understanding in the field. We also highlight the important issues on the horizon that will need addressing in clinical identification, management, and risk reduction for people with coexistent T2D and cognitive dysfunction.
Collapse
Affiliation(s)
- Chris Moran
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Alfred Health, Melbourne, 3004 Victoria, Australia
| | - Stephanie Than
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
| | - Michele Callisaya
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| | - Richard Beare
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, 3052 Victoria, Australia
| | - Velandai Srikanth
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| |
Collapse
|
44
|
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
|
45
|
Wimalarathne D, Ruan W, Sun X, Liu F, Gai Y, Liu Q, Hu F, Lan X. Impact of TOF on Brain PET With Short-Lived 11C-Labeled Tracers Among Suspected Patients With AD/PD: Using Hybrid PET/MRI. Front Med (Lausanne) 2022; 9:823292. [PMID: 35308534 PMCID: PMC8926006 DOI: 10.3389/fmed.2022.823292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To explore the impact of the time-of-flight (TOF) reconstruction on brain PET with short-lived 11C-labeled tracers in PET magnetic resonance (PET/MR) brain images among suspected patients with Alzheimer's and Parkinson's disease (AD/PD). Methods Patients who underwent 11C-2-ß-carbomethoxy-3-b-(4-fluorophenyl) tropane (11C-CFT) and 2-(4-N-[11C] methylaminophenyl)-6-hydroxybenzothiazole (11C-PiB) PET/MRI were retrospectively included in the study. Each PET LIST mode data were reconstructed with and without the TOF reconstruction algorithm. Standard uptake values (SUVs) of Caudate Nucleus (CN), Putamen (PU), and Whole-brain (WB) were measured. TOF and non-TOF SUVs were assessed by using paired t-test. Standard formulas were applied to measure contrast, signal-to-noise ratio (SNR), and percentage relative average difference of SUVs (%RAD-SUVs). Results Total 75 patients were included with the median age (years) and body mass index (BMI-kg/m2) of 60.2 ± 10.9 years and 23.9 ± 3.7 kg/m2 in 11C-CFT (n = 41) and 62.2 ± 6.8 years and 24.7 ± 2.9 kg/m2 in 11C-PiB (n = 34), respectively. Higher average SUVs and positive %RAD-SUVs were observed in CN and PU in TOF compared with non-TOF reconstructions for the two 11C-labeled radiotracers. Differences of SUVmean were significant (p < 0.05) in CN and PU for both 11C-labeled radiotracers. SUVmax was enhanced significantly in CN and PU for 11C-CFT and CN for 11C-PiB, but not in PU. Significant contrast enhancement was observed in PU for both 11C-labeled radiotracers, whereas SNR gain was significant in PU, only for 11C-PiB in TOF reconstruction. Conclusion Time-of-flight leads to a better signal vs. noise trade-off than non-TOF in 11C-labeled tracers between CN and PU, improving the SUVs, contrast, and SNR, which were valuable for reducing injected radiation dose. Improved timing resolution aided the rapid decay rate of short-lived 11C-labeled tracers, and it shortened the scan time, increasing the patient comfort, and reducing the motion artifact among patients with AD/PD. However, one should adopt the combined TOF algorithm with caution for the quantitative analysis because it has different effects on the SUVmax, contrast, and SNR of different brain regions.
Collapse
Affiliation(s)
- D.D.N Wimalarathne
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Radiography and Radiotherapy, Faculty of Allied Health Sciences, General Sir John Kotelawala Defence University, Rathmalana, Sri Lanka
| | - Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xun Sun
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Vodičková A, Koren SA, Wojtovich AP. Site-specific mitochondrial dysfunction in neurodegeneration. Mitochondrion 2022; 64:1-18. [PMID: 35182728 PMCID: PMC9035127 DOI: 10.1016/j.mito.2022.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
Abstract
Mitochondria are essential for neuronal survival and mitochondrial dysfunction is a hallmark of neurodegeneration. The loss in mitochondrial energy production, oxidative stress, and changes in calcium handling are associated with neurodegenerative diseases; however, different sites and types of mitochondrial dysfunction are linked to distinct neuropathologies. Understanding the causal or correlative relationship between changes in mitochondria and neuropathology will lead to new therapeutic strategies. Here, we summarize the evidence of site-specific mitochondrial dysfunction and mitochondrial-related clinical trials for neurodegenerative diseases. We further discuss potential therapeutic approaches, such as mitochondrial transplantation, restoration of mitochondrial function, and pharmacological alleviation of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Anežka Vodičková
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Shon A Koren
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Andrew P Wojtovich
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA; Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
47
|
Lange C, Mäurer A, Suppa P, Apostolova I, Steffen IG, Grothe MJ, Buchert R. Brain FDG PET for Short- to Medium-Term Prediction of Further Cognitive Decline and Need for Assisted Living in Acutely Hospitalized Geriatric Patients With Newly Detected Clinically Uncertain Cognitive Impairment. Clin Nucl Med 2022; 47:123-129. [PMID: 35006106 DOI: 10.1097/rlu.0000000000003981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE The aim of this study was to evaluate brain FDG PET for short- to medium-term prediction of cognitive decline, need for assisted living, and survival in acutely hospitalized geriatric patients with newly detected clinically uncertain cognitive impairment (CUCI). MATERIALS AND METHODS The study included 96 patients (62 females, 81.4 ± 5.4 years) hospitalized due to (sub)acute admission indications with newly detected CUCI (German Clinical Trials Register DRKS00005041). FDG PET was categorized as "neurodegenerative" (DEG+) or "nonneurodegenerative" (DEG-) based on visual inspection by 2 independent readers. In addition, each individual PET was tested voxel-wise against healthy controls (P < 0.001 uncorrected). The resulting total hypometabolic volume (THV) served as reader-independent measure of the spatial extent of neuronal dysfunction/degeneration. FDG PET findings at baseline were tested for association with the change in living situation and change in vital status 12 to 24 months after PET. The association with the annual change of the CDR-SB (Clinical Dementia Rating Sum of Boxes) after PET was tested in a subsample of 72 patients. RESULTS The mean time between PET and follow-up did not differ between DEG+ and DEG- patients (1.37 ± 0.27 vs 1.41 ± 0.27 years, P = 0.539). Annual change of CDR-SB was higher in DEG+ compared with DEG- patients (2.78 ± 2.44 vs 0.99 ± 1.81, P = 0.001), and it was positively correlated with THV (age-corrected Spearman ρ = 0.392, P = 0.001). DEG+ patients moved from at home to assisted living significantly earlier than DEG- patients (P = 0.050). Survival was not associated with DEG status or with THV. CONCLUSIONS In acutely hospitalized geriatric patients with newly detected CUCI, the brain FDG PET can contribute to the prediction of further cognitive/functional decline and the need for assisted living within 1 to 2 years.
Collapse
Affiliation(s)
- Catharina Lange
- From the Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin
| | | | | | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Ingo G Steffen
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg
| |
Collapse
|
48
|
Metabolic Features of Brain Function with Relevance to Clinical Features of Alzheimer and Parkinson Diseases. Molecules 2022; 27:molecules27030951. [PMID: 35164216 PMCID: PMC8839962 DOI: 10.3390/molecules27030951] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Brain metabolism is comprised in Alzheimer’s disease (AD) and Parkinson’s disease (PD). Since the brain primarily relies on metabolism of glucose, ketone bodies, and amino acids, aspects of these metabolic processes in these disorders—and particularly how these altered metabolic processes are related to oxidative and/or nitrosative stress and the resulting damaged targets—are reviewed in this paper. Greater understanding of the decreased functions in brain metabolism in AD and PD is posited to lead to potentially important therapeutic strategies to address both of these disorders, which cause relatively long-lasting decreased quality of life in patients.
Collapse
|
49
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
50
|
Odano I, Maeyatsu F, Asari M, Yamaguchi S, Miura T, Taki Y. Whole-body vibration exercise and training increase regional CBF in mild cognitive impairment with enhanced cognitive function. Ann Nucl Med 2022; 36:82-94. [PMID: 34762232 DOI: 10.1007/s12149-021-01687-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES Preclinical and non-medicinal interventions are essential for preventing and treating cognitive decline in patients with mild cognitive impairment (MCI). Whole-body vibration (WBV) exercise is conducted on a platform that generates vertical sinusoidal vibrations, and WBV training may improve regional cerebral blood flow (rCBF) and cognitive function, however, the underlying mechanism remains unclear. The aim of the present study was to investigate whether WBV exercise and a 24-week WBV training protocol increased rCBF and enhanced cognitive function in patients with amnestic MCI (aMCI). METHODS [99mTc]-ECD and SPECT studies were performed on 16 aMCI patients at baseline, during WBV exercise, and on 6 of the 16 patients after 24-week WBV training. To diagnose SPECT images and select the patients, a Z-score mapping approach was used, which revealed pathological hypoperfusion in the parietal association cortex, precuneus and/or posterior cingulate gyrus for MCI at baseline. rCBF was semi-quantitatively measured and underestimation in the high flow range was corrected. Since it is difficult to quantitatively measure rCBF during WBV exercise, the rCBFratio was obtained by standardizing with the average of individual mean SPECT counts with correcting underestimation in the high flow range. The rCBFratios at baseline and after WBV training were also obtained in a similar manner. Since the changes in rCBF were regarded as corresponding to the changes in rCBFratio, the ratios were compared. Cognitive function was also evaluated and compared. RESULTS We found that the rCBFratio changed with an average range of 11.5% during WBV exercise, and similar changes were observed after 24-week WBV training with a 13.0% change, resulting in improved cognitive function (MoCA-J, P = 0.028). The rCBFratio increased in the parietal association cortex and occipital lobes, including the precuneus and posterior cingulate gyrus, at which hypoperfusion was detected at baseline, but decreased in the frontal lobe and anterior cingulate gyrus. The rCBFratio increased on the right side of several motion-suppressive nuclei by WBV exercise; the bilateral red nuclei and right medial globus pallidus by WBV training. CONCLUSION WBV exercise and training increase rCBF in aMCI patients, and WBV training enhances cognitive function and may increase the cognitive reserve. Further investigation is necessary.
Collapse
Affiliation(s)
- Ikuo Odano
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan.
- Department of Neurology and Radiology, Miyagi Koseikyokai, Izumi Hospital, Sendai, Japan.
| | - Fumio Maeyatsu
- Department of Neurology and Radiology, Miyagi Koseikyokai, Izumi Hospital, Sendai, Japan
| | - Mami Asari
- Department of Neurology and Radiology, Miyagi Koseikyokai, Izumi Hospital, Sendai, Japan
| | - Sayaka Yamaguchi
- Department of Neurology and Radiology, Miyagi Koseikyokai, Izumi Hospital, Sendai, Japan
| | - Tsukasa Miura
- Department of Neurology and Radiology, Miyagi Koseikyokai, Izumi Hospital, Sendai, Japan
| | - Yasuyuki Taki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|