1
|
Querry M, Botzung A, Cretin B, Demuynck C, Muller C, Ravier A, Schorr B, Mondino M, Sanna L, de Sousa PL, Philippi N, Blanc F. Neuroanatomical substrates of depression in dementia with Lewy bodies and Alzheimer's disease. GeroScience 2024; 46:5725-5744. [PMID: 38750385 PMCID: PMC11493943 DOI: 10.1007/s11357-024-01190-4] [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: 11/23/2023] [Accepted: 05/01/2024] [Indexed: 10/23/2024] Open
Abstract
Dementia with Lewy bodies (DLB) and Alzheimer's disease (AD) are often associated with depressive symptoms from the prodromal stage. The aim of the present study was to investigate the neuroanatomical correlates of depression in prodromal to mild DLB patients compared with AD patients. Eighty-three DLB patients, 37 AD patients, and 18 healthy volunteers were enrolled in this study. Depression was evaluated with the Mini International Neuropsychiatric Interview (MINI), French version 5.0.0. T1-weighted three-dimensional anatomical images were acquired for all participants. Regression and comparison analyses were conducted using a whole-brain voxel-based morphometry (VBM) approach on the grey matter volume (GMV). DLB patients presented a significantly higher mean MINI score than AD patients (p = 0.004), 30.1% of DLB patients had clinical depression, and 56.6% had a history of depression, while 0% of AD patients had clinical depression and 29.7% had a history of depression. VBM regression analyses revealed negative correlations between the MINI score and the GMV of right prefrontal regions in DLB patients (p < 0.001, uncorrected). Comparison analyses between DLB patients taking and those not taking an antidepressant mainly highlighted a decreased GMV in the bilateral middle/inferior temporal gyrus (p < 0.001, uncorrected) in treated DLB patients. In line with the literature, our behavioral analyses revealed higher depression scores in DLB patients than in AD patients. We also showed that depressive symptoms in DLB are associated with decreased GMV in right prefrontal regions. Treated DLB patients with long-standing depression would be more likely to experience GMV loss in the bilateral middle/inferior temporal cortex. These findings should be taken into account when managing DLB patients.
Collapse
Affiliation(s)
- Manon Querry
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France.
| | - Anne Botzung
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Benjamin Cretin
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
- CM2R, Neuropsychology Unit, Neurology Department, Head and Neck Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Catherine Demuynck
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Candice Muller
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Alix Ravier
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Benoît Schorr
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Mary Mondino
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
| | - Léa Sanna
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Paulo Loureiro de Sousa
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
| | - Nathalie Philippi
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
- CM2R, Neuropsychology Unit, Neurology Department, Head and Neck Division, University Hospitals of Strasbourg, Strasbourg, France
| | - Frédéric Blanc
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatric Day Hospital, Geriatrics Division, University Hospitals of Strasbourg, Strasbourg, France
| |
Collapse
|
2
|
Dubois B, Villain N, Schneider L, Fox N, Campbell N, Galasko D, Kivipelto M, Jessen F, Hanseeuw B, Boada M, Barkhof F, Nordberg A, Froelich L, Waldemar G, Frederiksen KS, Padovani A, Planche V, Rowe C, Bejanin A, Ibanez A, Cappa S, Caramelli P, Nitrini R, Allegri R, Slachevsky A, de Souza LC, Bozoki A, Widera E, Blennow K, Ritchie C, Agronin M, Lopera F, Delano-Wood L, Bombois S, Levy R, Thambisetty M, Georges J, Jones DT, Lavretsky H, Schott J, Gatchel J, Swantek S, Newhouse P, Feldman HH, Frisoni GB. Alzheimer Disease as a Clinical-Biological Construct-An International Working Group Recommendation. JAMA Neurol 2024:2825806. [PMID: 39483064 DOI: 10.1001/jamaneurol.2024.3770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Importance Since 2018, a movement has emerged to define Alzheimer disease (AD) as a purely biological entity based on biomarker findings. The recent revision of the Alzheimer Association (AA) criteria for AD furthers this direction. However, concerns about a purely biological definition of AD being applied clinically, the understanding of AD by society at large, and the translation of blood-based biomarkers into clinical practice prompt these International Working Group (IWG) updated recommendations. Objective To consider the revised AA criteria and to offer an alternative definitional view of AD as a clinical-biological construct for clinical use. The recommendations of the 2021 IWG diagnostic criteria are updated for further elaborating at-risk and presymptomatic states. Evidence Review PubMed was searched for articles published between July 1, 2020, and March 1, 2024, using the terms "biomarker" OR "amyloid" OR "tau" OR "neurodegeneration" OR "preclinical" OR "CSF" OR "PET" OR "plasma" AND "Alzheimer's disease." The references of relevant articles were also searched. Findings In the new AA diagnostic criteria, AD can be defined clinically as encompassing cognitively normal people having a core 1 AD biomarker. However, recent literature shows that the majority of biomarker-positive cognitively normal individuals will not become symptomatic along a proximate timeline. In the clinical setting, disclosing a diagnosis of AD to cognitively normal people with only core 1 AD biomarkers represents the most problematic implication of a purely biological definition of the disease. Conclusions and Relevance The ultimate aim of the field was to foster effective AD treatments, including preventing symptoms and dementia. The approach of diagnosing AD without a clinical and biological construct would be unwarranted and potentially concerning without a clear knowledge of when or whether symptoms will ever develop. It is recommended that those who are amyloid-positive only and, more generally, most biomarker-positive cognitively normal individuals, should not be labeled as having AD. Rather, they should be considered as being at risk for AD. The expansion of presymptomatic AD is viewed as a better diagnostic construct for those with a specific pattern of biomarkers, indicating that they are proximate to the expression of symptoms in the near future.
Collapse
Affiliation(s)
- Bruno Dubois
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, FrontLab, Paris, France
| | - Nicolas Villain
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, Maladie d'Alzheimer, Maladies à Prions, Paris, France
| | - Lon Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles
| | - Nick Fox
- Department of Neurodegenerative Disease, Dementia Research Centre, and the United Kingdom Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Noll Campbell
- Purdue University College of Pharmacy, West Lafayette, Indiana
- Purdue University Center for Aging and the Life Course, West Lafayette, Indiana
- Indiana University Center for Aging Research, Indianapolis
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Bernard Hanseeuw
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Institute of Neurosciences, UC Louvain, Brussels, Belgium
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Bioengineering, University College London, London, United Kingdom
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, The Aging Brain, Karolinska University Hospital, Stockholm, Sweden
| | - Lutz Froelich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alessandro Padovani
- Neurology and Neurophysiology Section, Department Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Hospital Department of Continuità di Cura e Fragilità, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Vincent Planche
- Univ. Bordeaux, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources et de Recherche, CHU de Bordeaux, Bordeaux, France
| | - Christopher Rowe
- Department of Molecular Imaging and Therapy, Austin Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Stefano Cappa
- University School for Advanced Studies, Pavia, Italy
- RCCS Mondino Foundation, Pavia, Italy
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Nitrini
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Allegri
- Department of Cognitive Neurology, Fleni Neurological Institute, Buenos Aires, Argentina
- Department of Cognitive Neurosciences, Universidad de la Costa, Barranquilla, Colombia
| | - Andrea Slachevsky
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Memory and Neuropsychiatric Center Neurology Department, Hospital del Salvador and Neuropsychology and Clinical Neuroscience Laboratory, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Chile
- Neurology and Psychiatry Department, Clínica Alemana-Universidad Desarrollo, Santiago, Chile
| | - Leonardo Cruz de Souza
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andrea Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill
| | - Eric Widera
- Division of Geriatrics, University of California, San Francisco
- Hospice & Palliative Care, San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Craig Ritchie
- Brain Health and Neurodegenerative Medicine, University of St Andrews, Scotland, United Kingdom
- Scottish Brain Sciences, Edinburgh, Scotland, United Kingdom
| | - Marc Agronin
- Medical Office for MIND Institute, Miami, Florida
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Lisa Delano-Wood
- Veterans Affairs San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego Health, La Jolla, California
- Center for Stress and Mental Health, VA San Diego Healthcare System, San Diego, California
| | - Stéphanie Bombois
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
| | - Richard Levy
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, FrontLab, Paris, France
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Helen Lavretsky
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior
- David Geffen School of Medicine, University of California, Los Angeles
| | - Jonathan Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jennifer Gatchel
- Department of Psychiatry, Massachusetts General Hospital, Boston
- McLean Hospital, Belmont, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Psychiatry, Baylor College of Medicine, Houston, Texas
- Michael E. DeBakey VA Medical Center, Houston, Texas
| | - Sandra Swantek
- American Association for Geriatric Psychiatry, Brentwood, Tennessee
| | - Paul Newhouse
- Vanderbilt University, Nashville, Tennessee
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, Tennessee
- VA-TVHS Geriatric Research Education and Clinical Center, Nashville, Tennessee
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, La Jolla
- Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Luikku AJ, Nerg O, Koivisto AM, Hänninen T, Junkkari A, Kemppainen S, Juopperi SP, Sinisalo R, Pesola A, Soininen H, Hiltunen M, Leinonen V, Rauramaa T, Martiskainen H. Deep learning assisted quantitative analysis of Aβ and microglia in patients with idiopathic normal pressure hydrocephalus in relation to cognitive outcome. J Neuropathol Exp Neurol 2024; 83:967-978. [PMID: 39101555 PMCID: PMC11487103 DOI: 10.1093/jnen/nlae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Abstract
Neuropathologic changes of Alzheimer disease (AD) including Aβ accumulation and neuroinflammation are frequently observed in the cerebral cortex of patients with idiopathic normal pressure hydrocephalus (iNPH). We created an automated analysis platform to quantify Aβ load and reactive microglia in the vicinity of Aβ plaques and to evaluate their association with cognitive outcome in cortical biopsies of patients with iNPH obtained at the time of shunting. Aiforia Create deep learning software was used on whole slide images of Iba1/4G8 double immunostained frontal cortical biopsies of 120 shunted iNPH patients to identify Iba1-positive microglia somas and Aβ areas, respectively. Dementia, AD clinical syndrome (ACS), and Clinical Dementia Rating Global score (CDR-GS) were evaluated retrospectively after a median follow-up of 4.4 years. Deep learning artificial intelligence yielded excellent (>95%) precision for tissue, Aβ, and microglia somas. Using an age-adjusted model, higher Aβ coverage predicted the development of dementia, the diagnosis of ACS, and more severe memory impairment by CDR-GS whereas measured microglial densities and Aβ-related microglia did not correlate with cognitive outcome in these patients. Therefore, cognitive outcome seems to be hampered by higher Aβ coverage in cortical biopsies in shunted iNPH patients but is not correlated with densities of surrounding microglia.
Collapse
Affiliation(s)
- Antti J Luikku
- Institute of Clinical Medicine—Neurosurgery, University of Eastern Finland, Kuopio, Finland
- Neurosurgery of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Ossi Nerg
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine—Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anne M Koivisto
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine—Neurology, University of Eastern Finland, Kuopio, Finland
- Department of Neurosciences, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Department of Geriatrics/Rehabilitation and Internal Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Tuomo Hänninen
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine—Neurology, University of Eastern Finland, Kuopio, Finland
| | - Antti Junkkari
- Neurosurgery of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | - Rosa Sinisalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Alli Pesola
- Institute of Clinical Medicine—Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine—Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ville Leinonen
- Institute of Clinical Medicine—Neurosurgery, University of Eastern Finland, Kuopio, Finland
- Neurosurgery of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine—Pathology, University of Eastern Finland, Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
De Simone MS, Lombardi MG, De Tollis M, Perri R, Fadda L, Caltagirone C, Carlesimo GA. Forgetting rate for the familiarity and recollection components of recognition in amnestic mild cognitive impairment: A longitudinal study. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:1411-1423. [PMID: 36264763 DOI: 10.1080/23279095.2022.2135441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Here we aimed to investigate the rate of forgetting of the familiarity and recollection components of recognition in patients at the onset of medial temporal lobe (MTL) pathology and destined to convert to Alzheimer's disease (AD). For this purpose, we conducted a longitudinal study of 13 patients who were diagnosed with amnestic mild cognitive impairment (a-MCI) at the first assessment and followed-up for 3 years. During this time, five patients converted to AD and eight remained in a stable condition of cognitive impairment. A group of 15 healthy subjects were enrolled as the control group (HC). In order to separately quantify the contribution of recollection and familiarity to recognition memory performance, the experimental sample was submitted to a modified version of Huppert and Piercy's procedure that included a Remember/Know paradigm. Data demonstrated that both stable and converter a-MCI patients forgot memory traces relative to the familiarity components of recognition at the same rate as HC. Conversely, converter a-MCI patients showed accelerated long-term forgetting specifically for the recollection component of recognition compared to stable a-MCI and HC. This is the first empirical demonstration that familiarity and recollection components of declarative memory are subject to different rates of forgetting in a-MCI patients as a function of their longitudinal clinical outcome. Our finding of accelerated long-term forgetting of the recollection component of recognition disclosed by converter a-MCI patients suggests that atrophy in the MTL not only interferes with the storage aspects but also disrupts the consolidation of memory traces.
Collapse
Affiliation(s)
- Maria Stefania De Simone
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Giovanna Lombardi
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Massimo De Tollis
- Technology and Training Methods for Disability Care Laboratory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Roberta Perri
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Lucia Fadda
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Carlo Caltagirone
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
- Technology and Training Methods for Disability Care Laboratory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giovanni Augusto Carlesimo
- Laboratory of Neuropsychology of Memory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| |
Collapse
|
5
|
Golipoor M, Rafat Z, Saberi A, Roostaei D, Shabanpour AM. Comparing the frequency, antifungal susceptibility, and enzymatic profiles of the oral fungal composition in patients with and without Alzheimer's disease admitted to a neurology clinic. Front Cell Infect Microbiol 2024; 14:1477230. [PMID: 39492992 PMCID: PMC11527782 DOI: 10.3389/fcimb.2024.1477230] [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: 08/08/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Background Studies have shown that changes in the frequency of oral microorganisms may play a key role in the development of Alzheimer's disease (AD). However, no research has been conducted on the oral fungal composition in AD-patients. The present study aimed to investigate the changes in the frequency of oral fungal composition, the antifungal susceptibility, and the enzymatic profiles of oral fungal composition in patients suffering from AD compared to non-AD individuals. Materials and methods In the present analytical cross-sectional study during 12 months, 76 hospitalized patients with AD were matched with 76 individuals without AD. A sterile serum physiology-moistened cotton-tipped swab was used to sample the mouth area. All swabs were cultured on Sabouraud Chloramphenicol Agar. Fungal identified were confirmed through the PCR-sequencing techniques. Enzyme activity index (EAI) for important pathogenic factors including proteinase, esterase and hemolysin was measured using relevant protocols. The susceptibility to 8 antifungal agents (nystatin, voriconazole, itraconazole, fluconazole, posaconazole, amphotericin B, 5-fluorocytosine, and caspofungin) against fungal strains obtained from AD-patients was evaluated according to the Clinical and Laboratory Standards Institute (CLSI) guidelines, document M38-A2 for filamentous fungi, and document M27-A4 for yeasts. Results The results showed that compared to the non-AD individuals, the prevalence of oral fungal composition in AD group was 1.6 times higher. Candida albicans was the most common fungal species isolated from oral swab samples of AD group (n=53, 80%) and non-AD group (n=28, 40%), and the diversity of the oral fungal composition in AD-patients were lower than non-AD individuals. Among the 3 investigated virulence factors, a statistically significant difference was shown in terms of hemolysin activity level between the two studied groups (p<0.05) and the activity level of esterase and proteinase enzymes did not show a significant difference in the two studied groups (p>0.05). The results showed that almost all of the tested isolates were susceptible to nystatin, the most widely prescribed antifungal to treat superficial infections, and only 1.69 % (2/118) of the Candida isolates were resistant to this antifungal drug. Conclusion Understanding the changes in the frequency of oral fungal composition the antifungal susceptibility, and the enzymatic profiles of oral fungal composition in patients suffering from AD compared to non-AD individuals makes it possible to better understand the etiology of this disease.
Collapse
Affiliation(s)
- Mandana Golipoor
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Rafat
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alia Saberi
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Roostaei
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir-Mohammad Shabanpour
- Student research committee, Anzali International Medical Campus, Guilan University of Medical Sciences, Guilan, Iran
| |
Collapse
|
6
|
Chen ZY, Shi Q, Xiao K, Kong Y, Liang DL, Wang YH, Min R, Zhang J, Wang Z, Ye H, Gao R, Chu M, Nan HT, Jiang DM, Li JJ, Wang L, Zou WQ, Wu LY, Dong XP. Multisite Skin Biopsies vs Cerebrospinal Fluid for Prion Seeding Activity in the Diagnosis of Prion Diseases. JAMA Neurol 2024:2824416. [PMID: 39401015 DOI: 10.1001/jamaneurol.2024.3458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Importance Recent studies have revealed that autopsy skin samples from cadavers with prion diseases (PRDs) exhibited a positive prion seeding activity similar to cerebrospinal fluid (CSF). It is worthwhile to validate the findings with a large number of biopsy skin samples and compare the clinical value of prion seeding activity between skin biopsies and concurrent CSF specimens. Objective To compare the prion seeding activity of skin biopsies and CSF samples and to determine the effectiveness of combination of the skin biopsies from multiple sites and numerous dilutions on the diagnosis for various types of PRDs. Design, Setting, and Participants In the exploratory cohort, patients were enrolled from September 15, 2021, to December 15, 2023, and were followed up every 3 months until April 2024. The confirmatory cohort enrolled patients from December 16, 2023, to June 31, 2024. The exploratory cohort was conducted at a single center, the neurology department at Xuanwu Hospital. The confirmatory cohort was a multicenter study involving 4 hospitals in China. Participants included those diagnosed with probable sporadic Creutzfeldt-Jakob disease or genetically confirmed PRDs. Patients with uncertain diagnoses or those lost to follow-up were excluded. All patients with PRDs underwent skin sampling at 3 sites (the near-ear area, upper arm, lower back, and inner thigh), and a portion of them had CSF samples taken simultaneously. In the confirmatory cohort, a single skin biopsy site and CSF samples were simultaneously collected from a portion of patients with PRDs. Exposures The skin and CSF prion seeding activity was assessed using the real-time quaking-induced conversion (RT-QUIC) assay, with rHaPrP90-231, a Syrian hamster recombinant prion protein, as the substrate. In the exploratory cohort, skin samples were tested at dilutions of 10-2 through 10-4. In the confirmatory cohort, skin samples were tested at a dilution of 10-2. A total of four 15-μL wells of CSF were used in the RT-QUIC assay. Main Outcomes and Measures Correlations between RT-QUIC results from the skin and CSF and the final diagnosis of enrolled patients. Results In the exploratory cohort, the study included 101 patients (mean [SD] age, 60.9 [10.2] years; 63 female [62.4%]) with PRD and 23 patients (mean [SD] age, 63.4 [9.1] years; 13 female [56.5%]) without PRD. A total of 94 patients had CSF samples taken simultaneously with the skin biopsy samples. In the confirmatory cohort, a single skin biopsy site and CSF sample were taken simultaneously in 43 patients with PRDs. Using an experimental condition of 10-2 dilution, the RT-QUIC positive rates of skin samples from different sites were comparable with those of the CSF (skin: 18 of 26 [69.2%] to 74 of 93 [79.6%] vs CSF: 71 of 94 [75.5%]). When tested at 3 different dilutions, all skin sample positivity rates increased to over 80.0% (79 of 93 for the near-ear area, 21 of 26 for the upper arm, 77 of 92 for the lower back, and 78 of 92 for the inner thigh). Combining samples from skin sites near the ear, inner thigh, and lower back in pairs yielded positivity rates exceeding 92.1% (93 of 101), significantly higher than CSF alone (71 of 94 [75.5%]; P =.002). When all skin sample sites were combined and tested at 3 dilution concentrations for RT-QUIC, the sensitivity reached 95.0% (96 of 101). In the confirmatory cohort, the RT-QUIC positive rate of a single skin biopsy sample was slightly higher than that of the CSF (34 of 43 [79.1%] vs 31 of 43 [72.1%]; P = .45). Conclusions and Relevance Results of this diagnostic study suggest that the sensitivity of an RT-QUIC analysis of a combination of 2 or more skin sites was superior to that of CSF in diagnosing PRDs.
Collapse
Affiliation(s)
- Zhong-Yun Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Shi
- National Key-Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key-Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yu Kong
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dong-Lin Liang
- National Key-Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yi-Hao Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rong Min
- Department of Clinical Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jing Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong Ye
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Gao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Chu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hai-Tian Nan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - De-Ming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jun-Jie Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lin Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wen-Quan Zou
- Institute of Neurology, Jiangxi Academy of Clinical Medical Sciences, Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li-Yong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiao-Ping Dong
- National Key-Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
7
|
Quan M. Timing of biomarker changes preceding Alzheimer's disease: for diagnosis, treatment or prevention? SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2733-7. [PMID: 39422811 DOI: 10.1007/s11427-024-2733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing, 100053, China.
| |
Collapse
|
8
|
Ma SJ, Yu YX, Tian K, Yong W, Yu WL, Bai RY, Wu LE, Guo X. Prevalence and risk factors of subjective cognitive decline in older adults in Baotou, China: a cross-sectional study. Front Aging Neurosci 2024; 16:1422258. [PMID: 39444802 PMCID: PMC11496101 DOI: 10.3389/fnagi.2024.1422258] [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: 04/23/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Objectives Subjective cognitive decline (SCD) as a stage between healthy cognition and early neurocognitive disorders, has been proposed to be helpful in the diagnosis of prodromal neurocognitive disorders. To investigate the prevalence of SCD and the related risk factors on the prevalence. Methods A cross-sectional study involving 1,120 elderly subjects residing in Baotou, China. From June 2021 to June 2023, the data were gathered by research assistants with training utilizing standardized questionnaires. The following factors were evaluated: subjective cognitive decline, physical and cognitive activity levels, past medical history, demographics, instrumental activities of daily living, and cognitive function. Risk factors of SCD were used chi-square tests and multivariate logistic regression analysis. Results The prevalence of SCD was 43.8%. Permanent residence, marital status, BMI, dietary habits, average sleep duration per night, smoking, diabetes, coronary heart disease, and visual impairment were significantly associated with SCD (p < 0 0.05). Multivariable logistic regression analysis showed obesity, vegetarian-based, smoking for a long time, diabetes and coronary heart disease, visual impairment, no spouse, and average sleep duration per night <6 h were independent risk factors for SCD. Based on the gender analysis, the difference in marital status, dietary habits, average sleep duration per night, smoking, drinking, and hypertension was statistically significant (p < 0.001). Conclusion The prevalence of subjective cognitive decline was high among elder adults. We discovered significant differences in the prevalence or risk factors for SCD between men and women based on their sex. This study provides a more theoretical basis for the early prevention and screening of cognitive impairment diseases in the elderly population.
Collapse
Affiliation(s)
- Shang-Jia Ma
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Yan-Xue Yu
- Department of Neurological Function, Luoyang Central Hospital, Luoyang, China
| | - Kai Tian
- Department of Psychological Rehabilitation, The Third Hospital of Baogang Group, Baotou, China
| | - Wen Yong
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Wen-Long Yu
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Ru-Yu Bai
- Department of Neurology, The Ninth People’s Hospital of Shenyang, Shenyang, China
| | - Li-E Wu
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Xia Guo
- Department of Neurology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| |
Collapse
|
9
|
Shaib AH, Chouaib AA, Chowdhury R, Altendorf J, Mihaylov D, Zhang C, Krah D, Imani V, Spencer RKW, Georgiev SV, Mougios N, Monga M, Reshetniak S, Mimoso T, Chen H, Fatehbasharzad P, Crzan D, Saal KA, Alawieh MM, Alawar N, Eilts J, Kang J, Soleimani A, Müller M, Pape C, Alvarez L, Trenkwalder C, Mollenhauer B, Outeiro TF, Köster S, Preobraschenski J, Becherer U, Moser T, Boyden ES, Aricescu AR, Sauer M, Opazo F, Rizzoli SO. One-step nanoscale expansion microscopy reveals individual protein shapes. Nat Biotechnol 2024:10.1038/s41587-024-02431-9. [PMID: 39385007 DOI: 10.1038/s41587-024-02431-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
The attainable resolution of fluorescence microscopy has reached the subnanometer range, but this technique still fails to image the morphology of single proteins or small molecular complexes. Here, we expand the specimens at least tenfold, label them with conventional fluorophores and image them with conventional light microscopes, acquiring videos in which we analyze fluorescence fluctuations. One-step nanoscale expansion (ONE) microscopy enables the visualization of the shapes of individual membrane and soluble proteins, achieving around 1-nm resolution. We show that conformational changes are readily observable, such as those undergone by the ~17-kDa protein calmodulin upon Ca2+ binding. ONE is also applied to clinical samples, analyzing the morphology of protein aggregates in cerebrospinal fluid from persons with Parkinson disease, potentially aiding disease diagnosis. This technology bridges the gap between high-resolution structural biology techniques and light microscopy, providing new avenues for discoveries in biology and medicine.
Collapse
Affiliation(s)
- Ali H Shaib
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany.
| | - Abed Alrahman Chouaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Rajdeep Chowdhury
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Department of Chemistry, GITAM School of Science, GITAM, Hyderabad, India
| | - Jonas Altendorf
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Chi Zhang
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Donatus Krah
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Vanessa Imani
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Russell K W Spencer
- Institute for Theoretical Physics, Georg-August University, Göttingen, Germany
| | - Svilen Veselinov Georgiev
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Nikolaos Mougios
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Mehar Monga
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
| | - Sofiia Reshetniak
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Mimoso
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Han Chen
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Parisa Fatehbasharzad
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Dagmar Crzan
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Kim-Ann Saal
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Mohamad Mahdi Alawieh
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadia Alawar
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Janna Eilts
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Jinyoung Kang
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alireza Soleimani
- Institute for Theoretical Physics, Georg-August University, Göttingen, Germany
| | - Marcus Müller
- Institute for Theoretical Physics, Georg-August University, Göttingen, Germany
| | - Constantin Pape
- Institute of Computer Science, Georg-August University Göttingen, Göttingen, Germany
| | | | - Claudia Trenkwalder
- Department of Neurosurgery, University Medical Center, Göttingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Julia Preobraschenski
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Ute Becherer
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Tobias Moser
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Edward S Boyden
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Felipe Opazo
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- NanoTag Biotechnologies GmbH, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
10
|
Fabara-Rodríguez AC, García-Bravo C, García-Bravo S, Quirosa-Galán I, Rodríguez-Pérez MP, Pérez-Corrales J, Fernández-Gómez G, Donovan M, Huertas-Hoyas E. Quality-of-Life- and Cognitive-Oriented Rehabilitation Program through NeuronUP in Older People with Alzheimer's Disease: A Randomized Clinical Trial. J Clin Med 2024; 13:5982. [PMID: 39408042 PMCID: PMC11477658 DOI: 10.3390/jcm13195982] [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: 08/14/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
(1) Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by cognitive decline and functional impairment. The NeuronUP platform is a computer program whose main function is cognitive stimulation through three types of activities that change so that the user does not manage to learn it. This program provides opportunities to work on various domains, including activities of daily living (ADLs), social skills, and cognitive functions. The main objective of this randomized clinical trial was to assess the impact of integrating the NeuronUP platform with conventional occupational therapy to enhance or maintain cognitive, perceptual, and quality of life (QoL) abilities in people with AD compared to a control group. (2) Methods: A randomized, single-blind clinical trial was conducted. The sample was randomized using a software program, OxMar, which allowed the separation of the sample into a control group (CG) that received their conventional occupational therapy sessions and an experimental group (EG) that received therapy with NeuronUP, in addition to their conventional occupational therapy sessions. An eighteen-week intervention was conducted. (3) Results: The study included 20 participants, and significant differences were observed in most variables analyzed, indicating improvements after the intervention, particularly in measures of QoL and cognitive status. (4) Conclusions: Our findings demonstrate that an eighteen-week experimental protocol, incorporating the NeuronUP platform alongside conventional occupational therapy, led to improvements in cognitive status and QoL in older adults with AD. Thus, integrating the NeuronUP platform as a complementary tool to occupational therapy can be a valuable resource for enhancing the QoL of individuals with AD. However, due to the small sample size, further studies are needed to corroborate these findings.
Collapse
Affiliation(s)
| | - Cristina García-Bravo
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Research Group of Humanities and Qualitative Research in Health Science (Hum&QRinHS), 28922 Alcorcón, Spain;
- Physiocare Madrid, Physiotherapy Clinic, 28026 Madrid, Spain;
| | - Sara García-Bravo
- Physiocare Madrid, Physiotherapy Clinic, 28026 Madrid, Spain;
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (M.P.R.-P.); (E.H.-H.)
| | - Isabel Quirosa-Galán
- PhD Program in Health Sciences, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (I.Q.-G.); (G.F.-G.)
| | - Mª Pilar Rodríguez-Pérez
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (M.P.R.-P.); (E.H.-H.)
| | - Jorge Pérez-Corrales
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Research Group of Humanities and Qualitative Research in Health Science (Hum&QRinHS), 28922 Alcorcón, Spain;
| | - Gemma Fernández-Gómez
- PhD Program in Health Sciences, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (I.Q.-G.); (G.F.-G.)
- TANGRAM, Center for Comprehensive Care for Children and Adolescents, 28032 Madrid, Spain
| | | | - Elisabet Huertas-Hoyas
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (M.P.R.-P.); (E.H.-H.)
| |
Collapse
|
11
|
Li XL, Wang RT, Tan CC, Tan L, Xu W. Systolic blood pressure variability in late-life predicts cognitive trajectory and risk of Alzheimer's disease. Front Aging Neurosci 2024; 16:1448034. [PMID: 39420926 PMCID: PMC11483855 DOI: 10.3389/fnagi.2024.1448034] [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: 06/12/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Background The relationship of systolic blood pressure variability (SBPV) with Alzheimer's disease (AD) remains controversial. We aimed to explore the roles of SBPV in predicting AD incidence and to test the pathways that mediated the relationship of SBPV with cognitive functions. Methods Longitudinal data across 96 months (T0 to T4) were derived from the Alzheimer's disease Neuroimaging Initiative cohort. SBPV for each participant was calculated based on the four measurements of SBP across 24 months (T0 to T3). At T3, logistic regression models were used to test the SBPV difference between 86 new-onset AD and 743 controls. Linear regression models were used to test the associations of SBPV with cognition and AD imaging endophenotypes for 743 non-demented participants (median age = 77.0, female = 42%). Causal mediation analyses were conducted to explore the effects of imaging endophenotypes in mediating the relationships of SBPV with cognitive function. Finally, Cox proportional hazard model was utilized to explore the association of SBPV with incident risk of AD (T3 to T4, mean follow-up = 3.5 years). Results Participants with new-onset AD at T3 had significantly higher SBPV compared to their controls (p = 0.018). Higher SBPV was associated with lower scores of cognitive function (p = 0.005 for general cognition, p = 0.029 for memory, and p = 0.016 for executive function), higher cerebral burden of amyloid deposition by AV45 PET (p = 0.044), lower brain metabolism by FDG PET (p = 0.052), and higher burden of white matter hyperintensities (WMH) (p = 0.012). Amyloid pathology, brain metabolism, and WMH partially (ranging from 17.44% to 36.10%) mediated the associations of SBPV with cognition. Higher SBPV was significantly associated with elevated risk of developing AD (hazard ratio = 1.29, 95% confidence interval = 1.07 to 1.57, p = 0.008). Conclusion These findings supported that maintaining stable SBP in late life helped lower the risk of AD, partially by modulating amyloid pathology, cerebral metabolism, and cerebrovascular health.
Collapse
Affiliation(s)
- Xiao-Lu Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Medical College, Qingdao University, Qingdao, China
| | - Ruo-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Kugler BA, Lysaker CR, Franczak E, Hauger BM, Csikos V, Stopperan JA, Allen JA, Stanford JA, Koch LG, Britton SL, Thyfault JP, Wilkins HM. Intrinsic aerobic capacity modulates Alzheimer's disease pathological hallmarks, brain mitochondrial function and proteome during aging. GeroScience 2024; 46:4955-4967. [PMID: 38867031 PMCID: PMC11336007 DOI: 10.1007/s11357-024-01248-3] [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: 04/02/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
Low aerobic capacity is strongly associated with all-cause mortality and risk for Alzheimer's disease (AD). Individuals with early dementia and AD have lower aerobic capacity compared to age-matched controls. The mechanism by which aerobic capacity influences AD risk is unknown but is likely mediated by sexual dimorphism and tissue-level differences in mitochondrial energetics. Here, we used rats selectively bred for large differences in intrinsic aerobic exercise capacity. Brain tissue from 18-month and 24-month-old female and male low-capacity runner (LCR) and high-capacity runner (HCR) rats were analyzed for markers of mitochondrial function and AD-associated pathologies. LCR rats, irrespective of sex, exhibited a greater increase in brain amyloid beta (Aβ42) and tau hyperphosphorylation (pTauthr181/total tau) with aging. In female LCR rats, brain mitochondrial respiration at states 3, 4, and FCCP-induced uncoupling, when stimulated with pyruvate/malate, was reduced at 18 and 24 months, leading to lower ATP-linked mitochondrial respiration compared to mitochondria from HCR rats. Male LCR rats also showed reduced complex II-stimulated mitochondrial respiration (succinate + rotenone) at 24 months compared to HCR rats. Differences in mitochondrial respiration were associated with tau hyperphosphorylation and Aβ42 alterations in both HCR and LCR strains. Proteomic analysis unveiled a distinct difference in the mitochondrial proteome, wherein female LCR rats displayed diminished mitochondrial translation and oxidative phosphorylation (OXPHOS) proteins at 18 months compared to female HCR rats. Conversely, male LCR rats exhibited increased OXPHOS protein abundance but reduced tricarboxylic acid (TCA) cycle proteins compared to male HCR rats. These findings underscore a robust association between intrinsic aerobic exercise capacity, brain mitochondrial function, and AD pathologies during aging.
Collapse
Affiliation(s)
- Benjamin A Kugler
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Colton R Lysaker
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Edziu Franczak
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Brittany M Hauger
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Vivien Csikos
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Julia A Stopperan
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Julie A Allen
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - John A Stanford
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - John P Thyfault
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
- Research Service, Kansas City VA Medical Center Department of Veterans Affairs, University of Kansas Diabetes Center, Kansas City, KS, USA
- University of Kansas Medical Center Department of Molecular Biology and Biochemistry, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA.
- University of Kansas Medical Center Department of Molecular Biology and Biochemistry, Kansas City, KS, USA.
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
13
|
Lanni I, Chiacchierini G, Papagno C, Santangelo V, Campolongo P. Treating Alzheimer's disease with brain stimulation: From preclinical models to non-invasive stimulation in humans. Neurosci Biobehav Rev 2024; 165:105831. [PMID: 39074672 DOI: 10.1016/j.neubiorev.2024.105831] [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: 05/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) is a severe and progressive neurodegenerative condition that exerts detrimental effects on brain function. As of now, there is no effective treatment for AD patients. This review explores two distinct avenues of research. The first revolves around the use of animal studies and preclinical models to gain insights into AD's underlying mechanisms and potential treatment strategies. Specifically, it delves into the effectiveness of interventions such as Optogenetics and Chemogenetics, shedding light on their implications for understanding pathophysiological mechanisms and potential therapeutic applications. The second avenue focuses on non-invasive brain stimulation (NiBS) techniques in the context of AD. Evidence suggests that NiBS can successfully modulate cognitive functions associated with various neurological and neuropsychiatric disorders, including AD, as demonstrated by promising findings. Here, we critically assessed recent findings in AD research belonging to these lines of research and discuss their potential impact on the clinical horizon of AD treatment. These multifaceted approaches offer hope for advancing our comprehension of AD pathology and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Ilenia Lanni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Chiacchierini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Costanza Papagno
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Valerio Santangelo
- Functional Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
14
|
Bizzozero-Peroni B, Díaz-Goñi V, Beneit N, Oliveira A, Jiménez-López E, Martínez-Vizcaíno V, Mesas AE. Nut consumption is associated with a lower risk of all-cause dementia in adults: a community-based cohort study from the UK Biobank. GeroScience 2024:10.1007/s11357-024-01365-z. [PMID: 39343863 DOI: 10.1007/s11357-024-01365-z] [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: 07/04/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
This cohort study aimed to analyze the relationship between nut consumption and the risk of all-cause dementia in adults from the United Kingdom (UK). Data from participants in the UK Biobank cohort between 2007-2012 (baseline) and 2013-2023 (follow-up) were analyzed. Baseline information on nut consumption was obtained using the Oxford WebQ 24-h questionnaire. All-cause dementia (i.e. Alzheimer's disease, frontotemporal dementia, or vascular dementia) was assessed at baseline and follow-up through self-reported medical diagnosis, hospitalization, or death records. Hazard regression models were used to estimate the association between nut consumption and the risk of developing all-cause dementia, with adjustments made for sociodemographic, lifestyle, hearing problems, self-rated health, and the number of chronic diseases. Participants with all-cause dementia at baseline were excluded. A total of 50,386 participants (mean age 56.5 ± 7.7 years, 49.2% women) were included in the prospective analyses. The incidence of all-cause dementia was 2.8% (n = 1422 cases). Compared with no consumption, daily nut consumption (> 0 to 3 or more handfuls) was significantly associated with a 12% lower risk of all-cause dementia (hazard ratio = 0.88; 95% confidence interval, 0.77-0.99) after 7.1 mean years of follow-up, regardless of the potential confounders considered. No statistically significant interactions were observed between nut consumption and any of the covariates included in the hazard regression models. Stratified analyses revealed that nut consumption of up to 1 handful of 30 g/day and consumption of unsalted nuts were associated with the greatest protective benefits. The daily consumption of nuts may play a protective role in the prevention of dementia.
Collapse
Affiliation(s)
- Bruno Bizzozero-Peroni
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Instituto Superior de Educación Física, Universidad de la República, Rivera, Uruguay
| | - Valentina Díaz-Goñi
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Nuria Beneit
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain.
| | - Andreia Oliveira
- EPIUnit, Institute of Public Health, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health, University of Porto, Porto, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Estela Jiménez-López
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Arthur Eumann Mesas
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| |
Collapse
|
15
|
Shi J, Touchon J, Middleton LT, Rovira MB, Vassar R, Vellas B, Shen Y. Now and future: Strategies for diagnosis, prevention and therapies for Alzheimer's disease. Sci Bull (Beijing) 2024:S2095-9273(24)00715-1. [PMID: 39443183 DOI: 10.1016/j.scib.2024.09.042] [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: 08/29/2023] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 10/25/2024]
Abstract
After a number of failed drug studies on Alzheimer's disease (AD) over the past decade, clinical trials of AD started to show encouraging results and were approved or pending approval for clinical use. However, controversies on the clinically meaningful benefits and risks of brain edema and microhemorrhages have reminded us to think further about monitoring treatment and developing new drug targets. The goal of this review is to find insights from clinical trials that aimed at two key pathological features of AD, i.e., amyloid-β (Aβ) and tau protein, and to explore other targets such as anti-inflammation in AD. The complex pathophysiology of AD may require combination therapies rather than monotherapy. Throughout the course of AD, multiple pathways are disrupted, presenting a multitude of possible therapeutic targets for designing prevention and intervention for AD.
Collapse
Affiliation(s)
- Jiong Shi
- Department of Neurology, Institute on Aging and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| | - Jacques Touchon
- Institute of Neuroscience - University Hospital Gui de Chauliac-Montpellier, Montpellier 34295, France
| | - Lefkos T Middleton
- Ageing Epidemiology (AGE) Research, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - Mercé Boada Rovira
- Centro de Investigación Biomédica en Red sobre, Enfermedades Neurodegenerativas (CIBERNED), Universitat International de Catalunya-Barcelona, Barcelona 08028, Spain
| | - Robert Vassar
- Department of Cell Biology, Medical School, Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bruno Vellas
- IHU HealthAge, WHO Collaborating Center for Frailty, Clinical & Geoscience Research and Geriatric Training, Toulouse University Hospital, INSERM UMR 1295, University Paul Sabatier, Toulouse 31000, France.
| | - Yong Shen
- Department of Neurology, Institute on Aging and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
16
|
Huang B, Zheng W, Mu R, Yang P, Li X, Liu F, Qin X, Zhu X. Disparities in the diagnostic efficacy of radiomics models in predicting various degrees of cognitive impairment in patients with cerebral small vessel disease. BMC Med Imaging 2024; 24:257. [PMID: 39333913 PMCID: PMC11428886 DOI: 10.1186/s12880-024-01431-0] [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: 04/10/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Aim to validate the diagnostic efficacy of radiomics models for predicting various degrees of cognitive impairment in patients with cerebral small vessel disease (CSVD). METHODS Participants were divided into mild cognitive impairment group (mild-CSVD group) and sever cognitive impairment group (sever-CSVD group) according to Montreal Cognitive Assessment (MoCA) performance, 98 gender-age-education matched subjects served as normal controls. Radiomic features were extracted from the segmented hippocampus using PyRadiomics. The feature preprocessing involved replacing missing values with the mean, applying stratified random sampling to allocate subjects into training (80%) and testing (20%) sets, ensuring balance among the three classes (normal controls, mild-CSVD group, and sever-CSVD group). A feature selection method was applied to identify discriminative radiomic features, with the optimal texture feature chosen for developing diagnostic models. Performance was evaluated in both the training and testing sets using receiver operating characteristic (ROC) curve analysis. RESULTS The radiomics model achieved an accuracy of 0.625, an AUC of 0.593, a sensitivity of 0.828, and a specificity of 0.316 in distinguishing mild-CSVD group from normal controls. When distinguishing mild-CSVD group from sever-CSVD group, the radiomics model reached an accuracy of 0.683, an AUC of 0.660, a sensitivity of 0.167, and a specificity of 0.897. Similarly, in distinguishing sever-CSVD group from normal controls, the radiomics model exhibited an accuracy of 0.781, an AUC of 0.818, a sensitivity of 0.538, and a specificity of 0.947. CONCLUSION Radiomics model based on hippocampal texture had disparities in the diagnostic efficacy of radiomics models in predicting various degrees of cognitive impairment in patients with CSVD.
Collapse
Affiliation(s)
- Bingqin Huang
- Graduate School, Guilin Medical University, Guilin, 541002, China
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Wei Zheng
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Ronghua Mu
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Peng Yang
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Xin Li
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Fuzhen Liu
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Xiaoyan Qin
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China.
| | - Xiqi Zhu
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
| |
Collapse
|
17
|
Bieger A, Brum WS, Borelli WV, Therriault J, De Bastiani MA, Moreira AG, Benedet AL, Ferrari-Souza JP, Da Costa JC, Souza DO, Castilhos RM, Schumacher Schuh AF, Fagundes Chaves ML, Schöll M, Zetterberg H, Blennow K, Pascoal TA, Gauthier S, Rosa-Neto P, Schilling LP, Zimmer ER. Influence of Different Diagnostic Criteria on Alzheimer Disease Clinical Research. Neurology 2024; 103:e209753. [PMID: 39167736 PMCID: PMC11338500 DOI: 10.1212/wnl.0000000000209753] [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: 10/16/2023] [Accepted: 06/14/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Updates in Alzheimer disease (AD) diagnostic guidelines by the National Institute on Aging-Alzheimer's Association (NIA-AA) and the International Working Group (IWG) over the past 11 years may affect clinical diagnoses. We assessed how these guidelines affect clinical AD diagnosis in a cohort of cognitively unimpaired (CU) and cognitively impaired (CI) individuals. METHODS We applied clinical and biomarker data in algorithms to classify individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort according to the following diagnostic guidelines for AD: 2011 NIA-AA, 2016 IWG-2, 2018 NIA-AA, and 2021 IWG-3, assigning the following generic diagnostic labels: (1) not AD (nAD), (2) increased risk of developing AD (irAD), and (3) AD. Diagnostic labels were compared according to their frequency, convergence across guidelines, biomarker profiles, and prognostic value. We also evaluated the diagnostic discordance among the criteria. RESULTS A total of 1,195 individuals (mean age 73.2 ± 7.2 years, mean education 16.1 ± 2.7, 44.0% female) presented different repartitions of diagnostic labels according to the 2011 NIA-AA (nAD = 37.8%, irAD = 23.0%, AD = 39.2%), 2016 IWG-2 (nAD = 37.7%, irAD = 28.7%, AD = 33.6%), 2018 NIA-AA (nAD = 40.7%, irAD = 9.3%, AD = 50.0%), and 2021 IWG-3 (nAD = 51.2%, irAD = 8.4%, AD = 48.3%) frameworks. Discordant diagnoses across all guidelines were found in 512 participants (42.8%) (138 [91.4%] occurring in only β-amyloid [CU 65.4%, CI 34.6%] and 191 [78.6%] in only tau-positive [CU 71.7%, CI 28.3%] individuals). Differences in predicting cognitive impairment between nAD and irAD groups were observed with the 2011 NIA-AA (hazard ratio [HR] 2.21, 95% CI 1.34-3.65, p = 0.002), 2016 IWG-2 (HR 2.81, 95% CI 1.59-4.96, p < 0.000), and 2021 IWG-3 (HR 3.61, 95% CI 2.09-6.23, p < 0.000), but not with 2018 NIA-AA (HR 1.69, 95% CI 0.87-3.28, p = 0.115). DISCUSSION Over 42% of the studied population presented discordant diagnoses when using the different examined AD criteria, mostly in individuals with a single positive biomarker. Except for 2018 NIA-AA, all guidelines identified asymptomatic individuals at risk of cognitive impairment. Our findings highlight important differences between the guidelines, emphasizing the necessity for updated criteria with enhanced staging metrics, considering clinical, research, therapeutic, and trial design aspects.
Collapse
Affiliation(s)
- Andrei Bieger
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wagner S Brum
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wyllians V Borelli
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Joseph Therriault
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marco A De Bastiani
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Amanda G Moreira
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Andrea L Benedet
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Jaderson C Da Costa
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Diogo O Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Raphael M Castilhos
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Artur Francisco Schumacher Schuh
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marcia L Fagundes Chaves
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Michael Schöll
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Henrik Zetterberg
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Kaj Blennow
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Tharick A Pascoal
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Serge Gauthier
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Lucas P Schilling
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| |
Collapse
|
18
|
Mohammadzadeh M, Khoshakhlagh AH, Grafman J. Air pollution: a latent key driving force of dementia. BMC Public Health 2024; 24:2370. [PMID: 39223534 PMCID: PMC11367863 DOI: 10.1186/s12889-024-19918-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Many researchers have studied the role of air pollutants on cognitive function, changes in brain structure, and occurrence of dementia. Due to the wide range of studies and often contradictory results, the present systematic review was conducted to try and clarify the relationship between air pollutants and dementia. To identify studies for this review, a systematic search was conducted in Scopus, PubMed, and Web of Science databases (without historical restrictions) until May 22, 2023. The PECO statement was created to clarify the research question, and articles that did not meet the criteria of this statement were excluded. In this review, animal studies, laboratory studies, books, review articles, conference papers and letters to the editors were avoided. Also, studies focused on the effect of air pollutants on cellular and biochemical changes (without investigating dementia) were also excluded. A quality assessment was done according to the type of design of each article, using the checklist developed by the Joanna Briggs Institute (JBI). Finally, selected studies were reviewed and discussed in terms of Alzheimer's dementia and non-Alzheimer's dementia. We identified 14,924 articles through a systematic search in databases, and after comprehensive reviews, 53 articles were found to be eligible for inclusion in the current systematic review. The results showed that chronic exposure to higher levels of air pollutants was associated with adverse effects on cognitive abilities and the presence of dementia. Studies strongly supported the negative effects of PM2.5 and then NO2 on the brain and the development of neurodegenerative disorders in old age. Because the onset of brain structural changes due to dementia begins decades before the onset of disease symptoms, and that exposure to air pollution is considered a modifiable risk factor, taking preventive measures to reduce air pollution and introducing behavioral interventions to reduce people's exposure to pollutants is advisable.
Collapse
Affiliation(s)
- Mahdiyeh Mohammadzadeh
- Department of Health in Emergencies and Disasters, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Climate Change and Health Research Center (CCHRC), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Khoshakhlagh
- Department of Occupational Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran.
| | - Jordan Grafman
- Department of Physical Medicine & Rehabilitation, Neurology, Cognitive Neurology and Alzheimer's Center, Department of Psychiatry, Feinberg School of Medicine & Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| |
Collapse
|
19
|
Macchitella L, Tosi G, Giaquinto F, Iaia M, Rizzi E, Chiarello Y, Bertoux M, Angelelli P, Romano DL. Genuine Memory Deficits as Assessed by the Free and Cued Selective Reminding Test (FCSRT) in the Behavioural Variant of Frontotemporal Dementia. A Systematic Review and Meta-analysis Study. Neuropsychol Rev 2024; 34:823-837. [PMID: 37736861 PMCID: PMC11473568 DOI: 10.1007/s11065-023-09613-3] [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: 06/29/2022] [Accepted: 08/07/2023] [Indexed: 09/23/2023]
Abstract
The current diagnostic criteria for the behavioural variant of frontotemporal dementia (bvFTD) foresee a relative sparing of long-term memory. Although bvFTD patients were thought to report secondary memory deficits associated with prefrontal dysfunctions, some studies indicated the presence of a "genuine memory deficit" related to mesial temporal lobe dysfunctions. Among various neuropsychological tests, the Free and Cue Selective Reminding Test (FCSRT) has been recommended to distinguish genuine from apparent amnesia. We conducted a systematic review and a random effect Bayesian meta-analysis to evaluate the nature and severity of memory deficit in bvFTD. Our objective was to determine whether the existing literature offers evidence of genuine or apparent amnesia in patients with bvFTD, as assessed via the FCSRT. On 06/19/2021, we conducted a search across four databases (PMC, Scopus, Web of Science, and PubMed). We included all studies that evaluated memory performance using the FCSRT in patients with bvFTD, as long as they also included either cognitively unimpaired participants or AD groups. We tested publication bias through the Funnel plot and Egger's test. To assess the quality of studies, we used the Newcastle-Ottawa quality assessment scale adapted for cross-sectional studies. We included 16 studies in the meta-analysis. The results showed that bvFTD patients perform better than AD patients (pooled effects between 0.95 and 1.14), as their memory performance stands between AD and control groups (pooled effects between - 2.19 and - 1.25). Moreover, patients with bvFTD present both genuine and secondary memory disorders. As a major limitation of this study, due to our adoption of a rigorous methodology and stringent inclusion criteria, we ended up with just 16 studies. Nonetheless, our robust findings can contribute to the ongoing discussion on international consensus criteria for bvFTD and the selection of appropriate neuropsychological tools to facilitate the differential diagnosis between AD and bvFTD.
Collapse
Affiliation(s)
- Luigi Macchitella
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
- Scientific Institute I.R.C.C.S. "E. Medea", Unit for Severe Disabilities in Developmental Age and Young Adults (Developmental Neurology and Neurorehabilitation), Piazza Di Summa, 72100, Brindisi, Italy
| | - Giorgia Tosi
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy.
- Department of Psychology, University of Milano-Bicocca, Piazza Ateneo Nuovo 1, 20126, Milan, Italy.
| | - Francesco Giaquinto
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
| | - Marika Iaia
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
| | - Ezia Rizzi
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
| | - Ylenia Chiarello
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
| | - Maxime Bertoux
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, LiCEND, DISTALZ, 42 rue Paul Duez, 59000, Lille, France
| | - Paola Angelelli
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
| | - Daniele Luigi Romano
- Department of Human and Social Sciences, University of Salento, Piazza Tancredi 7, 73100, Lecce, Italy
- Department of Psychology, University of Milano-Bicocca, Piazza Ateneo Nuovo 1, 20126, Milan, Italy
| |
Collapse
|
20
|
Hadad R, Mandelli ML, Rankin KP, Toohey C, Sturm VE, Javandel S, Milicic A, Knudtson M, Allen IE, Hoffmann N, Friedberg A, Possin K, Valcour V, Miller BL. Itching Frequency and Neuroanatomic Correlates in Frontotemporal Lobar Degeneration. JAMA Neurol 2024; 81:977-984. [PMID: 39037825 PMCID: PMC11264090 DOI: 10.1001/jamaneurol.2024.2213] [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: 01/07/2024] [Accepted: 05/24/2024] [Indexed: 07/24/2024]
Abstract
Importance Itching is common in geriatric populations and is frequently linked to dermatological or systemic conditions. Itching engages specific brain regions that are implicated in the pathogenesis of frontotemporal lobar degeneration spectrum disorders (FTLD-SD). Thus, itching of undetermined origin (IUO) may indicate the presence of a neurodegenerative process. Objective To compare the frequency of itching in FTLD-SD and Alzheimer disease (AD) and to determine the neuroanatomical underpinnings of IUO. Design, Setting, and Participants This case-control study evaluated data and brain magnetic resonance images (MRIs) for participants with FTLD-SD or AD. Participants of a research study on FTLD-SD at the University of California, San Francisco, Memory and Aging Center were evaluated from May 1, 2002, to December 31, 2021. The exposure group underwent structural brain MRI within 6 months of initial diagnosis. Research visit summaries were reviewed to validate qualitative details and accurately identify itching with undetermined origin (IUO). Exposures Symptoms suggestive of FTLD-SD or AD. Main Outcomes and Measures Frequency of itching in FTLD-SD and AD and neuroanatomic correlates. Results A total of 2091 research visit summaries were reviewed for 1112 patients exhibiting symptoms indicative of FTLD-SD or AD. From 795 records where itching or a related phrase was endorsed, 137 had IUO. A total of 454 participants were included in the study: 137 in the itching group (mean [SD] age, 62.7 [9.9] years; 74 [54%] females and 63 males [46%]) and 317 in the nonitching group (mean [SD] age, 60.7 [10.8] years; 154 [49%] females and 163 males [51%]). Groups were similar in age, sex, and disease severity. More frequent itching was found in FTLD-SD (95/248 patients [38%], of which 44 [46%] had behavioral variant frontotemporal dementia [bvFTD]) compared with the AD group (14/77 patients [18%]; P = .001). The odds of itching were 2.4 (95% CI, 1.48-3.97) times higher for FTLD-SD compared with all other cases of dementia. Compared with healthy controls, the group with IUO exhibited greater gray matter atrophy bilaterally in the amygdala, insula, precentral gyrus, and cingulum, as well as in the right frontal superior gyrus and thalamus. Among patients with bvFTD and itching vs bvFTD without itching, itching was associated with right-lateralized gray matter atrophy affecting the insula, thalamus, superior frontal gyrus, and cingulum. Conclusions and Relevance Among individuals with IUO, FTLD-SD was disproportionately represented compared with AD. In FTLD-SD, dysfunction in the right anterior insula and its connected regions, including the right precentral gyrus, cingulum, and bilateral amygdala, contribute to dysregulation of the itching-scratching networks, resulting in uncontrollable itching or skin picking. Awareness among physicians about the relationship between neurodegeneration and itching may help in the management of itch in older individuals. Further studies are needed to determine the best treatments for these symptoms in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
- Rafi Hadad
- Stroke and Cognition Institute, Department of Neurology, Rambam Health Care Campus, Haifa, Israel
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
| | - Maria Luisa Mandelli
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Katherine P. Rankin
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Charlie Toohey
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Virginia E. Sturm
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
- Department of Psychiatry and Behavioral Sciences University of California, San Francisco
| | - Shireen Javandel
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Andjelika Milicic
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Marguerite Knudtson
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Isabel Elaine Allen
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Nathalia Hoffmann
- Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Adit Friedberg
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Katherine Possin
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Victor Valcour
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Bruce L. Miller
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| |
Collapse
|
21
|
Dayarathna T, Roseborough AD, Gomes J, Khazaee R, Silveira CRA, Borron K, Yu S, Coleman K, Jesso S, Finger E, MacDonald P, Borrie M, Wells J, Bartha R, Zou G, Whitehead SN, Leong HS, Pasternak SH. Nanoscale flow cytometry-based quantification of blood-based extracellular vesicle biomarkers distinguishes MCI and Alzheimer's disease. Alzheimers Dement 2024; 20:6094-6106. [PMID: 38958575 PMCID: PMC11497682 DOI: 10.1002/alz.14087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Accurate testing for Alzheimer's disease (AD) represents a crucial step for therapeutic advancement. Currently, tests are expensive and require invasive sampling or radiation exposure. METHODS We developed a nanoscale flow cytometry (nFC)-based assay of extracellular vesicles (EVs) to screen biomarkers in plasma from mild cognitive impairment (MCI), AD, or controls. RESULTS Circulating amyloid beta (Aβ), tau, phosphorylated tau (p-tau)181, p-tau231, p-tau217, p-tauS235, ubiquitin, and lysosomal-associated membrane protein 1-positive EVs distinguished AD samples. p-tau181, p-tau217, p-tauS235, and ubiquitin-positive EVs distinguished MCI samples. The most sensitive marker for AD distinction was p-tau231, with an area under the receiver operating characteristic curve (AUC) of 0.96 (sensitivity 0.95/specificity 1.0) improving to an AUC of 0.989 when combined with p-tauS235. DISCUSSION This nFC-based assay accurately distinguishes MCI and AD plasma without EV isolation, offering a rapid approach requiring minute sample volumes. Incorporating nFC-based measurements in larger populations and comparison to "gold standard" biomarkers is an exciting next step for developing AD diagnostic tools. HIGHLIGHTS Extracellular vesicles represent promising biomarkers of Alzheimer's disease (AD) that can be measured in the peripheral circulation. This study demonstrates the utility of nanoscale flow cytometry for the measurement of circulating extracellular vesicles (EVs) in AD blood samples. Multiple markers including amyloid beta, tau, phosphorylated tau (p-tau)181, p-tau231, p-tau217, and p-tauS235 accurately distinguished AD samples from healthy controls. Future studies should expand blood and cerebrospinal fluid-based EV biomarker development using nanoflow cytometry approaches.
Collapse
Affiliation(s)
- Thamara Dayarathna
- Institute for Genomic MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Austyn D. Roseborough
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Janice Gomes
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Reza Khazaee
- Department of BiologyWestern UniversityLondonOntarioCanada
- Biotron Integrated Microscopy FacilityWestern UniversityLondonOntarioCanada
| | - Carolina R. A. Silveira
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
| | - Kathy Borron
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
| | - Soojung Yu
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
| | - Kristy Coleman
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
| | - Sarah Jesso
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
| | - Elizabeth Finger
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Penny MacDonald
- Department of Clinical Neurological Sciences, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Michael Borrie
- Department of Geriatric Medicine, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Jennie Wells
- Department of Geriatric Medicine, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Robert Bartha
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Guangyong Zou
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Shawn N. Whitehead
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Hon S. Leong
- Sunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Stephen H. Pasternak
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
- Cognitive Neurology and Alzheimer's Disease Research CentreParkwood Institute, St. Joseph's Health Care CentreLondonOntarioCanada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| |
Collapse
|
22
|
Lee EH, Yoo H, Kim YJ, Cheon BK, Ryu S, Chang Y, Yun J, Jang H, Kim JP, Kim HJ, Koh SB, Jeong JH, Na DL, Seo SW, Kang SH. Different associations between body mass index and Alzheimer's markers depending on metabolic health. Alzheimers Res Ther 2024; 16:194. [PMID: 39210402 PMCID: PMC11363444 DOI: 10.1186/s13195-024-01563-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Increasing evidence supports the association between body mass index (BMI), Alzheimer's disease, and vascular markers. Recently, metabolically unhealthy conditions have been reported to affect the expression of these markers. We aimed to investigate the effects of BMI status on Alzheimer's and vascular markers in relation to metabolic health status. METHODS We recruited 1,736 Asians without dementia (71.6 ± 8.0 years). Participants were categorized into underweight, normal weight, or obese groups based on their BMI. Each group was further divided into metabolically healthy (MH) and unhealthy (MU) groups based on the International Diabetes Foundation definition of metabolic syndrome. The main outcome was Aβ positivity, defined as a Centiloid value of 20.0 or above and the presence of vascular markers, defined as severe white matter hyperintensities (WMH). Logistic regression analyses were performed for Aβ positivity and severe WMH with BMI status or interaction terms between BMI and metabolic health status as predictors. Mediation analyses were performed with hippocampal volume (HV) and baseline Mini-Mental State Examination (MMSE) scores as the outcomes, and linear mixed models were performed for longitudinal change in MMSE scores. RESULTS Being underweight increased the risk of Aβ positivity (odds ratio [OR] = 2.37, 95% confidence interval [CI] 1.13-4.98), whereas obesity decreased Aβ positivity risk (OR = 0.63, 95% CI 0.50-0.80). Especially, obesity decreased the risk of Aβ positivity (OR = 0.38, 95% CI 0.26-0.56) in the MH group, but not in the MU group. Obesity increased the risk of severe WMH (OR = 1.69, 1.16-2.47). Decreased Aβ positivity mediate the relationship between obesity and higher HV and MMSE scores, particularly in the MH group. Obesity demonstrated a slower decline in MMSE (β = 1.423, p = 0.037) compared to being normal weight, especially in the MH group. CONCLUSIONS Our findings provide new evidence that metabolic health has a significant effect on the relationship between obesity and Alzheimer's markers, which, in turn, lead to better clinical outcomes.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Heejin Yoo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Young Ju Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Bo Kyoung Cheon
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jihwan Yun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul National University college of Medicine, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Sung Hoon Kang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
23
|
Afrashteh F, Almasi-Dooghaee M, Kamyari N, Rajabi R, Baradaran HR. Is Montreal Cognitive Assessment a valuable test for the differentiation of Alzheimer's disease, frontotemporal dementia, dementia with Lewy body, and vascular dementia? Dement Neuropsychol 2024; 18:e20230124. [PMID: 39193467 PMCID: PMC11348881 DOI: 10.1590/1980-5764-dn-2023-0124] [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: 12/23/2023] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 08/29/2024] Open
Abstract
Dementia is one of the growing diseases in the world and has different types based on its definition. The Montreal Cognitive Assessment (MoCA) test has been employed to screen patients with dementia, cognitive impairment, and disruption of daily activities. Objective This study examined the diagnostic value of the total MoCA score and its subscores in differentiating Alzheimer's disease (AD), frontotemporal dementia (FTD), dementia with Lewy body (DLB), and vascular dementia (VaD). Methods A total of 241 patients (AD=110, FTD=90, DLB=28, and VaD=13) and 59 healthy persons, who were referred to a dementia clinic with memory impairment in Firoozgar Hospital, were included in this study. MoCA tests were performed in all patients and normal persons. Results By using the receiver operating characteristic (ROC) curve and measuring the area under the curve (AUC) for the total MoCA score in each group, AUC was 0.616, 0.681, 0.6117, and 0.583 for differentiating AD, FTD, DLB, and VaD patients, respectively. Among the groups, just the VaD group showed no significant usefulness in using the total MoCA score to differentiate it. To compare MoCA subscores, AD patients had higher scores in digit span, literal fluency, and abstraction but lower delayed recall scores compared with FTD patients. Conclusion The total MoCA score and its subscores could not differentiate people with different types of dementia in the setting of screening.
Collapse
Affiliation(s)
- Fatemeh Afrashteh
- Iran University of Medical Sciences, School of Medicine, Tehran, Iran
| | - Mostafa Almasi-Dooghaee
- Iran University of Medical Sciences, Department of Neurology, Firoozgar Clinical Research Development Center (FCRDC), Tehran, Iran
| | - Naser Kamyari
- Abadan University of Medical Sciences, Department of Biostatistics and Epidemiology, School of Health, Abadan, Iran
| | - Rayan Rajabi
- Iran University of Medical Sciences, School of Medicine, Tehran, Iran
| | - Hamid Reza Baradaran
- Iran University of Medical Sciences, Department of Epidemiology, School of Public Health, Tehran, Iran
- University of Aberdeen, Ageing Clinical and Experimental Research Team, Institute of Applied Health Sciences, Aberdeen, UK
| |
Collapse
|
24
|
Hok-A-Hin YS, Vermunt L, Peeters CF, van der Ende EL, de Boer SC, Meeter LH, van Swieten JC, Hu WT, Lleó A, Alcolea D, Engelborghs S, Sieben A, Chen-Plotkin A, Irwin DJ, van der Flier WM, Pijnenburg YA, Teunissen CE, del Campo M. Large-scale CSF proteome profiling identifies biomarkers for accurate diagnosis of Frontotemporal Dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312100. [PMID: 39228745 PMCID: PMC11370532 DOI: 10.1101/2024.08.19.24312100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Diagnosis of Frontotemporal dementia (FTD) and the specific underlying neuropathologies (frontotemporal lobar degeneration; FTLD- Tau and FTLD-TDP) is challenging, and thus fluid biomarkers are needed to improve diagnostic accuracy. We used proximity extension assays to analyze 665 proteins in cerebrospinal fluid (CSF) samples from a multicenter cohort including patients with FTD (n = 189), Alzheimer's Disease dementia (AD; n = 232), and cognitively unimpaired individuals (n = 196). In a subset, FTLD neuropathology was determined based on phenotype or genotype (FTLD-Tau = 87 and FTLD-TDP = 68). Forty three proteins were differentially regulated in FTD compared to controls and AD, reflecting axon development, regulation of synapse assembly, and cell-cell adhesion mediator activity pathways. Classification analysis identified a 14- and 13-CSF protein panel that discriminated FTD from controls (AUC: 0.96) or AD (AUC: 0.91). Custom multiplex panels confirmed the highly accurate discrimination between FTD and controls (AUCs > 0.96) or AD (AUCs > 0.88) in three validation cohorts, including one with autopsy confirmation (AUCs > 0.90). Six proteins were differentially regulated between FTLD-TDP and FTLD-Tau, but no reproducible classification model could be generated (AUC: 0.80). Overall, this study introduces novel FTD-specific biomarker panels with potential use in diagnostic setting.
Collapse
Affiliation(s)
- Yanaika S. Hok-A-Hin
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Carel F.W. Peeters
- Mathematical & Statistical Methods group – Biometris, Wageningen University & Research, Wageningen, The Netherlands
| | - Emma L. van der Ende
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Sterre C.M. de Boer
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Lieke H. Meeter
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John C. van Swieten
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William T. Hu
- Department of Neurology, Center for Neurodegenerative Diseases Research, Emory University School of Medicine, Atlanta, USA
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Neuroprotection and Neuromodulation Research Group (NEUR), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| | - Anne Sieben
- Lab of neuropathology, Neurobiobank, Institute Born-Bunge, Antwerp University, Edegem, Belgium
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wiesje M. van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Yolande A.L. Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San PabloCEU, CEU Universities, Madrid, Spain
| |
Collapse
|
25
|
Schäfer S, Tröger J, Kray J. Modern scores for traditional tests - Review of the diagnostic potential of scores derived from word list learning tests in mild cognitive impairment and early Alzheimer's Disease. Neuropsychologia 2024; 201:108908. [PMID: 38744410 DOI: 10.1016/j.neuropsychologia.2024.108908] [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: 09/21/2023] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Episodic memory impairment is one of the early hallmarks in Alzheimer's Disease. In the clinical diagnosis and research, episodic memory impairment is typically assessed using word lists that are repeatedly presented to and recalled by the participant across several trials. Until recently, total learning scores, which consist of the total number of words that are recalled by participants, were almost exclusively used for diagnostic purposes. The present review aims at summarizing evidence on additional scores derived from the learning trials which have recently been investigated more frequently regarding their diagnostic potential. These scores reflect item acquisition, error frequencies, strategy use, intertrial fluctuations, and recall consistency. Evidence was summarized regarding the effects of clinical status on these scores. Preclinical, mild cognitive impairment and mild Alzheimer's Disease stages were associated with a pattern of reduced item acquisition, more errors, less strategy use, and reduced access of items, indicating slowed and erroneous encoding. Practical implications and limitations of the present research will be discussed.
Collapse
Affiliation(s)
| | | | - Jutta Kray
- Saarland University, Saarbrücken, Germany
| |
Collapse
|
26
|
Vicario R, Fragkogianni S, Weber L, Lazarov T, Hu Y, Hayashi SY, Craddock BP, Socci ND, Alberdi A, Baako A, Ay O, Ogishi M, Lopez-Rodrigo E, Kappagantula R, Viale A, Iacobuzio-Donahue CA, Zhou T, Ransohoff RM, Chesworth R, Abdel-Wahab O, Boisson B, Elemento O, Casanova JL, Miller WT, Geissmann F. A microglia clonal inflammatory disorder in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577216. [PMID: 38328106 PMCID: PMC10849735 DOI: 10.1101/2024.01.25.577216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however few studies have investigated its role in neurodegenerative processes such as Alzheimer's Disease (AD). Here we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in human, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Leslie Weber
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Samantha Y. Hayashi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Barbara P. Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Rajya Kappagantula
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christine A. Iacobuzio-Donahue
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
27
|
Liang Y, Liu C, Cheng M, Geng L, Li J, Du W, Song M, Chen N, Yeleen TAN, Song L, Wang X, Han Y, Sheng C. The link between gut microbiome and Alzheimer's disease: From the perspective of new revised criteria for diagnosis and staging of Alzheimer's disease. Alzheimers Dement 2024; 20:5771-5788. [PMID: 38940631 PMCID: PMC11350031 DOI: 10.1002/alz.14057] [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: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Over the past decades, accumulating evidence suggests that the gut microbiome exerts a key role in Alzheimer's disease (AD). The Alzheimer's Association Workgroup is updating the diagnostic criteria for AD, which changed the profiles and categorization of biomarkers from "AT(N)" to "ATNIVS." Previously, most of studies focus on the correlation between the gut microbiome and amyloid beta deposition ("A"), the initial AD pathological feature triggering the "downstream" tauopathy and neurodegeneration. However, limited research investigated the interactions between the gut microbiome and other AD pathogenesis ("TNIVS"). In this review, we summarize current findings of the gut microbial characteristics in the whole spectrum of AD. Then, we describe the association of the gut microbiome with updated biomarker categories of AD pathogenesis. In addition, we outline the gut microbiome-related therapeutic strategies for AD. Finally, we discuss current key issues of the gut microbiome research in the AD field and future research directions. HIGHLIGHTS: The new revised criteria for Alzheimer's disease (AD) proposed by the Alzheimer's Association Workgroup have updated the profiles and categorization of biomarkers from "AT(N)" to "ATNIVS." The associations of the gut microbiome with updated biomarker categories of AD pathogenesis are described. Current findings of the gut microbial characteristics in the whole spectrum of AD are summarized. Therapeutic strategies for AD based on the gut microbiome are proposed.
Collapse
Affiliation(s)
- Yuan Liang
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Congcong Liu
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Manman Cheng
- Department of Respiratory MedicineThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Lijie Geng
- Department of RadiologyThe People's Hospital of YanzhouJiningChina
| | - Jing Li
- Department of EmergencyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Wenying Du
- Department of NeurologyChina‐Japan Friendship HospitalBeijingChina
| | - Minfang Song
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Nian Chen
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | | | - Li Song
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| | - Xiaoni Wang
- Department of NeurologySir Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Key Laboratory of Biomedical Engineering of Hainan ProvinceSchool of Biomedical EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
| | - Can Sheng
- Department of NeurologyThe Affiliated Hospital of Jining Medical UniversityJiningChina
| |
Collapse
|
28
|
Sainsily-Cesarus A, Schmitt E, Landre L, Botzung A, Rauch L, Demuynck C, Philippi N, de Sousa PL, Mutter C, Cretin B, Martin-Hunyadi C, Blanc F. Dementia with Lewy bodies and gait neural basis: a cross-sectional study. Alzheimers Res Ther 2024; 16:170. [PMID: 39080741 PMCID: PMC11287986 DOI: 10.1186/s13195-024-01539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Dementia with Lewy Bodies (DLB) is responsible for cognitive-behavioural disorders but also for gait disorders. The latter are thought to be related to parkinsonism, but the neural bases of these disorders are not well known, especially in the early stages. The aim of this study was to investigate by volumetric Magnetic Resonance Imaging the neuronal basis of gait disorders in DLB patients, compared to Healthy Elderly Controls and Alzheimer's Disease patients. METHODS Clinical examination with motor assessment including 10-meter walking speed, one-leg balance and Timed Up and Go test, a comprehensive neuropsychological evaluation and 3D brain Magnetic Resonance Imaging were performed on 84 DLB patients, 39 Alzheimer's Disease patients and 22 Healthy Elderly Controls. We used Statistical Parametric Mapping 12 to perform a one-sample t-test to investigate the correlation between each gait score and gray matter volume (P ≤ 0.05 corrected for family-wise error). RESULTS We found a correlation for DLB patients between walking speed and gray matter decrease (P < 0.05, corrected for family-wise error) in caudate nuclei, anterior cingulate cortex, mid-cingulate cortex, hippocampi, supplementary motor area, right cerebellar cortex and left parietal operculum. We found no correlation with Timed Up and Go test and one-leg balance. CONCLUSION Gait disorders are underpinned by certain classical regions such as the cerebellum and the supplementary motor area. Our results suggest there may be a motivational and emotional component of voluntary gait in DLB subjects, underpinned by the cingulate cortex, a spatial orientation component, underpinned by hippocampi and suggest the involvement of brain processing speed and parkinsonism, underpinned by the caudate nuclei. TRIAL REGISTRATION The study protocol has been registered on ClinicalTrials.gov. (NCT01876459) on June 12, 2013.
Collapse
Affiliation(s)
- Adele Sainsily-Cesarus
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Elise Schmitt
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France.
- Faculty of Medicine, University of Strasbourg, Strasbourg, EA-3072, France.
| | - Lionel Landre
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Anne Botzung
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
| | - Lucie Rauch
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
| | - Catherine Demuynck
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
| | - Nathalie Philippi
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Paulo Loureiro de Sousa
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Catherine Mutter
- University Hospital of Strasbourg, CIC INSERM 1434, Strasbourg, France
| | - Benjamin Cretin
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Catherine Martin-Hunyadi
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| | - Frederic Blanc
- Geriatrics Department, University Hospital of Strasbourg, CM2R (Memory Resource and Research Centre), Strasbourg, France
- University of Strasbourg, CNRS, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS, ICube laboratory, Strasbourg, France
| |
Collapse
|
29
|
Wertman E. Essential New Complexity-Based Themes for Patient-Centered Diagnosis and Treatment of Dementia and Predementia in Older People: Multimorbidity and Multilevel Phenomenology. J Clin Med 2024; 13:4202. [PMID: 39064242 PMCID: PMC11277671 DOI: 10.3390/jcm13144202] [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: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a highly prevalent condition with devastating clinical and socioeconomic sequela. It is expected to triple in prevalence by 2050. No treatment is currently known to be effective. Symptomatic late-onset dementia and predementia (SLODP) affects 95% of patients with the syndrome. In contrast to trials of pharmacological prevention, no treatment is suggested to remediate or cure these symptomatic patients. SLODP but not young onset dementia is intensely associated with multimorbidity (MUM), including brain-perturbating conditions (BPCs). Recent studies showed that MUM/BPCs have a major role in the pathogenesis of SLODP. Fortunately, most MUM/BPCs are medically treatable, and thus, their treatment may modify and improve SLODP, relieving suffering and reducing its clinical and socioeconomic threats. Regrettably, the complex system features of SLODP impede the diagnosis and treatment of the potentially remediable conditions (PRCs) associated with them, mainly due to failure of pattern recognition and a flawed diagnostic workup. We suggest incorporating two SLODP-specific conceptual themes into the diagnostic workup: MUM/BPC and multilevel phenomenological themes. By doing so, we were able to improve the diagnostic accuracy of SLODP components and optimize detecting and favorably treating PRCs. These revolutionary concepts and their implications for remediability and other parameters are discussed in the paper.
Collapse
Affiliation(s)
- Eli Wertman
- Department of Neurology, Hadassah University Hospital, The Hebrew University, Jerusalem 9190500, Israel;
- Section of Neuropsychology, Department of Psychology, The Hebrew University, Jerusalem 9190500, Israel
- Or’ad: Organization for Cognitive and Behavioral Changes in the Elderly, Jerusalem 9458118, Israel
- Merhav Neuropsychogeriatric Clinics, Nehalim 4995000, Israel
| |
Collapse
|
30
|
Lopez de Coca T, Moreno L, Pardo J, Pérez-Tur J, Ramos H, Villagrasa V. Influence of daily life and health profile in subtle cognitive decline of women residing in Spanish religious communities: DeCo religious orders study. Front Public Health 2024; 12:1395877. [PMID: 39086806 PMCID: PMC11288981 DOI: 10.3389/fpubh.2024.1395877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background Prior to the onset of dementia, individuals commonly undergo a phase marked by subtle cognitive changes, known as subtle cognitive decline. Recognizing these early cognitive alterations is crucial, as they can serve as indicators of an impending decline in cognitive function, warranting timely intervention and support. Objectives To determine the incidence of subtle cognitive decline in a population of Spanish women and establish the relationship with possible protective and/or risk factors such as cognitive reserve, cardiovascular risk factors, medication consumption and psychosocial factors. Design and participants This is a cross-sectional observational study with women from the general population and a more homogeneous population composed of nuns from the Valencian region (Spain). Measurements A validated questionnaire was used including lifestyle variables, chronic illnesses, level of education and pharmacological treatments. Three validated subtle cognitive decline screening tests with varying levels of sensitivity and specificity were used: Memory Impairment Screening, Pfeiffer's Short Portable Mental State Questionnaire, and Semantic Verbal Fluency. Results Our results suggest that nuns may have a significantly reduced risk of cognitive decline compared to the general population (20.67% in nuns vs. 36.63% in the general population). This lower risk for subtle cognitive decline in nuns may be partly attributed to their higher cognitive reserve and long-time engagement in intellectually stimulating activities. Additionally, nuns tend to adopt healthy lifestyles, they are not isolated because they live in community and obtained lower scores for risk factors such as depression, anticholinergic burden, and benzodiazepine consumption. Conclusion A healthy lifestyle combined with intellectually stimulating activities is related with preserved cognitive function.
Collapse
Affiliation(s)
- Teresa Lopez de Coca
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Lucrecia Moreno
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Juan Pardo
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Embedded Systems and Artificial Intelligence Group, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Jordi Pérez-Tur
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Institut de Biomedicina de València-CSIC, CIBERNED, ISCIII, Valencia, Spain
| | - Hernán Ramos
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Victoria Villagrasa
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| |
Collapse
|
31
|
Papamichail P, Sagredaki ML, Bouzineki C, Kanellopoulou S, Lyros E, Christakou A. The Effectiveness of an Exercise Program on Muscle Strength and Range of Motion on Upper Limbs, Functional Ability and Depression at Early Stage of Dementia. J Clin Med 2024; 13:4136. [PMID: 39064174 PMCID: PMC11278101 DOI: 10.3390/jcm13144136] [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: 06/15/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Dementia involves the loss of cognitive abilities and represents a decline from the prior level of function, which impairs functional abilities in day-to-day life. The purpose of the present study is to examine the effectiveness of an exercise program on the muscle strength and range of motion of the upper limbs, the functional status, and the depression of elderly people with early stage dementia. Methods: The sample consisted of 60 elderly people with early stage dementia who were randomly divided into a control and an experimental group of 30 participants each. The experimental group received a 12-week Otago exercise program with 45 min duration of each session. The control group received usual care without doing exercise. The outcome measures of muscle strength and range of motion of the upper limbs, the functional status, and the depression were assessed by valid instruments and tests at the beginning and at the end of the intervention program. Repeated measures one-way ANOVA and Mann-Whitney tests examined the differences between the two groups at the end of the 12-week exercise program. Results: Statistically significant differences were found between the experimental and control groups in the entire outcome measures (functional ability F = 9.35 p < 0.05; muscle strength right hand F = 32.33, p < 0.05 left hand U = 95.50 p < 0.01; e.g., range of motion shoulder extension U = 104.00 p < 0.01), except depression. Conclusions: Both muscle strength and range of motion of the upper limbs, as well as the functional ability, were improved by the exercise program. Further research is needed to investigate the present results, in particular to explore the long-term cognitive, behavioral, and functional status outcomes of exercise in the early stages of dementia.
Collapse
Affiliation(s)
- Panagiotis Papamichail
- Department of Physiotherapy, School of Health Sciences, University of Peloponnese, 23100 Sparta, Greece; (P.P.); (M.-L.S.); (E.L.)
- Intensive Care Unit, Bioclinic Athens, 11525 Athens, Greece
| | - Maria-Louiza Sagredaki
- Department of Physiotherapy, School of Health Sciences, University of Peloponnese, 23100 Sparta, Greece; (P.P.); (M.-L.S.); (E.L.)
| | | | | | - Epameinondas Lyros
- Department of Physiotherapy, School of Health Sciences, University of Peloponnese, 23100 Sparta, Greece; (P.P.); (M.-L.S.); (E.L.)
| | - Anna Christakou
- Department of Physiotherapy, Lab Biomechanics, School of Health Sciences, University of Peloponnese, 23100 Sparta, Greece
- Department of Physiotherapy, University of West Attica, 12243 Athens, Greece
| |
Collapse
|
32
|
Wang J, Feng Y, Sun Y. ACOT7, a candidate and novel serum biomarker of Alzheimer's disease. Front Aging Neurosci 2024; 16:1345668. [PMID: 39026992 PMCID: PMC11254632 DOI: 10.3389/fnagi.2024.1345668] [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: 11/28/2023] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Alzheimer's disease (AD) is the most common fatal neurodegenerative disease among the elderly worldwide, characterized by memory and cognitive impairment. The identification of biomarkers for AD is crucial and urgent to facilitate the diagnosis and intervention. The aim of this study was to evaluate the diagnostic value of acyl-Coenzyme A thioesterase 7 (ACOT7) as a serum biomarker for the prediction of AD. In our study, we observed a significant increase in ACOT7 expression in patients (n = 366) with AD and animal (n = 8-12) models of AD, compared to the control group. A significant negative correlation was found between ACOT7 levels and Mini-Mental State Examination (MMSE) scores (r = -0.85; p < 0.001). The analysis of the receiver operating characteristic curve (ROC) showed that the area under the curve (AUC) for ACOT7 was 0.83 (95% confidence intervals: 0.80-0.86). The optimal cut-off point of 62.5 pg./mL was selected with the highest sum of sensitivity and specificity. The diagnostic accuracy of serum ACOT7 for AD was 77% (95% confidence intervals: 72-82%), with a sensitivity of 80% (95% confidence intervals: 75-84%) and a specificity of 74% (95% confidence intervals: 69-79%). Moreover, the ROC analysis showed that the AUC of Aβ42/40 ratio is 0.70, and the diagnostic accuracy was 72%, with 69% sensitivity and 76% specificity. Compared with the AD traditional marker Aβ42/40 ratio, ACOT7 shows better superiority as a new serum candidate biomarker of AD. By suppressing the ACOT7 gene, our study provides evidence of the involvement of ACOT7 in the metabolism of amyloid precursor protein (APP), resulting in alterations in the expression levels of Aβ42, BACE1 and βCTF. ACOT7 has the ability to modulate the amyloidogenic pathway of APP metabolism, while it does not have an impact on the non-amyloidogenic pathway. In conclusion, the findings of our study suggest that serum ACOT7 may serve as a promising and non-invasive biomarker for AD.
Collapse
Affiliation(s)
- Jintao Wang
- Department of Pharmacy, First People’s Hospital of Wenling, Wenling, China
| | - Yong Feng
- Department of Medical Research, Qingdao Huangdao People’s Hospital, Qingdao, China
| | - Yingni Sun
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, China
- Beijing Handian Pharmaceutical Co, Ltd., Beijing, China
- School of Life Sciences, Ludong University, Yantai, China
| |
Collapse
|
33
|
Levak N, Lehtisalo J, Thunborg C, Westman E, Andersen P, Andrieu S, Broersen LM, Coley N, Hartmann T, Irving GF, Mangialasche F, Ngandu T, Pantel J, Rosenberg A, Sindi S, Soininen H, Solomon A, Wang R, Kivipelto M. Nutrition guidance within a multimodal intervention improves diet quality in prodromal Alzheimer's disease: Multimodal Preventive Trial for Alzheimer's Disease (MIND-AD mini). Alzheimers Res Ther 2024; 16:147. [PMID: 38961421 PMCID: PMC11221015 DOI: 10.1186/s13195-024-01522-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Multimodal lifestyle interventions can benefit overall health, including cognition, in populations at-risk for dementia. However, little is known about the effect of lifestyle interventions in patients with prodromal Alzheimer's disease (AD). Even less is known about dietary intake and adherence to dietary recommendations within this population making it difficult to design tailored interventions for them. METHOD A 6-month MIND-ADmini pilot randomized controlled trial (RCT) was conducted among 93 participants with prodromal AD in Sweden, Finland, Germany, and France. Three arms were included in the RCT: 1) multimodal lifestyle intervention (nutritional guidance, exercise, cognitive training, vascular/metabolic risk management, and social stimulation); 2) multimodal lifestyle intervention + medical food product; and 3) regular health advice (control group). Adherence to dietary advice was assessed with a brief food intake questionnaire by using the Healthy Diet Index (HDI) and Mediterranean Diet Adherence Screener (MEDAS). The intake of macro- and micronutrients were analyzed on a subsample using 3-day food records. RESULTS The dietary quality in the intervention groups, pooled together, improved compared to that of the control group at the end of the study, as measured with by HDI (p = 0.026) and MEDAS (p = 0.008). The lifestyle-only group improved significantly more in MEDAS (p = 0.046) and almost significantly in HDI (p = 0.052) compared to the control group, while the lifestyle + medical food group improved in both HDI (p = 0.042) and MEDAS (p = 0.007) during the study. There were no changes in macro- or micronutrient intake for the intervention groups at follow-up; however, the intakes in the control group declined in several vitamins and minerals when adjusted for energy intake. CONCLUSION These results suggest that dietary intervention as part of multimodal lifestyle interventions is feasible and results in improved dietary quality in a population with prodromal AD. Nutrient intakes remained unchanged in the intervention groups while the control group showed a decreasing nutrient density. TRIAL REGISTRATION ClinicalTrials.gov NCT03249688, 2017-07-08.
Collapse
Affiliation(s)
- Nicholas Levak
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Karolinska Vägen 37 A, 171 64, Solna, Sweden.
| | - Jenni Lehtisalo
- Population Health Unit, Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
| | - Charlotta Thunborg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Caring Sciences, Faculty of Health and Occupational Studies, University of Gävle, Gävle, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Pia Andersen
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Karolinska Vägen 37 A, 171 64, Solna, Sweden
| | - Sandrine Andrieu
- Department of Clinical Epidemiology and Public Health, UMR 1295, CHU de Toulouse, and Aging Research Team, CERPOP Inserm, Jules Guesde, 31000, Toulouse, France
- IHU HealthAge, Toulouse, 31059, France
| | | | - Nicola Coley
- Department of Clinical Epidemiology and Public Health, UMR 1295, CHU de Toulouse, and Aging Research Team, CERPOP Inserm, Jules Guesde, 31000, Toulouse, France
- IHU HealthAge, Toulouse, 31059, France
| | - Tobias Hartmann
- German Institute for Dementia Prevention (DIDP), Saarland University, 66424, Homburg, Germany
- Department of Experimental Neurology, Medical Faculty, Saarland University, 66424, Homburg, Germany
| | - Gerd Faxén Irving
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
| | - Francesca Mangialasche
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
| | - Tiia Ngandu
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Population Health Unit, Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Johannes Pantel
- Institute of General Practice, Goethe University Frankfurt a.M., Frankfurt, Germany
| | - Anna Rosenberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
| | - Alina Solomon
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Rui Wang
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Physical Activity and Health, the Swedish School of Sport and Health Sciences, 114 86, Stockholm, Sweden
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave J5/1 Mezzanine, Madison, WI, 53792, USA
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Center for Alzheimer Research QA32, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Karolinska Vägen 37 A, 171 64, Solna, Sweden
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 8, 70210, Kuopio, Finland
| |
Collapse
|
34
|
Cardoso S, Barros R, Marôco J, de Mendonça A, Guerreiro M. Different MMSE domains are associated to cognitive decline and education. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:533-539. [PMID: 35234096 DOI: 10.1080/23279095.2022.2041018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The Mini-Mental State Examination (MMSE) is a long-established test to screen for dementia, estimate the severity and monitor the progression of cognitive impairment. The MMSE total score is dependent upon demographic factors, particularly education, but little is known about how education influences the 6 distinct MMSE cognitive domains. The present study aims to understand how the performances in the MMSE cognitive domains reflect clinical diagnosis and educational level. The study recruited 1043 participants, comprising 388 healthy controls, 360 patients with Mild Cognitive Impairment (MCI) and 295 patients with dementia. The association of the MMSE cognitive domains scores with clinical diagnosis (healthy, MCI, dementia) and educational level (primary education, middle school, high school and university/college) was analyzed with a multivariate ordinal regression model. The scores in all MMSE domains were generally higher in healthy controls as compared to patients with MCI, and higher in these as compared to patients with dementia. The MMSE domain Constructional ability was associated to the education level, the domains Orientation, Recall and Language were associated to diagnosis, Attention and calculation was associated to both education level and diagnosis, and Registration was not associated to either education or diagnosis. In conclusion, impairment in specific MMSE domains pinpoints cognitive decline, probably indicating brain areas affected by neurodegeneration, and impairment in others reflects lower education levels and the lack of acquisition of relevant schooling abilities.
Collapse
Affiliation(s)
- Sandra Cardoso
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Ricardo Barros
- Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - João Marôco
- ISPA -Instituto Universitário, Lisbon, Portugal
| | | | | |
Collapse
|
35
|
Li Y, El Habib Daho M, Conze PH, Zeghlache R, Le Boité H, Tadayoni R, Cochener B, Lamard M, Quellec G. A review of deep learning-based information fusion techniques for multimodal medical image classification. Comput Biol Med 2024; 177:108635. [PMID: 38796881 DOI: 10.1016/j.compbiomed.2024.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/18/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Multimodal medical imaging plays a pivotal role in clinical diagnosis and research, as it combines information from various imaging modalities to provide a more comprehensive understanding of the underlying pathology. Recently, deep learning-based multimodal fusion techniques have emerged as powerful tools for improving medical image classification. This review offers a thorough analysis of the developments in deep learning-based multimodal fusion for medical classification tasks. We explore the complementary relationships among prevalent clinical modalities and outline three main fusion schemes for multimodal classification networks: input fusion, intermediate fusion (encompassing single-level fusion, hierarchical fusion, and attention-based fusion), and output fusion. By evaluating the performance of these fusion techniques, we provide insight into the suitability of different network architectures for various multimodal fusion scenarios and application domains. Furthermore, we delve into challenges related to network architecture selection, handling incomplete multimodal data management, and the potential limitations of multimodal fusion. Finally, we spotlight the promising future of Transformer-based multimodal fusion techniques and give recommendations for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Yihao Li
- LaTIM UMR 1101, Inserm, Brest, France; University of Western Brittany, Brest, France
| | - Mostafa El Habib Daho
- LaTIM UMR 1101, Inserm, Brest, France; University of Western Brittany, Brest, France.
| | | | - Rachid Zeghlache
- LaTIM UMR 1101, Inserm, Brest, France; University of Western Brittany, Brest, France
| | - Hugo Le Boité
- Sorbonne University, Paris, France; Ophthalmology Department, Lariboisière Hospital, AP-HP, Paris, France
| | - Ramin Tadayoni
- Ophthalmology Department, Lariboisière Hospital, AP-HP, Paris, France; Paris Cité University, Paris, France
| | - Béatrice Cochener
- LaTIM UMR 1101, Inserm, Brest, France; University of Western Brittany, Brest, France; Ophthalmology Department, CHRU Brest, Brest, France
| | - Mathieu Lamard
- LaTIM UMR 1101, Inserm, Brest, France; University of Western Brittany, Brest, France
| | | |
Collapse
|
36
|
Kintz S, Kim H, Wright HH. A preliminary investigation on core lexicon analysis in dementia of the Alzheimer's type. INTERNATIONAL JOURNAL OF LANGUAGE & COMMUNICATION DISORDERS 2024; 59:1336-1350. [PMID: 38165595 DOI: 10.1111/1460-6984.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/01/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Core lexicon (CL) analysis is a time efficient and possibly reliable measure that captures discourse production abilities. For people with aphasia, CL scores have demonstrated correlations with aphasia severity, as well as other discourse and linguistic measures. It was also found to be clinician-friendly and clinically sensitive enough to capture longitudinal changes in aphasia. To our knowledge, CL has never been investigated in individuals with neurologically progressive disease. AIMS As a preliminary investigation, we sought to investigate (1) whether CL scores correlate with dementia severity, (2) whether CL scores correlate with measures of discourse quality, and (3) whether CL scores correlate with other measures of lexical/semantic access. METHODS & PROCEDURES Twelve participants with a cognitive impairment associated with dementia of the Alzheimer's type (DAT) completed several measures of language and cognitive ability, as well as provide a language sample from the wordless picture book, Picnic. RESULTS & CONCLUSION Results are informative, as they provide insight into characteristics of CL and provide support for potential use of CL in individuals with neurologically progressive disease. The results indicated that CL scores do correlate with dementia severity and several measures of language ability, indicating they may provide a useful measure of language abilities in DAT, but more research is needed. WHAT THIS PAPER ADDS What is already known on the subject Core lexicon (CL) analysis is an assessment measure of discourse ability, most closely related to informativeness or productivity, used in aphasiology that is easier to use and less time consuming than previous measures of informativeness, such as correct information units or type-token ratio (TTR). For people with aphasia, CL analysis correlates with aphasia severity, measures of informativeness, as well as other measures of discourse quality. It has also been shown to be faster and more reliable between scorers than other informativeness measures. What this study adds Core lexicon analysis is a new simple and online method for assessing the informativeness of a discourse sample without the need to record or transcribe the language sample. CL is receiving a lot of attention in aphasia, correlating with everything from aphasia severity to measures of productivity and lexical access, as well as measures of informativeness. Unfortunately, no one has investigated CL analysis in dementia. The study demonstrates the first evidence that CL analysis may be a useful measure for determining dementia severity and language quality in people with dementia. What are the clinical implications of this work? Core lexicon analysis may provide clinicians and researchers with an easy method for assessing the discourse of people with a cognitive impairment associated with dementia of the Alzheimer's type. This will improve initial assessment, as well as improve ongoing language assessment that may provide clues into their functional ability to communicate effectively.
Collapse
Affiliation(s)
- Stephen Kintz
- Department of Speech Language Pathology, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Hana Kim
- Department of Communication Sciences & Disorders, University of South Florida, Tampa Bay, Florida, USA
| | - Heather Harris Wright
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
37
|
Bonato G, Cimino P, Pistonesi F, Salviati L, Bertolin C, Carecchio M. Non-Motor Symptoms in Primary Familial Brain Calcification. J Clin Med 2024; 13:3873. [PMID: 38999439 PMCID: PMC11242504 DOI: 10.3390/jcm13133873] [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: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: Primary Familial Brain Calcification is a rare neurodegenerative disorder of adulthood characterized by calcium deposition in the basal ganglia and other brain areas; the main clinical manifestations include movement disorders, mainly parkinsonism. Non-motor symptoms are not well defined in PFBC. This work aims at defining the burden of non-motor symptoms in PFBC. Methods: A clinical, genetic and neuropsychological evaluation of a cohort of PFBC patients, COMPASS-31 scale administration. Results: A total of 50 PFBC patients were recruited; in 25, the genetic test was negative; 10 carried mutations in SLC20A2 gene, 8 in MYORG, 3 in PDGFB, 1 in PDGFRB, 2 in JAM2 (single mutations), and one test is still ongoing. The main motor manifestation was parkinsonism. Headache was reported in 26% of subjects (especially in PDGFB mutation carriers), anxiety or depression in 62%, psychosis or hallucinations in 10-12%, sleep disturbances in 34%; 14% of patients reported hyposmia, 32% constipation, and 34% urinary disturbances. A neuropsychological assessment revealed cognitive involvement in 56% (sparing memory functions, to some extent). The COMPASS-31 mean score was 20.6, with higher sub-scores in orthostatic intolerance and gastrointestinal problems. MYORG patients and subjects with cognitive decline tended to have higher scores and bladder involvement compared to other groups. Conclusions: The presence of non-motor symptoms is frequent in PFBC and should be systematically assessed to better meet patients' needs.
Collapse
Affiliation(s)
- Giulia Bonato
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| | - Paola Cimino
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Francesca Pistonesi
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Leonardo Salviati
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Cinzia Bertolin
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| |
Collapse
|
38
|
Manenti R, Baglio F, Pagnoni I, Gobbi E, Campana E, Alaimo C, Rossetto F, Di Tella S, Pagliari C, Geviti A, Bonfiglio NS, Calabrò RS, Cimino V, Binetti G, Quartarone A, Bramanti P, Cappa SF, Rossini PM, Cotelli M. Long-lasting improvements in episodic memory among subjects with mild cognitive impairment who received transcranial direct current stimulation combined with cognitive treatment and telerehabilitation: a multicentre, randomized, active-controlled study. Front Aging Neurosci 2024; 16:1414593. [PMID: 38966802 PMCID: PMC11223647 DOI: 10.3389/fnagi.2024.1414593] [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: 04/09/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024] Open
Abstract
Background In recent years, an increasing number of studies have examined the potential efficacy of cognitive training procedures in individuals with normal ageing and mild cognitive impairment (MCI). Objective The aims of this study were to (i) evaluate the efficacy of the cognitive Virtual Reality Rehabilitation System (VRRS) combined with anodal transcranial direct current stimulation (tDCS) applied to the left dorsolateral prefrontal cortex compared to placebo tDCS stimulation combined with VRRS and (ii) to determine how to prolong the beneficial effects of the treatment. A total of 109 subjects with MCI were assigned to 1 of 5 study groups in a randomized controlled trial design: (a) face-to-face (FTF) VRRS during anodal tDCS followed by cognitive telerehabilitation (TR) (clinic-atDCS-VRRS+Tele@H-VRRS); (b) FTF VRRS during placebo tDCS followed by TR (clinic-ptDCS-VRRS+Tele@H-VRRS); (c) FTF VRRS followed by cognitive TR (clinic-VRRS+Tele@H-VRRS); (d) FTF VRRS followed by at-home unstructured cognitive stimulation (clinic-VRRS+@H-UCS); and (e) FTF cognitive treatment as usual (clinic-TAU). Results An improvement in episodic memory was observed after the end of clinic-atDCS-VRRS (p < 0.001). We found no enhancement in episodic memory after clinic-ptDCS-VRRS or after clinic-TAU.Moreover, the combined treatment led to prolonged beneficial effects (clinic-atDCS-VRRS+Tele@H-VRRS vs. clinic-ptDCS-VRRS+Tele@H-VRRS: p = 0.047; clinic-atDCS-VRRS+Tele@H-VRRS vs. clinic-VRRS+Tele@H-VRRS: p = 0.06). Discussion The present study provides preliminary evidence supporting the use of individualized VRRS combined with anodal tDCS and cognitive telerehabilitation for cognitive rehabilitation. Clinical trial registration https://clinicaltrials.gov/study/NCT03486704?term=NCT03486704&rank=1, NCT03486704.
Collapse
Affiliation(s)
- Rosa Manenti
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Ilaria Pagnoni
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Elena Gobbi
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Elena Campana
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Cristina Alaimo
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Sonia Di Tella
- IRCCS Fondazione Don Carlo Gnocchi – ONLUS, Milan, Italy
- Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | | | - Andrea Geviti
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | | | | | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Placido Bramanti
- IRCCS Centro Neurolesi “Bonino Pulejo”, Messina, Italy
- Università Degli Studi eCAMPUS, Novedrate, Italy
| | - Stefano F. Cappa
- Istituto Universitario Studi Superiori IUSS, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Paolo Maria Rossini
- Department Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Rome, Italy
| | - Maria Cotelli
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
39
|
Zhu W, Zhou X, Ren M, Yin W, Tang Y, Yin J, Sun Y, Zhu X, Sun Z. Process approach as a cognitive biomarker related to gray matter volume in mild cognitive impairment and Alzheimer's disease. BMC Neurol 2024; 24:199. [PMID: 38872077 PMCID: PMC11170873 DOI: 10.1186/s12883-024-03711-2] [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: 01/14/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Process approach is valuable for memory assessment in Alzheimer's disease (AD) and mild cognitive impairment (MCI), yet its underlying mechanisms remain elusive. This study aims to synergize the process approach with brain structure analysis to explore both the discriminative capacity and potential mechanisms underlying the process approach. METHODS 37 subjects of MCI, 35 subjects of AD and 38 subjects of healthy control (HC) were included. The process approach in Auditory Verbal Learning Test (AVLT), including discriminability (A'), response bias (B"D), semantic clustering (LBCsem) and serial clustering (LBCser) was performed. The gray matter volume (GMV) was analyzed by voxel-based morphometry. Receiver operating characteristic (ROC) analysis and partial correlations were conducted to explore the value of the process approach and investigate the relationship between the process approach, traditional indices of AVLT and GMV. RESULTS ROC analysis showed the value of A', B"D and LBCser in differentiating MCI and AD. Combining AVLT-Immediately Recall (AVLT-IR) and LBCser showed a higher value in diagnosing MCI. Partial correlations revealed that in the MCI group, A' and B"D were mainly positively associated with GMV of the hippocampus and temporal lobe. CONCLUSION This study indicated that the process approach is a promising cognitive biomarker to detect MCI and AD.
Collapse
Affiliation(s)
- Wenhao Zhu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Xia Zhou
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Mengmeng Ren
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Wenwen Yin
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Yating Tang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Jiabin Yin
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Yue Sun
- Department of Neurology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoqun Zhu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China
| | - Zhongwu Sun
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, China.
| |
Collapse
|
40
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
41
|
Chidambaram SB, Anand N, Varma SR, Ramamurthy S, Vichitra C, Sharma A, Mahalakshmi AM, Essa MM. Superoxide dismutase and neurological disorders. IBRO Neurosci Rep 2024; 16:373-394. [PMID: 39007083 PMCID: PMC11240301 DOI: 10.1016/j.ibneur.2023.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/21/2023] [Indexed: 07/16/2024] Open
Abstract
Superoxide dismutase (SOD) is a common antioxidant enzyme found majorly in living cells. The main physiological role of SOD is detoxification and maintain the redox balance, acts as a first line of defence against Reactive nitrogen species (RNS), Reactive oxygen species (ROS), and other such potentially hazardous molecules. SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide dismutases (SODs) are expressed in neurons and glial cells throughout the CNS both intracellularly and extracellularly. Endogenous oxidative stress (OS) linked with enlarged production of reactive oxygen metabolites (ROMs), inflammation, deregulation of redox balance, mitochondrial dysfunction and bioenergetic crisis are found to be prerequisite for neuronal loss in neurological diseases. Clinical and genetic studies indicate a direct correlation between mutations in SOD gene and neurodegenerative diseases, like Amyotrophic Lateral Sclerosis (ALS), Huntington's disease (HD), Parkinson's Disease (PD) and Alzheimer's Disease (AD). Therefore, inhibitors of OS are considered as an optimistic approach to prevent neuronal loss. SOD mimetics like Metalloporphyrin Mn (II)-cyclic polyamines, Nitroxides and Mn (III)- Salen complexes are designed and used as therapeutic extensively in the treatment of neurological disorders. SODs and SOD mimetics are promising future therapeutics in the field of various diseases with OS-mediated pathology.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department of Pharmacology, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Antigua, Antigua and Barbuda
| | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Ajman University, 346 Ajman, the United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, 346 Ajman, the United Arab Emirates
| | - Srinivasan Ramamurthy
- College of Pharmacy & Health Sciences, University of Science and Technology of Fujairah, 2202 Fujairah, the United Arab Emirates
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ambika Sharma
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
42
|
Xiang C, Zhang Y. Comparison of Cognitive Intervention Strategies for Individuals With Alzheimer's Disease: A Systematic Review and Network Meta-analysis. Neuropsychol Rev 2024; 34:402-416. [PMID: 36929474 PMCID: PMC11166762 DOI: 10.1007/s11065-023-09584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 12/22/2022] [Indexed: 03/18/2023]
Abstract
Accumulating evidence has shown the effectiveness of cognitive interventions, which can be divided into cognitive training (CT), cognitive stimulation (CS), cognitive rehabilitation (CR), and combined interventions (i.e., cognitive interventions combined with other non-pharmacological interventions such as physical exercise), in individuals with Alzheimer's disease (AD). However, the effectiveness of cognitive interventions varies greatly among studies and more comprehensive studies are required. We aimed to evaluate whether the current evidence shows that cognitive interventions are effective at improving cognition, neuropsychiatric symptoms, depression, quality of life, and basic activities of daily living among individuals with possible or probable AD. Randomized controlled trials of all types of cognitive intervention were identified for inclusion in pairwise and network meta-analyses. There was a moderate and statistically significant post-intervention improvement in global cognition among individuals with AD for all types of cognitive intervention compared to control interventions (39 studies, g = 0.43, 95% CI: 0.28 to 0.58, p < 0.01; Q = 102.27, df = 38, p < 0.01; I2 = 61.97%, τ2 = 0.13). Regarding the specific types of cognitive intervention, combined interventions had the highest surface under the cumulative ranking curve (SUCRA) value (90.7%), followed by CT (67.8%), CS (53.4%), and lastly CR (28.9%). Significant effects of cognitive interventions were also found for working memory, verbal memory, verbal fluency, confrontation naming, attention, neuropsychiatric symptoms, basic activities of daily living, and quality of life.
Collapse
Affiliation(s)
- Chunchen Xiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yumei Zhang
- Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Center of Stroke, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing Institute for Brain Disorders, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
43
|
van Dijk SE, Drenth N, Hafkemeijer A, Labadie G, Witjes-Ané MNW, Blauw GJ, Rombouts SARB, van der Grond J, van Rooden S. Neurovascular coupling in early stage dementia - A case-control study. J Cereb Blood Flow Metab 2024; 44:1013-1023. [PMID: 37994030 PMCID: PMC11318393 DOI: 10.1177/0271678x231214102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/11/2023] [Accepted: 10/05/2023] [Indexed: 11/24/2023]
Abstract
Cerebral amyloid angiopathy (CAA) is frequently found post mortem in Alzheimer's dementia, but often undetected during life especially since in vivo hallmarks of CAA and its vascular damage become overt relatively late in the disease process. Decreased neurovascular coupling to visual stimulation has been put forward as an early MRI marker for CAA disease severity. The current study investigates the role of neurovascular coupling in AD related dementia and its early stages. We included 25 subjective cognitive impairment, 33 mild cognitive impairment and 17 dementia patients and 44 controls. All participants underwent magnetic resonance imaging of the brain and neuropsychological assessment. Univariate general linear modeling analyses were used to assess neurovascular coupling between patient groups and controls. Moreover, linear regression analyses was used to assess the associations between neurovascular coupling and cognition. Our data show that BOLD amplitude is lower in dementia (mean 0.8 ± 0.2, p = 0.001) and MCI patients (mean 0.9 ± 0.3, p = 0.004) compared with controls (mean 1.1 ± 0.2). A low BOLD amplitude was associated with low scores in multiple cognitive domains. We conclude that cerebrovascular dysfunction, most likely due CAA, is an important comorbidity in early stages of dementia and has an independent effect on cognition.
Collapse
Affiliation(s)
- Suzanne E van Dijk
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadieh Drenth
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Hafkemeijer
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Psychology, Leiden University, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Gerda Labadie
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Noëlle W Witjes-Ané
- Department of Geriatrics and Psychiatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Gerard J Blauw
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Geriatrics, Haaglanden Medical Center, The Hague, the Netherlands
| | - Serge ARB Rombouts
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Psychology, Leiden University, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sanneke van Rooden
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
44
|
Bodryzlova Y, Moullec G, Kelly MP. The Dynamic Model of Health Assets: a model development. Glob Health Promot 2024:17579759241248624. [PMID: 38822628 DOI: 10.1177/17579759241248624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
AIM Epidemiological research on resistance and resilience can build on models of health developed in health promotion. Nevertheless, these models need to be adjusted to approaches currently employed in epidemiology; namely, included concepts should be easy to operationalize, and links between them should be simple enough to enable statistical modeling. In addition, these models should include both individual and environmental assets. The objective of this study is to consolidate the current knowledge on health assets, adjust them to epidemiological research needs, and propose a new model of health assets for epidemiological studies on health. DESIGN The conceptual paper was conducted according to the guidelines for the model development. METHODS The development of the new model was made from the perspective of salutogenesis - the branch of health promotion studying the origins of health. The analysis of literature on health promotion, public health, and positive psychology was conducted to find the links connecting individual and environmental assets. RESULTS The newly developed Dynamic Model of Health Assets circularly links individual characteristics, actions, environments, and support. Each preceding component of the model contributes to the following one; each component also independently contributes to resistance and resilience. The new model may guide large-scale epidemiological research on resistance and resilience. The model's components are easy to operationalize; the model allows for constructing multilevel models and accounting for the dynamic nature of the relationships between components. It is also generic enough to be adjusted to studying contributors to resistance and resilience to different specific diseases. CONCLUSION The new model can guide epidemiological studies on resistance and resilience.
Collapse
Affiliation(s)
- Yuliya Bodryzlova
- École de santé publique, Université de Montréal, Canada
- Centre de recherche du CIUSSS du Nord-de-l' Île-de-Montréal, Montréal, Canada
| | - Gregory Moullec
- École de santé publique, Université de Montréal, Canada
- Centre de recherche du CIUSSS du Nord-de-l' Île-de-Montréal, Montréal, Canada
| | - Michael P Kelly
- Department of Public Health and Primary Care, University of Cambridge, UK
| |
Collapse
|
45
|
Vecchio F, Miraglia F, Pappalettera C, Nucci L, Cacciotti A, Rossini PM. Small World derived index to distinguish Alzheimer's type dementia and healthy subjects. Age Ageing 2024; 53:afae121. [PMID: 38935531 PMCID: PMC11210397 DOI: 10.1093/ageing/afae121] [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: 12/27/2023] [Revised: 04/26/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND This article introduces a novel index aimed at uncovering specific brain connectivity patterns associated with Alzheimer's disease (AD), defined according to neuropsychological patterns. METHODS Electroencephalographic (EEG) recordings of 370 people, including 170 healthy subjects and 200 mild-AD patients, were acquired in different clinical centres using different acquisition equipment by harmonising acquisition settings. The study employed a new derived Small World (SW) index, SWcomb, that serves as a comprehensive metric designed to integrate the seven SW parameters, computed across the typical EEG frequency bands. The objective is to create a unified index that effectively distinguishes individuals with a neuropsychological pattern compatible with AD from healthy ones. RESULTS Results showed that the healthy group exhibited the lowest SWcomb values, while the AD group displayed the highest SWcomb ones. CONCLUSIONS These findings suggest that SWcomb index represents an easy-to-perform, low-cost, widely available and non-invasive biomarker for distinguishing between healthy individuals and AD patients.
Collapse
Affiliation(s)
- Fabrizio Vecchio
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Francesca Miraglia
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Chiara Pappalettera
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Lorenzo Nucci
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Alessia Cacciotti
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
46
|
Hong S, Choi Y, Lee MB, Rhee HY, Park S, Ryu CW, Cho AR, Kwon OI, Jahng GH. Increased extra-neurite conductivity of brain in patients with Alzheimer's disease: A pilot study. Psychiatry Res Neuroimaging 2024; 340:111807. [PMID: 38520873 DOI: 10.1016/j.pscychresns.2024.111807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/31/2024] [Accepted: 03/03/2024] [Indexed: 03/25/2024]
Abstract
The objectives of this study were to investigate how the extra-neurite conductivity (EC) and intra-neurite conductivity (IC) were reflected in Alzheimer's disease (AD) patients compared with old cognitively normal (CN) people and patients with amnestic mild cognitive impairment (MCI) and to evaluate the association between those conductivity values and cognitive decline. To do this, high-frequency conductivity (HFC) at the Larmor frequency was obtained using MRI-based electrical property tomography (MREPT) and was decomposed into EC and IC using information of multi-shell multi-gradient direction diffusion tensor images. This prospective single-center study included 20 patients with mild or moderate AD, 25 patients with amnestic MCI, and 21 old CN participants. After decomposing EC and IC from HFC for all participants, we performed voxel-based and regions-of-interest analyses to compare conductivity between the three participant groups and to evaluate the association with either age or the Mini-Mental State Examination (MMSE) scores. We found increased EC in AD compared to CN and MCI. EC was significantly negatively associated with MMSE scores in the insula, and middle temporal gyrus. EC might be used as an imaging biomarker for helping to monitor cognitive function.
Collapse
Affiliation(s)
- Seowon Hong
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea
| | - Yunjeong Choi
- Department of Biomedical Engineering, Undergraduate School, College of Electronics and Information, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Mun Bae Lee
- Department of Mathematics, College of Basic Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hak Young Rhee
- Department of Neurology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea; Department of Medicine, Kyung Hee University College of Medicine, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Soonchan Park
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea; Department of Medicine, Kyung Hee University College of Medicine, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chang-Woo Ryu
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea; Department of Medicine, Kyung Hee University College of Medicine, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ah Rang Cho
- Department of Medicine, Kyung Hee University College of Medicine, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Psychiatry, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea
| | - Oh In Kwon
- Department of Mathematics, College of Basic Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Republic of Korea; Department of Medicine, Kyung Hee University College of Medicine, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
47
|
Xu J, Sun Y, Zhu X, Pan S, Tong Z, Jiang K. Tactile discrimination as a diagnostic indicator of cognitive decline in patients with mild cognitive impairment: A narrative review. Heliyon 2024; 10:e31256. [PMID: 38803967 PMCID: PMC11129005 DOI: 10.1016/j.heliyon.2024.e31256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Background Tactile discrimination, a cognitive task reliant on fingertip touch for stimulus discrimination, encompasses the somatosensory system and working memory, with its acuity diminishing with advancing age. Presently, the evaluation of cognitive capacity to differentiate between individuals with early Alzheimer's disease (AD) and typical older adults predominantly relies on visual or auditory tasks, yet the efficacy of discrimination remains constrained. Aims To review the existing tactile cognitive tasks and explore the interaction between tactile perception and the pathological process of Alzheimer's disease. The tactile discrimination task may be used as a reference index of cognitive decline in patients with mild cognitive impairment and provide a new method for clinical evaluation. Methods We searched four databases (Embase, PubMed, Web of Science and Google scholar). The reference coverage was from 1936 to 2023. The search terms included "Alzheimer disease" "mild cognitive impairment" "tactile" "tactile discrimination" "tactile test" and so on. Reviews and experimental reports in the field were examined and the effectiveness of different types of tactile tasks was compared. Main results Individuals in the initial phases of Alzheimer's spectrum disease, specifically those in the stage of mild cognitive impairment (MCI), exhibit notable impairments in tasks involving tactile discrimination. These tasks possess certain merits, such as their quick and straightforward comparability, independence from educational background, and ability to circumvent the limitations associated with conventional cognitive assessment scales. Furthermore, tactile discrimination tasks offer enhanced accuracy compared to cognitive tasks that employ visual or auditory stimuli. Conclusions Tactile discrimination has the potential to serve as an innovative reference indicator for the swift diagnosis of clinical MCI patients, thereby assisting in the screening process on a clinical scale.
Collapse
Affiliation(s)
- Jinan Xu
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
- School of Mental Health, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuqi Sun
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
- School of Mental Health, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xianghe Zhu
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
- School of Mental Health, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Sipei Pan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhiqian Tong
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
- School of Mental Health, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ke Jiang
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
- School of Mental Health, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
- Center for Brain, Mind and Education, Shaoxing University, China
| |
Collapse
|
48
|
Thunborg C, Wang R, Rosenberg A, Sindi S, Andersen P, Andrieu S, Broersen LM, Coley N, Couderc C, Duval CZ, Faxen-Irving G, Hagman G, Hallikainen M, Håkansson K, Kekkonen E, Lehtisalo J, Levak N, Mangialasche F, Pantel J, Rydström A, Stigsdotter-Neely A, Wimo A, Ngandu T, Soininen H, Hartmann T, Solomon A, Kivipelto M. Integrating a multimodal lifestyle intervention with medical food in prodromal Alzheimer's disease: the MIND-AD mini randomized controlled trial. Alzheimers Res Ther 2024; 16:118. [PMID: 38812047 PMCID: PMC11138035 DOI: 10.1186/s13195-024-01468-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: 02/18/2024] [Accepted: 04/27/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) showed cognitive benefits from a multidomain lifestyle intervention in at-risk older people. The LipiDiDiet trial highlighted benefits of medical food in prodromal Alzheimer's disease (AD). However, the feasibility and impact of multimodal interventions combining lifestyle with medical food in prodromal AD is unclear. METHODS MIND-ADmini was a 6-month multinational (Sweden, Finland, Germany, France) proof-of-concept randomized controlled trial (RCT). Participants were 60-85 years old, had prodromal AD (International Working Group-1 criteria), and vascular/lifestyle risk factors. The parallel-group RCT had three arms: multimodal lifestyle intervention (nutritional guidance, exercise, cognitive training, vascular/metabolic risk management and social stimulation); multimodal lifestyle intervention + medical food (Fortasyn Connect); and regular health advice/care (control). Participants were randomized 1:1:1 (computer-generated allocation at each site). Outcome evaluators were blinded to randomization. Primary outcome was feasibility of the multimodal intervention, evaluated by recruitment rate during a 6-month recruitment phase, overall adherence in each intervention arm, and 6-month retention rate. Successful adherence was pre-specified as attending ≥ 40% of sessions/domain in ≥ 2/4 domains (lifestyle intervention), and consuming ≥ 60% of the medical food (lifestyle intervention + medical food). The secondary outcomes included adherence/participation to each intervention component and overall adherence to healthy lifestyle changes, measured using a composite score for healthy lifestyle. Cognitive assessments were included as exploratory outcomes, e.g. Clinical Dementia Rating scale. RESULTS During September 2017-May 2019, 93 individuals were randomized (32 lifestyle intervention, 31 lifestyle + medical food, and 30 control group). Overall recruitment rate was 76.2% (64.8% during the first 6 months). Overall 6-month retention rate was 91.4% (lifestyle intervention 87.5%; lifestyle + medical food 90.3%; control 96.7%). Domain-specific adherence in the lifestyle intervention group was 71.9% to cognitive training, 78.1% exercise, 68.8% nutritional guidance, and 81.3% vascular risk management; and in the lifestyle + medical food group, 90.3% to cognitive training, 87.1% exercise, 80.7% nutritional guidance, 87.1% vascular risk management, and 87.1% medical food. Compared with control, both intervention arms showed healthy diet improvements (βLifestyle×Time = 1.11, P = 0.038; βLifestyle+medical food×Time = 1.43, P = 0.007); the lifestyle + medical food group also showed vascular risk reduction (P = 0.043) and less cognitive-functional decline (P < 0.05, exploratory analysis). There were 5 serious adverse events (control group: 1; lifestyle intervention: 3; lifestyle + medical food: 1) unrelated to interventions. CONCLUSIONS The multidomain lifestyle intervention, alone or combined with medical food, had good feasibility and adherence in prodromal AD. Longer-term cognitive and other health benefits should be further investigated in a larger-scale trial. TRIAL REGISTRATION ClinicalTrials.gov NCT03249688.
Collapse
Affiliation(s)
- Charlotta Thunborg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden
- Department of Caring Sciences, Faculty of Health and Occupational Studies, Gävle, 801 76, Sweden
| | - Rui Wang
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Department of Physical Activity and Health, the Swedish School of Sport and Health Sciences, Stockholm, SE-114 86, Sweden
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave J5/1 Mezzanine, Madison, WI, 53792, USA
| | - Anna Rosenberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, Kuopio, 70210, Finland
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Pia Andersen
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden
| | - Sandrine Andrieu
- Department of Clinical Epidemiology and Public Health, CHU de Toulouse, and Aging Research Team, CERPOP Inserm UMR 1295, Jules Guesde, Toulouse, 31000, France
- IHU HealthAge, Toulouse, 31059, France
| | | | - Nicola Coley
- Department of Clinical Epidemiology and Public Health, CHU de Toulouse, and Aging Research Team, CERPOP Inserm UMR 1295, Jules Guesde, Toulouse, 31000, France
- IHU HealthAge, Toulouse, 31059, France
| | - Celine Couderc
- Department of Clinical Epidemiology and Public Health, CHU de Toulouse, and Aging Research Team, CERPOP Inserm UMR 1295, Jules Guesde, Toulouse, 31000, France
| | - Celine Z Duval
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, 66424, Germany
- Institute of General Practice, Goethe University Frankfurt a.M, Frankfurt, 60590, Germany
| | - Gerd Faxen-Irving
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
| | - Göran Hagman
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden
| | - Merja Hallikainen
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Krister Håkansson
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden
- Stockholm Gerontology Research Center, Tomtebodavägen 18a, Solna, 171 65, Sweden
| | - Eija Kekkonen
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Jenni Lehtisalo
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
- Population Health Unit, Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Mannerheimintie 166, P.O. Box 30, Helsinki, Finland
| | - Nicholas Levak
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden
| | - Francesca Mangialasche
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
| | - Johannes Pantel
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, 66424, Germany
- Institute of General Practice, Goethe University Frankfurt a.M, Frankfurt, 60590, Germany
| | - Anders Rydström
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
| | - Anna Stigsdotter-Neely
- Department of Social and Psychological Studies, Karlstad University, Karlstad, 651 88, Sweden
- Engineering Psychology, Luleå University of Technology, Luleå, 971 87, Sweden
| | - Anders Wimo
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
| | - Tiia Ngandu
- Population Health Unit, Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Mannerheimintie 166, P.O. Box 30, Helsinki, Finland
| | - Hilkka Soininen
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Tobias Hartmann
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, 66424, Germany
- Department of Experimental Neurology, Medical Faculty, Saarland University, Homburg, 66424, Germany
| | - Alina Solomon
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, Kuopio, 70210, Finland
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research QA32, Karolinska Institute, Karolinska Vägen 37 A, Solna, 171 64, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska Vägen 37 A, Solna, Stockholm, 171 64, Sweden.
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 8, Kuopio, 70210, Finland.
- The Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Mary's Hospital, Norfolk Place, London, W2 1PG, UK.
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 8, Kuopio, 70210, Finland.
| |
Collapse
|
49
|
Ali A, Milman S, Weiss EF, Gao T, Napolioni V, Barzilai N, Zhang ZD, Lin JR. Rare genetic coding variants associated with age-related episodic memory decline implicate distinct memory pathologies in the hippocampus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.21.24307692. [PMID: 38826255 PMCID: PMC11142267 DOI: 10.1101/2024.05.21.24307692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Approximately 40% of people aged 65 or older experience memory loss, particularly in episodic memory. Identifying the genetic basis of episodic memory decline is crucial for uncovering its underlying causes. Methods We investigated common and rare genetic variants associated with episodic memory decline in 742 (632 for rare variants) Ashkenazi Jewish individuals (mean age 75) from the LonGenity study. All-atom MD simulations were performed to uncover mechanistic insights underlying rare variants associated with episodic memory decline. Results In addition to the common polygenic risk of Alzheimer's Disease (AD), we identified and replicated rare variant association in ITSN1 and CRHR2 . Structural analyses revealed distinct memory pathologies mediated by interfacial rare coding variants such as impaired receptor activation of corticotropin releasing hormone and dysregulated L-serine synthesis. Discussion Our study uncovers novel risk loci for episodic memory decline. The identified underlying mechanisms point toward heterogeneous memory pathologies mediated by rare coding variants.
Collapse
|
50
|
Li Q, Li B, Liu L, Wang KJ, Liu MY, Deng Y, Li Z, Zhao WD, Wu LY, Chen YH, Zhang K. Monocytes release cystatin F dimer to associate with Aβ and aggravate amyloid pathology and cognitive deficits in Alzheimer's disease. J Neuroinflammation 2024; 21:125. [PMID: 38730470 PMCID: PMC11088181 DOI: 10.1186/s12974-024-03119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Understanding the molecular mechanisms of Alzheimer's disease (AD) has important clinical implications for guiding therapy. Impaired amyloid beta (Aβ) clearance is critical in the pathogenesis of sporadic AD, and blood monocytes play an important role in Aβ clearance in the periphery. However, the mechanism underlying the defective phagocytosis of Aβ by monocytes in AD remains unclear. METHODS Initially, we collected whole blood samples from sporadic AD patients and isolated the monocytes for RNA sequencing analysis. By establishing APP/PS1 transgenic model mice with monocyte-specific cystatin F overexpression, we assessed the influence of monocyte-derived cystatin F on AD development. We further used a nondenaturing gel to identify the structure of the secreted cystatin F in plasma. Flow cytometry, enzyme-linked immunosorbent assays and laser scanning confocal microscopy were used to analyse the internalization of Aβ by monocytes. Pull down assays, bimolecular fluorescence complementation assays and total internal reflection fluorescence microscopy were used to determine the interactions and potential interactional amino acids between the cystatin F protein and Aβ. Finally, the cystatin F protein was purified and injected via the tail vein into 5XFAD mice to assess AD pathology. RESULTS Our results demonstrated that the expression of the cystatin F protein was specifically increased in the monocytes of AD patients. Monocyte-derived cystatin F increased Aβ deposition and exacerbated cognitive deficits in APP/PS1 mice. Furthermore, secreted cystatin F in the plasma of AD patients has a dimeric structure that is closely related to clinical signs of AD. Moreover, we noted that the cystatin F dimer blocks the phagocytosis of Aβ by monocytes. Mechanistically, the cystatin F dimer physically interacts with Aβ to inhibit its recognition and internalization by monocytes through certain amino acid interactions between the cystatin F dimer and Aβ. We found that high levels of the cystatin F dimer protein in blood contributed to amyloid pathology and cognitive deficits as a risk factor in 5XFAD mice. CONCLUSIONS Our findings highlight that the cystatin F dimer plays a crucial role in regulating Aβ metabolism via its peripheral clearance pathway, providing us with a potential biomarker for diagnosis and potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Qiang Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bing Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Li Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Neurology, Shenyang Fifth People Hospital, Shenyang, 110023, China
| | - Kang-Ji Wang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Ming-Yue Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Ze Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| | - Li-Yong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| |
Collapse
|