1
|
Moon WJ. [Preface for Special Issue on Alzheimer's Disease: New Diagnostic Criteria, Treatment, and the Role of Neuroimaging]. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2025; 86:4-5. [PMID: 39958500 PMCID: PMC11822280 DOI: 10.3348/jksr.2025.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
|
2
|
Nelson PT. New criteria to predict LATE-NC in the clinical setting: Probable/Possible LATE and LANS. J Neuropathol Exp Neurol 2025; 84:2-7. [PMID: 39441698 PMCID: PMC11659605 DOI: 10.1093/jnen/nlae113] [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: 10/25/2024] Open
Abstract
This review discusses terminology recently proposed for the classification of dementia and, more specifically, nosology related to aging-associated TDP-43 pathology: limbic-predominant age-related TDP-43 encephalopathy (LATE), and limbic-predominant amnestic neurodegenerative syndrome (LANS). While the "gold standard" for these clinical conditions is still LATE neuropathologic changes (LATE-NC), clinical criteria and biomarkers are evolving. The newly proposed clinical rubrics are discussed with emphasis on the need for terminology that acknowledges the distinctions between clinical syndrome-, molecular biomarker-, and pathologically defined disease concepts. As further progress is made on research into the specific biomarker-based detection and prediction of TDP-43 proteinopathy in the clinical setting, the definitions of "Probable" and "Possible" LATE are likely to become more useful clinically. For people interested in the pathological diagnoses or basic research related to LATE-NC, the relevant terminology remains unchanged by the newly proposed clinical criteria.
Collapse
Affiliation(s)
- Peter T Nelson
- Sanders-Brown Center on Aging, Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, KY 40536, United States
| |
Collapse
|
3
|
Santamaría-García H, Migeot J, Medel V, Hazelton JL, Teckentrup V, Romero-Ortuno R, Piguet O, Lawor B, Northoff G, Ibanez A. Allostatic Interoceptive Overload Across Psychiatric and Neurological Conditions. Biol Psychiatry 2025; 97:28-40. [PMID: 38964530 PMCID: PMC12012852 DOI: 10.1016/j.biopsych.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Emerging theories emphasize the crucial role of allostasis (anticipatory and adaptive regulation of the body's biological processes) and interoception (integration, anticipation, and regulation of internal bodily states) in adjusting physiological responses to environmental and bodily demands. In this review, we explore the disruptions in integrated allostatic interoceptive mechanisms in psychiatric and neurological disorders, including anxiety, depression, Alzheimer's disease, and frontotemporal dementia. We assess the biological mechanisms associated with allostatic interoception, including whole-body cascades, brain structure and function of the allostatic interoceptive network, heart-brain interactions, respiratory-brain interactions, the gut-brain-microbiota axis, peripheral biological processes (inflammatory, immune), and epigenetic pathways. These processes span psychiatric and neurological conditions and call for developing dimensional and transnosological frameworks. We synthesize new pathways to understand how allostatic interoceptive processes modulate interactions between environmental demands and biological functions in brain disorders. We discuss current limitations of the framework and future transdisciplinary developments. This review opens a new research agenda for understanding how allostatic interoception involves brain predictive coding in psychiatry and neurology, allowing for better clinical application and the development of new therapeutic interventions.
Collapse
Affiliation(s)
- Hernando Santamaría-García
- Pontificia Universidad Javeriana, PhD program of Neuroscience, Bogotá, Colombia; Hospital Universitario San Ignacio, Centro de Memoria y Cognición Intellectus, Bogotá, Colombia
| | - Joaquin Migeot
- Global Brain Health Institute, University California of San Francisco, San Francisco, California; Global Brain Health Institute, Trinity College of Dublin, Dublin, Ireland; Latin American Brain Health Institute, Universidad Adolfo Ibanez, Santiago, Chile
| | - Vicente Medel
- Latin American Brain Health Institute, Universidad Adolfo Ibanez, Santiago, Chile
| | - Jessica L Hazelton
- Latin American Brain Health Institute, Universidad Adolfo Ibanez, Santiago, Chile; School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Vanessa Teckentrup
- School of Psychology and Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Roman Romero-Ortuno
- Pontificia Universidad Javeriana, PhD program of Neuroscience, Bogotá, Colombia; Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Olivier Piguet
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Brian Lawor
- Pontificia Universidad Javeriana, PhD program of Neuroscience, Bogotá, Colombia
| | - George Northoff
- Institute of Mental Health Research, Mind, Brain Imaging and Neuroethics Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Agustin Ibanez
- Global Brain Health Institute, University California of San Francisco, San Francisco, California; Global Brain Health Institute, Trinity College of Dublin, Dublin, Ireland; Latin American Brain Health Institute, Universidad Adolfo Ibanez, Santiago, Chile; School of Psychology and Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Bendayan M, Boitrelle F, Maurens-Hamdi S. Congenital absence of vas deferens: Anatomical and embryological inputs from a series of autopsies reported in Europe throughout the 18th and 19th century. Andrology 2024. [PMID: 39648616 DOI: 10.1111/andr.13815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
Congenital absence of the vas deferens (CAVD) is a syndrome with a heterogeneous presentation: bilateral (CBAVD) or unilateral (CUAVD), complete or partial and associated or not with other anomalies of the male urogenital system. A turning point came in 1968 when CBAVD was associated with cystic fibrosis and its CFTR gene mutations. Genetic studies then revealed that a minority of CBAVD but a majority of CUAVD are CFTR-independent. In the literature, reference is classically made to two sources from the 18th and 19th century: Hunter and Reverdin. This scarcity prompted us to look for additional observations of CAVD. By a meticulous bibliographical search, we identified a corpus of 10 European observations (8 CUAVD and 2 CBAVD) some of them richly illustrated. They were collected between 1755 and 1876 throughout adult men autopsies. We also provided their primary and unambiguous sources. Analysis of the reported data revealed some interesting facts: both CBAVD cases were unlikely linked to cystic fibrosis and half of CUAVD cases were associated with an ipsilateral kidney absence, suggesting a CFTR-independent pathophysiology. Moreover, the anatomical details of the anomalies raise interesting embryological questions we have tried to address in the light of current data. This work made it possible to identify new historical sources dealing with male genital tract pathologies. It sheds light on the origins of andrology and opens up interesting prospects for research and education in the field.
Collapse
Affiliation(s)
- Marion Bendayan
- Service de Biologie de la Reproduction-Andrologie-CECOS, Hôpital de Poissy Saint Germain en Laye, Poissy, France
- École Nationale Vétérinaire d'Alfort, Université Paris-Saclay, BREED, Maisons-Alfort, France
| | - Florence Boitrelle
- Service de Biologie de la Reproduction-Andrologie-CECOS, Hôpital de Poissy Saint Germain en Laye, Poissy, France
- École Nationale Vétérinaire d'Alfort, Université Paris-Saclay, BREED, Maisons-Alfort, France
| | - Safouane Maurens-Hamdi
- Centre d'épidémiologie et de recherche en santé des populations (CERPOP), Université Toulouse III Paul-Sabatier, Toulouse, France
| |
Collapse
|
5
|
Leung KHC, Chan VT, Lam BYK, Mok VCT, Cheung CY. Retinal vascular changes are associated with PET-based biomarkers of Alzheimer's disease: A pilot study. J Alzheimers Dis Rep 2024; 8:1639-1648. [PMID: 40034363 PMCID: PMC11863728 DOI: 10.1177/25424823241300416] [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: 01/05/2024] [Accepted: 10/17/2024] [Indexed: 03/05/2025] Open
Abstract
Background Retina is a non-invasive channel for assessing changes in brain microvasculature, which has been implicated in the pathophysiology of Alzheimer's disease (AD). Previous studies revealed significant relationship between clinically diagnosed AD and retinal vasculature. However, clinical diagnosis has limited sensitivity and specificity, and those investigations were conducted from traditional retinal fundus photographs which only captured a narrow section of the fundus. Objective Determining changes in retinal vasculature from larger area of retina between subjects with positron emission tomography (PET) biomarker-confirmed AD compared to controls. Methods Participants were recruited from the community and cognitive disorder clinics. Diagnosis of AD was confirmed by significant amyloid-β (Aβ) and tau uptake on PET scan. Retinal vasculature was imaged with ultra-widefield (UWF) scanning laser ophthalmoscopy and a series of vessel parameters were quantified using the semi-automated Singapore I Vessel Assessment (SIVA) software. Statistical analyses were adjusted for age, gender and systolic blood pressure. In addition, arteriole parameters were adjusted against the same measurements in venules, and vice versa. Results Out of the 39 patients, 18 had radiologically confirmed AD. These individuals with AD showed significantly smaller arteriolar fractal dimension (p = 0.032) in UWF images and greater venular tortuosity (p = 0.011) in standard fundus images compared with controls. Presence of significant Aβ and tau burden was associated with lower arteriolar caliber (OR 3.857; 95% CI 1.014-14.67; p = 0.048). Conclusions Reduction of fractal dimension in retinal arterioles observed in UWF imaging is associated with cerebral Aβ and tau burden in people with biomarker-confirmed AD. Wide field retinal imaging provides an alternative perspective in demonstrating microvascular alterations related to AD in this pilot study.
Collapse
Affiliation(s)
- Kristie Hing Chi Leung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Brighton and Sussex University Hospitals NHS Trust, West Sussex, UK
| | - Victor T.T. Chan
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, China
| | - Bonnie Yin Ka Lam
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Gerald Choa Neuroscience Institute, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Vincent CT Mok
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Gerald Choa Neuroscience Institute, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
6
|
Pollard CA, Saito ER, Burns JM, Hill JT, Jenkins TG. Considering Biomarkers of Neurodegeneration in Alzheimer's Disease: The Potential of Circulating Cell-Free DNA in Precision Neurology. J Pers Med 2024; 14:1104. [PMID: 39590596 PMCID: PMC11595805 DOI: 10.3390/jpm14111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), are a growing public health crisis, exacerbated by an aging global population and the lack of effective early disease-modifying therapies. Early detection of neurodegenerative disorders is critical to delaying symptom onset and mitigating disease progression, but current diagnostic tools often rely on detecting pathology once clinical symptoms have emerged and significant neuronal damage has already occurred. While disease-specific biomarkers, such as amyloid-beta and tau in AD, offer precise insights, they are too limited in scope for broader neurodegeneration screening for these conditions. Conversely, general biomarkers like neurofilament light chain (NfL) provide valuable staging information but lack targeted insights. Circulating cell-free DNA (cfDNA), released during cell death, is emerging as a promising biomarker for early detection. Derived from dying cells, cfDNA can capture both general neurodegenerative signals and disease-specific insights, offering multi-layered genomic and epigenomic information. Though its clinical potential remains under investigation, advances in cfDNA detection sensitivity, standardized protocols, and reference ranges could establish cfDNA as a valuable tool for early screening. cfDNA methylation signatures, in particular, show great promise for identifying tissue-of-origin and disease-specific changes, offering a minimally invasive biomarker that could transform precision neurology. However, further research is required to address technological challenges and validate cfDNA's utility in clinical settings. Here, we review recent work assessing cfDNA as a potential early biomarker in AD. With continued advances, cfDNA could play a pivotal role in shifting care from reactive to proactive, improving diagnostic timelines and patient outcomes.
Collapse
Affiliation(s)
- Chad A. Pollard
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| | | | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS 66205, USA
| | - Jonathon T. Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Timothy G. Jenkins
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| |
Collapse
|
7
|
Thussu S, Naidu A, Manivannan S, Grossberg GT. Profiling aducanumab as a treatment option for Alzheimer's disease: an overview of efficacy, safety and tolerability. Expert Rev Neurother 2024; 24:1045-1053. [PMID: 39291991 DOI: 10.1080/14737175.2024.2402058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Alzheimer's disease is the most common form of dementia worldwide. Aducanumab, a monoclonal antibody targeting amyloid-beta, became the first disease-modifying treatment for mild cognitive impairment due to Alzheimer's disease (AD) and mild AD dementia and suggested that removing amyloid from the brain, especially in early AD, might make a difference in slowing cognitive decline. AREAS COVERED In this review, the authors outline aducanumab's clinical efficacy as shown through key clinical trials and discuss its approval by the Food and Drug Administration under the accelerated pathway, which sparked both hope and controversy. We also discuss the importance of amyloid-related imaging abnormalities as a major side effect of aducanumab and all subsequent monoclonal antibodies targeting amyloid-beta. EXPERT OPINION Aducanumab, became the first monoclonal antibody that provided at least partial support for the amyloid hypothesis by demonstrating slowed cognitive decline by removing amyloid from the brain, although full FDA approval now seems unlikely due to discontinuation of its development. Its introduction raised awareness of ARIA, highlighted the significant costs and need for informed consent in treatment, and emphasized the importance of long-term, diverse, and combination therapy data for future AD treatments targeting amyloid and tau.
Collapse
Affiliation(s)
- Shreeya Thussu
- Department of Psychiatry and Behavioral Neuroscience, Division of Geriatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Aniketh Naidu
- Department of Psychiatry and Behavioral Neuroscience, Division of Geriatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Sindhu Manivannan
- Department of Psychiatry and Behavioral Neuroscience, Division of Geriatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - George T Grossberg
- Department of Psychiatry and Behavioral Neuroscience, Division of Geriatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
- Division of Geriatric Psychiatry, Inaugural Henry & Amelia Nasrallah Endowed, St. Louis, MO, USA
| |
Collapse
|
8
|
Phillips WT, Schwartz JG. Nasal lymphatic obstruction of CSF drainage as a possible cause of Alzheimer's disease and dementia. Front Aging Neurosci 2024; 16:1482255. [PMID: 39497786 PMCID: PMC11532075 DOI: 10.3389/fnagi.2024.1482255] [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: 08/17/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Alzheimer's disease, the most common form of dementia among older adults, slowly destroys memory and thinking skills. In recent years, scientists have made tremendous progress in understanding Alzheimer's disease, still, they do not yet fully understand what causes the disease. This article proposes a novel etiology for Alzheimer's disease. Our hypothesis developed from a review of nuclear medicine scans, in which the authors observed a significant increase in nasal turbinate vasodilation and blood pooling in patients with hypertension, sleep apnea, diabetes and/or obesity, all risk factors for Alzheimer's disease. The authors propose that nasal turbinate vasodilation and resultant blood pooling lead to the obstruction of normal nasal lymphatic clearance of cerebrospinal fluid and its waste products from the brain. The nasal turbinate vasodilation, due to increased parasympathetic activity, occurs alongside the well-established increased sympathetic activity of the cardiovascular system as seen in patients with hypertension. The increased parasympathetic activity is likely due to an autonomic imbalance secondary to the increase in worldwide consumption of highly processed food associated with dysregulation of the glucose regulatory system. The authors' hypothesis offers a novel mechanism and a new paradigm for the etiology of Alzheimer's disease and helps explain the rapid worldwide rise in the disease and other dementias which are expected to double in the next 20 years. This new paradigm provides compelling evidence for the modulation of the parasympathetic nervous system as a novel treatment strategy for Alzheimer's disease and other degenerative brain diseases, specifically targeting nasal turbinate lymphatic flow.
Collapse
|
9
|
Patel AA, Zhu MH, Yan R, Antic SD. Ex vivo propagation of synaptically-evoked cortical depolarizations in a mouse model of Alzheimer's disease at 20 Hz, 40 Hz, or 83 Hz. Sci Rep 2024; 14:23365. [PMID: 39375474 PMCID: PMC11458755 DOI: 10.1038/s41598-024-74262-2] [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: 05/27/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024] Open
Abstract
Sensory stimulations at 40 Hz gamma (but not any other frequency), have shown promise in reversing Alzheimer's disease (AD)-related pathologies. What distinguishes 40 Hz? We hypothesized that stimuli at 40 Hz might summate more efficiently (temporal summation) or propagate more efficiently between cortical layers (vertically), or along cortical laminas (horizontally), compared to inputs at 20 or 83 Hz. To investigate these hypotheses, we used brain slices from AD mouse model animals (5xFAD). Extracellular (synaptic) stimuli were delivered in cortical layer 4 (L4). Leveraging a fluorescent voltage indicator (VSFP) expressed in cortical pyramidal neurons, we simultaneously monitored evoked cortical depolarizations at multiple sites, at 1 kHz sampling frequency. Experimental groups (AD-Female, CTRL-Female, AD-Male, and CTRL-Male) were tested at three stimulation frequencies (20, 40, and 83 Hz). Despite our initial hypothesis, two parameters-temporal summation of voltage waveforms and the strength of propagation through the cortical neuropil-did not reveal any distinct advantage of 40 Hz stimulation. Significant physiological differences between AD and Control mice were found at all stimulation frequencies tested, while the 40 Hz stimulation frequency was not remarkable.
Collapse
Affiliation(s)
- Aayushi A Patel
- Department of Neuroscience, School of Medicine, UConn Health, Farmington, CT, 06030, USA
- Touro College of Osteopathic Medicine, Middletown, NY, 10940, USA
| | - Mei Hong Zhu
- Department of Neuroscience, School of Medicine, UConn Health, Farmington, CT, 06030, USA
| | - Riqiang Yan
- Department of Neuroscience, School of Medicine, UConn Health, Farmington, CT, 06030, USA
| | - Srdjan D Antic
- Department of Neuroscience, School of Medicine, UConn Health, Farmington, CT, 06030, USA.
| |
Collapse
|
10
|
Khan AF, Iturria-Medina Y. Beyond the usual suspects: multi-factorial computational models in the search for neurodegenerative disease mechanisms. Transl Psychiatry 2024; 14:386. [PMID: 39313512 PMCID: PMC11420368 DOI: 10.1038/s41398-024-03073-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
From Alzheimer's disease to amyotrophic lateral sclerosis, the molecular cascades underlying neurodegenerative disorders remain poorly understood. The clinical view of neurodegeneration is confounded by symptomatic heterogeneity and mixed pathology in almost every patient. While the underlying physiological alterations originate, proliferate, and propagate potentially decades before symptomatic onset, the complexity and inaccessibility of the living brain limit direct observation over a patient's lifespan. Consequently, there is a critical need for robust computational methods to support the search for causal mechanisms of neurodegeneration by distinguishing pathogenic processes from consequential alterations, and inter-individual variability from intra-individual progression. Recently, promising advances have been made by data-driven spatiotemporal modeling of the brain, based on in vivo neuroimaging and biospecimen markers. These methods include disease progression models comparing the temporal evolution of various biomarkers, causal models linking interacting biological processes, network propagation models reproducing the spatial spreading of pathology, and biophysical models spanning cellular- to network-scale phenomena. In this review, we discuss various computational approaches for integrating cross-sectional, longitudinal, and multi-modal data, primarily from large observational neuroimaging studies, to understand (i) the temporal ordering of physiological alterations, i(i) their spatial relationships to the brain's molecular and cellular architecture, (iii) mechanistic interactions between biological processes, and (iv) the macroscopic effects of microscopic factors. We consider the extents to which computational models can evaluate mechanistic hypotheses, explore applications such as improving treatment selection, and discuss how model-informed insights can lay the groundwork for a pathobiological redefinition of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ahmed Faraz Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada.
| |
Collapse
|
11
|
Hendriksen HMA, de Rijke TJ, Fruijtier A, van de Giessen E, van Harten AC, van Leeuwenstijn‐Koopman MSSA, van der Schaar J, Trieu C, Visser D, Smets EMA, Visser LNC, van der Flier WM. Amyloid PET disclosure in subjective cognitive decline: Patient experiences over time. Alzheimers Dement 2024; 20:6556-6565. [PMID: 39087383 PMCID: PMC11497681 DOI: 10.1002/alz.14148] [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: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION We disclosed amyloid positron emission tomography (PET) results in individuals with subjective cognitive decline (SCD) and studied patient experiences and outcomes over a 6-month period. METHODS Fifty-seven participants from the Subjective Cognitive Impairment Cohort (SCIENCe) (66 ± 8 years, 21 [37%] F, Mini-Mental State Examination 29 ± 1, 15 [26%] amyloid positive [A+]) completed questionnaires 1 week prior (T0), 1 day after (T1), and 6 months after amyloid PET disclosure (T2). Questionnaires addressed patient-reported experiences and outcomes. RESULTS Independent of amyloid status, participants were satisfied with the consultation (scale 1-10; 7.9 ± 1.7) and information provided (scale 1-4; T1: 3.3 ± 0.9, T2: 3.2 ± 0.8). After 6 months, A+ participants reported more information needs (45% vs. 12%, p = 0.02). Independent of amyloid status, decision regret (scale 1-5; A+: 1.5 ± 0.9, A-: 1.4 ± 0.6, p = 0.53) and negative emotions (negative affect, uncertainty, anxiety) were low (all p > 0.15 and Pinteraction > 0.60). DISCUSSION Participants with SCD valued amyloid PET disclosure positively, regardless of amyloid status. The need for information after 6 months, which was stronger in A+ individuals, underscores the importance of follow-up. HIGHLIGHTS Participants with subjective cognitive decline (SCD) positively valued amyloid positron emission tomography (PET) disclosure. Participants with SCD experienced low levels of decision regret. We did not observe an increase in negative emotions. After 6 months, amyloid-positive individuals wanted more information.
Collapse
Affiliation(s)
- Heleen M. A. Hendriksen
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Tanja J. de Rijke
- Medical PsychologyAmsterdam UMC location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Amsterdam Public HealthQuality of CarePersonalized MedicineAmsterdamthe Netherlands
| | - Agnetha Fruijtier
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Medical PsychologyAmsterdam UMC location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Amsterdam Public HealthQuality of CarePersonalized MedicineAmsterdamthe Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamthe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Mardou S. S. A. van Leeuwenstijn‐Koopman
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Jetske van der Schaar
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Calvin Trieu
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Denise Visser
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamthe Netherlands
| | - Ellen M. A. Smets
- Medical PsychologyAmsterdam UMC location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Amsterdam Public HealthQuality of CarePersonalized MedicineAmsterdamthe Netherlands
| | - Leonie N. C. Visser
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Medical PsychologyAmsterdam UMC location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Amsterdam Public HealthQuality of CarePersonalized MedicineAmsterdamthe Netherlands
- Division of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Wiesje M. van der Flier
- Alzheimer Center AmsterdamDepartment of NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Epidemiology and Data ScienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamthe Netherlands
| |
Collapse
|
12
|
Marques BL, Campos AC. Cannabidiol and Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 177:121-134. [PMID: 39029982 DOI: 10.1016/bs.irn.2024.04.014] [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: 07/21/2024]
Abstract
Alzheimer's disease (AD) stands as the most prevalent form of neuropsychiatric disorder among the elderly population, impacting a minimum of 50 million individuals worldwide. Current pharmacological treatments rely on the prescribing cholinesterase inhibitors and memantine. However,recently anecdotal findings based on low-quality real-world data had prompted physicians, patients, and their relatives to consider the use of cannabinoids, especially Cannabidiol (CBD), for alleviating of AD symptoms. CBD the primary non-psychotomimetic compound found in the Cannabis sp. plant, exhibits promising therapeutic potential across various clinical contexts. Pre-clinical and in vitro studies indicate that CBD could mitigate cognitive decline and amyloid-beta-induced neurodegeneration by modulating oxidative stress and neuroinflammation. In addition, CBD demonstrates significant effects in promoting neuroplasticity, particularly in brain regions such as the hippocampus. However, the available clinical evidence presents conflicting results, and no randomized placebo-controlled trials have been published to date. In conclusion, although pre-clinical and in vitro studies offer encouraging insights into the potential benefits of CBD in AD models, new and well-designed clinical trials are imperative to ascertain the clinical relevance of CBD use in the management of AD symptoms, especially in comparison to conventional treatments.
Collapse
Affiliation(s)
- Bruno L Marques
- Pharmacology of Neuroplasticity Lab, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil; Center for Cannabinoid Research, Mental Health Building, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Alline C Campos
- Pharmacology of Neuroplasticity Lab, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil; Center for Cannabinoid Research, Mental Health Building, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
13
|
Zahr NM. Alcohol Use Disorder and Dementia: A Review. Alcohol Res 2024; 44:03. [PMID: 38812709 PMCID: PMC11135165 DOI: 10.35946/arcr.v44.1.03] [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: 05/31/2024] Open
Abstract
PURPOSE By 2040, 21.6% of Americans will be over age 65, and the population of those older than age 85 is estimated to reach 14.4 million. Although not causative, older age is a risk factor for dementia: every 5 years beyond age 65, the risk doubles; approximately one-third of those older than age 85 are diagnosed with dementia. As current alcohol consumption among older adults is significantly higher compared to previous generations, a pressing question is whether drinking alcohol increases the risk for Alzheimer's disease or other forms of dementia. SEARCH METHODS Databases explored included PubMed, Web of Science, and ScienceDirect. To accomplish this narrative review on the effects of alcohol consumption on dementia risk, the literature covered included clinical diagnoses, epidemiology, neuropsychology, postmortem pathology, neuroimaging and other biomarkers, and translational studies. Searches conducted between January 12 and August 1, 2023, included the following terms and combinations: "aging," "alcoholism," "alcohol use disorder (AUD)," "brain," "CNS," "dementia," "Wernicke," "Korsakoff," "Alzheimer," "vascular," "frontotemporal," "Lewy body," "clinical," "diagnosis," "epidemiology," "pathology," "autopsy," "postmortem," "histology," "cognitive," "motor," "neuropsychological," "magnetic resonance," "imaging," "PET," "ligand," "degeneration," "atrophy," "translational," "rodent," "rat," "mouse," "model," "amyloid," "neurofibrillary tangles," "α-synuclein," or "presenilin." When relevant, "species" (i.e., "humans" or "other animals") was selected as an additional filter. Review articles were avoided when possible. SEARCH RESULTS The two terms "alcoholism" and "aging" retrieved about 1,350 papers; adding phrases-for example, "postmortem" or "magnetic resonance"-limited the number to fewer than 100 papers. Using the traditional term, "alcoholism" with "dementia" resulted in 876 citations, but using the currently accepted term "alcohol use disorder (AUD)" with "dementia" produced only 87 papers. Similarly, whereas the terms "Alzheimer's" and "alcoholism" yielded 318 results, "Alzheimer's" and "alcohol use disorder (AUD)" returned only 40 citations. As pertinent postmortem pathology papers were published in the 1950s and recent animal models of Alzheimer's disease were created in the early 2000s, articles referenced span the years 1957 to 2024. In total, more than 5,000 articles were considered; about 400 are herein referenced. DISCUSSION AND CONCLUSIONS Chronic alcohol misuse accelerates brain aging and contributes to cognitive impairments, including those in the mnemonic domain. The consensus among studies from multiple disciplines, however, is that alcohol misuse can increase the risk for dementia, but not necessarily Alzheimer's disease. Key issues to consider include the reversibility of brain damage following abstinence from chronic alcohol misuse compared to the degenerative and progressive course of Alzheimer's disease, and the characteristic presence of protein inclusions in the brains of people with Alzheimer's disease, which are absent in the brains of those with AUD.
Collapse
Affiliation(s)
- Natalie M Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California. Center for Health Sciences, SRI International, Menlo Park, California
| |
Collapse
|
14
|
Chen H, Zhang CJ, Zhao ZY, Gao YY, Zhao JT, Li XX, Zhang M, Wang H. Mechanisms underlying LncRNA SNHG1 regulation of Alzheimer's disease involve DNA methylation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:428-435. [PMID: 38551404 DOI: 10.1080/15287394.2024.2334248] [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: 04/10/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease associated with long non-coding RNAs and DNA methylation; however, the mechanisms underlying the role of lncRNA small nucleolar RNA host gene 1 (lncRNA SNHG1) and subsequent involvement of DNA methylation in AD development are not known. The aim of this study was to examine the regulatory mechanisms attributed to lncRNA SNHG1 gene utilizing 2 strains of senescence-accelerated mouse prone 8 (SAMP8) model of AD and compared to senescence-accelerated mouse resistant (SAMR) considered a control. Both strains of the mouse were transfected with either blank virus, psLenti-U6-SNHG1(low gene expression) virus, and psLenti-pA-SNHG1(gene overexpression) virus via a single injection into the brains for 2 weeks. At 2 weeks mice were subjected to a Morris water maze to determine any behavioral effects followed by sacrifice to extract hippocampal tissue for Western blotting to measure protein expression of p-tau, DNMT1, DNMT3A, DNMT3B, TET1, and p-Akt. No marked alterations were noted in any parameters following blank virus transfection. In SAMP8 mice, a significant decrease was noted in protein expression of DNMT1, DNMT3A, DNMT3B, and p-Akt associated with rise in p-tau and TET1. Transfection with ps-Lenti-U6-SNHG1 alone in SAMR1 mice resulted in a significant rise in DNMTs and p-Akt and a fall in p-tau and TET1. Transfection of SAMP8 with ps-Lenti-U6-SNHG1 blocked effects on overexpression noted in this mouse strain. However, knockdown of lncRNA SNHG1 yielded the opposite results as found in SAMR1 mice. In conclusion, the knockdown of lncRNA SNHG1 enhanced DNA methylation through the PI3K/Akt signaling pathway, thereby reducing the phosphorylation levels of tau in SAMP8 AD model mice with ameliorating brain damage attributed to p-tau accumulation with consequent neuroprotection.
Collapse
Affiliation(s)
- Hong Chen
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Chun-Jie Zhang
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
- Center of Collaborative Innovation in Translational Medicine, Baotou Medical College, Inner Mongolia, China
| | - Zhi-Ying Zhao
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Yang-Yang Gao
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Jian-Tian Zhao
- Institute of Public Health, Baotou Medical College, Inner Mongolia, China
| | - Xiao-Xu Li
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - Ming Zhang
- Institute of Neuroscience and Medical Technology, Department of Anatomy, Baotou Medical College, Inner Mongolia, China
| | - He Wang
- School of Health Sciences, University of Newcastle, Newcastle, Australia
| |
Collapse
|
15
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
16
|
Hamilton LK, M'Bra PEH, Mailloux S, Galoppin M, Aumont A, Fernandes KJL. Central inhibition of stearoyl-CoA desaturase has minimal effects on the peripheral metabolic symptoms of the 3xTg Alzheimer's disease mouse model. Sci Rep 2024; 14:7742. [PMID: 38565895 PMCID: PMC10987571 DOI: 10.1038/s41598-024-58272-8] [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: 09/18/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Evidence from genetic and epidemiological studies point to lipid metabolism defects in both the brain and periphery being at the core of Alzheimer's disease (AD) pathogenesis. Previously, we reported that central inhibition of the rate-limiting enzyme in monounsaturated fatty acid synthesis, stearoyl-CoA desaturase (SCD), improves brain structure and function in the 3xTg mouse model of AD (3xTg-AD). Here, we tested whether these beneficial central effects involve recovery of peripheral metabolic defects, such as fat accumulation and glucose and insulin handling. As early as 3 months of age, 3xTg-AD mice exhibited peripheral phenotypes including increased body weight and visceral and subcutaneous white adipose tissue as well as diabetic-like peripheral gluco-regulatory abnormalities. We found that intracerebral infusion of an SCD inhibitor that normalizes brain fatty acid desaturation, synapse loss and learning and memory deficits in middle-aged memory-impaired 3xTg-AD mice did not affect these peripheral phenotypes. This suggests that the beneficial effects of central SCD inhibition on cognitive function are not mediated by recovery of peripheral metabolic abnormalities. Given the widespread side-effects of systemically administered SCD inhibitors, these data suggest that selective inhibition of SCD in the brain may represent a clinically safer and more effective strategy for AD.
Collapse
Affiliation(s)
- Laura K Hamilton
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Paule E H M'Bra
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Sophia Mailloux
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Manon Galoppin
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Anne Aumont
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Karl J L Fernandes
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada.
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada.
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada.
| |
Collapse
|
17
|
Therriault J, Schindler SE, Salvadó G, Pascoal TA, Benedet AL, Ashton NJ, Karikari TK, Apostolova L, Murray ME, Verberk I, Vogel JW, La Joie R, Gauthier S, Teunissen C, Rabinovici GD, Zetterberg H, Bateman RJ, Scheltens P, Blennow K, Sperling R, Hansson O, Jack CR, Rosa-Neto P. Biomarker-based staging of Alzheimer disease: rationale and clinical applications. Nat Rev Neurol 2024; 20:232-244. [PMID: 38429551 DOI: 10.1038/s41582-024-00942-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Disease staging, whereby the spatial extent and load of brain pathology are used to estimate the severity of Alzheimer disease (AD), is pivotal to the gold-standard neuropathological diagnosis of AD. Current in vivo diagnostic frameworks for AD are based on abnormal concentrations of amyloid-β and tau in the cerebrospinal fluid or on PET scans, and breakthroughs in molecular imaging have opened up the possibility of in vivo staging of AD. Focusing on the key principles of disease staging shared across several areas of medicine, this Review highlights the potential for in vivo staging of AD to transform our understanding of preclinical AD, refine enrolment criteria for trials of disease-modifying therapies and aid clinical decision-making in the era of anti-amyloid therapeutics. We provide a state-of-the-art review of recent biomarker-based AD staging systems and highlight their contributions to the understanding of the natural history of AD. Furthermore, we outline hypothetical frameworks to stage AD severity using more accessible fluid biomarkers. In addition, by applying amyloid PET-based staging to recently published anti-amyloid therapeutic trials, we highlight how biomarker-based disease staging frameworks could illustrate the numerous pathological changes that have already taken place in individuals with mildly symptomatic AD. Finally, we discuss challenges related to the validation and standardization of disease staging and provide a forward-looking perspective on potential clinical applications.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Liana Apostolova
- Department of Neurology, University of Indiana School of Medicine, Indianapolis, IN, USA
| | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Bermejo-Pareja F, del Ser T. Controversial Past, Splendid Present, Unpredictable Future: A Brief Review of Alzheimer Disease History. J Clin Med 2024; 13:536. [PMID: 38256670 PMCID: PMC10816332 DOI: 10.3390/jcm13020536] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Background: The concept of Alzheimer disease (AD)-since its histological discovery by Alzheimer to the present day-has undergone substantial modifications. Methods: We conducted a classical narrative review of this field with a bibliography selection (giving preference to Medline best match). Results: The following subjects are reviewed and discussed: Alzheimer's discovery, Kraepelin's creation of a new disease that was a rare condition until the 1970's, the growing interest and investment in AD as a major killer in a society with a large elderly population in the second half of the 20th century, the consolidation of the AD clinicopathological model, and the modern AD nosology based on the dominant amyloid hypothesis among many others. In the 21st century, the development of AD biomarkers has supported a novel biological definition of AD, although the proposed therapies have failed to cure this disease. The incidence of dementia/AD has shown a decrease in affluent countries (possibly due to control of risk factors), and mixed dementia has been established as the most frequent etiology in the oldest old. Conclusions: The current concept of AD lacks unanimity. Many hypotheses attempt to explain its complex physiopathology entwined with aging, and the dominant amyloid cascade has yielded poor therapeutic results. The reduction in the incidence of dementia/AD appears promising but it should be confirmed in the future. A reevaluation of the AD concept is also necessary.
Collapse
Affiliation(s)
- Félix Bermejo-Pareja
- CIBERNED, Institute of Health Carlos III, 28029 Madrid, Spain
- Institute of Research i+12, University Hospital “12 de Octubre”, 28041 Madrid, Spain
| | - Teodoro del Ser
- Alzheimer’s Centre Reina Sofia—CIEN Foundation, Institute of Health Carlos III, 28031 Madrid, Spain;
| |
Collapse
|
19
|
Høilund-Carlsen PF, Alavi A, Barrio JR, Castellani RJ, Costa T, Herrup K, Kepp KP, Neve RL, Perry G, Revheim ME, Robakis NK, Sensi SL, Vissel B. Revision of Alzheimer's diagnostic criteria or relocation of the Potemkin village. Ageing Res Rev 2024; 93:102173. [PMID: 38104639 DOI: 10.1016/j.arr.2023.102173] [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: 10/31/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
The recently announced revision of the Alzheimer's disease (AD) diagnostic ATN classification adds to an already existing disregard for clinical assessment the rejection of image-based in vivo assessment of the brain's condition. The revision suggests that the diagnosis of AD should be based solely on the presence of cerebral amyloid-beta and tau, indicated by the "A" and "T". The "N", which stands for neurodegeneration - detected by imaging - should no longer be given importance, except that A+ ± T + = AD with amyloid PET being the main method for demonstrating A+ . We believe this is an artificial and misleading suggestion. It is artificial because it relies on biomarkers whose significance remains obscure and where the detection of "A" is based on a never-validated PET method using a tracer that marks much more than amyloid-beta. It is misleading because many patients without dementia will be falsely classified as having AD, but nonetheless candidates for passive immunotherapy, which may be more harmful than beneficial, and sometimes fatal.
Collapse
Affiliation(s)
- Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA, USA
| | - Rudolph J Castellani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tommaso Costa
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; FOCUS Lab, Department of Psychology, University of Turin, Turin, Italy
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kasper P Kepp
- Section of Biophysical and Biomedicinal Chemistry, DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, MA, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Mona-Elisabeth Revheim
- The Intervention Centre, Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nikolaos K Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY, USA
| | - Stefano L Sensi
- Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; CAST-Center for Advanced Studies and Technology, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; ITAB-Institute of Advanced Biomedical Technology, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Bryce Vissel
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus Faculty of Medicine and Health, UNSW, Sydney, Australia; St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| |
Collapse
|
20
|
Høilund-Carlsen PF, Alavi A, Barrio JR. PET/CT/MRI in Clinical Trials of Alzheimer's Disease. J Alzheimers Dis 2024; 101:S579-S601. [PMID: 39422954 DOI: 10.3233/jad-240206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
With the advent of PET imaging in 1976, 2-deoxy-2-[18F]fluoro-D-glucose (FDG)-PET became the preferred method for in vivo investigation of cerebral processes, including regional hypometabolism in Alzheimer's disease. With the emergence of amyloid-PET tracers, [11C]Pittsburgh Compound-B in 2004 and later [18F]florbetapir, [18F]florbetaben, and [18F]flumetamol, amyloid-PET has replaced FDG-PET in Alzheimer's disease anti-amyloid clinical trial treatments to ensure "amyloid positivity" as an entry criterion, and to measure treatment-related decline in cerebral amyloid deposits. MRI has been used to rule out other brain diseases and screen for 'amyloid-related imaging abnormalities' (ARIAs) of two kinds, ARIA-E and ARIA-H, characterized by edema and micro-hemorrhage, respectively, and, to a lesser extent, to measure changes in cerebral volumes. While early immunotherapy trials of Alzheimer's disease showed no clinical effects, newer monoclonal antibody trials reported decreases of 27% to 85% in the cerebral amyloid-PET signal, interpreted by the Food and Drug Administration as amyloid removal expected to result in a reduction in clinical decline. However, due to the lack of diagnostic specificity of amyloid-PET tracers, amyloid positivity cannot prevent the inclusion of non-Alzheimer's patients and even healthy subjects in these clinical trials. Moreover, the "decreasing amyloid accumulation" assessed by amyloid-PET imaging has questionable quantitative value in the presence of treatment-related brain damage (ARIAs). Therefore, future Alzheimer's clinical trials should disregard amyloid-PET imaging and focus instead on assessment of regional brain function by FDG-PET and MRI monitoring of ARIAs and brain volume loss in all trial patients.
Collapse
Affiliation(s)
- Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
21
|
Fromm D, Dalton SG, Brick A, Olaiya G, Hill S, Greenhouse J, MacWhinney B. The Case of the Cookie Jar: Differences in Typical Language Use in Dementia. J Alzheimers Dis 2024; 100:1417-1434. [PMID: 38995772 PMCID: PMC11380261 DOI: 10.3233/jad-230844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Background Findings from language sample analyses can provide efficient and effective indicators of cognitive impairment in older adults. Objective This study used newly automated core lexicon analyses of Cookie Theft picture descriptions to assess differences in typical use across three groups. Methods Participants included adults without diagnosed cognitive impairments (Control), adults diagnosed with Alzheimer's disease (ProbableAD), and adults diagnosed with mild cognitive impairment (MCI). Cookie Theft picture descriptions were transcribed and analyzed using CLAN. Results Results showed that the ProbableAD group used significantly fewer core lexicon words overall than the MCI and Control groups. For core lexicon content words (nouns, verbs), however, both the MCI and ProbableAD groups produced significantly fewer words than the Control group. The groups did not differ in their use of core lexicon function words. The ProbableAD group was also slower to produce most of the core lexicon words than the MCI and Control groups. The MCI group was slower than the Control group for only two of the core lexicon content words. All groups mentioned a core lexicon word in the top left quadrant of the picture early in the description. The ProbableAD group was then significantly slower than the other groups to mention a core lexicon word in the other quadrants. Conclusions This standard and simple-to-administer task reveals group differences in overall core lexicon scores and the amount of time until the speaker produces the key items. Clinicians and researchers can use these tools for both early assessment and measurement of change over time.
Collapse
Affiliation(s)
- Davida Fromm
- Department of Psychology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Sarah Grace Dalton
- Department of Speech Pathology and Audiology, Marquette University, Milwaukee, WI, USA
| | - Alexander Brick
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Gbenuola Olaiya
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Sophia Hill
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Joel Greenhouse
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Brian MacWhinney
- Department of Psychology, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Petersen RC, Weintraub S, Sabbagh M, Karlawish J, Adler CH, Dilworth-Anderson P, Frank L, Huling Hummel C, Taylor A. A New Framework for Dementia Nomenclature. JAMA Neurol 2023; 80:1364-1370. [PMID: 37843871 DOI: 10.1001/jamaneurol.2023.3664] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Importance Nomenclature in the field of neurodegenerative diseases presents a challenging problem. Inconsistent use of terms such as Alzheimer disease and dementia has compromised progress in clinical care, research, and development of therapeutics. Dementia-associated stigma further contributes to inconsistent and imprecise language. The result is a lack of clarity that produces confusion with patients and the general public and presents communication challenges among researchers. Therefore, the Advisory Council on Research, Care, and Services of the National Plan to Address Alzheimer's Disease authorized a committee to make recommendations for improvement. Objective To establish a systematic neurodegenerative disease framework for information collection and communication to standardize language usage for research, clinical, and public health purposes. Evidence Review The Dementia Nomenclature Initiative organized into 3 major stakeholder working groups: clinicians, researchers, and the public (including individuals living with dementia and family caregivers). To inform the work, the initiative completed a narrative literature review of dementia nomenclature evolution over the last century across the PubMed, CINAHL, PsycInfo, and Scopus databases (January 1, 2000, through July 31, 2020). Initiative working groups used the results as a foundation for understanding current challenges with dementia nomenclature and implications for research, clinical practice, and public understanding. The initiative obtained additional input via focus groups with individuals living with dementia and caregivers, with separate groups for race and ethnicity (American Indian or Alaska Native, Asian or Pacific Islander, Black or African American, Hispanic or Latino, and White) as an initial assessment of the meaning of dementia-related terms to these groups. Findings From working group deliberations, the literature review, and focus group input, the initiative developed a framework clearly separating the clinical syndromic presentation experienced by affected individuals from possible underlying pathophysiologies. In the framework, domains of clinical impairment, such as cognitive, behavioral, motor, and other neurologic features, are graded by level of impairment between none and severe. Next, biomarker information describes underlying disease processes, explains the syndrome, and identifies possible disease labels: Alzheimer disease, frontotemporal degeneration, dementia with Lewy bodies, or vascular cognitive impairment dementia. Conclusions and Relevance The Dementia Nomenclature Initiative established a framework to guide communication about cognitive impairment among older adults. Wider testing and refinement of the framework will subsequently improve the information used in communicating about cognitive impairment and the way in which the information is used in clinical, research, and public settings.
Collapse
Affiliation(s)
- Ronald C Petersen
- Department of Neurology, Alzheimer's Disease Research Center, Mayo Clinic, Rochester, Minnesota
| | - Sandra Weintraub
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Marwan Sabbagh
- Alzheimer's and Memory Disorders Program, Barrow Neurological Institute, Phoenix, Arizona
| | - Jason Karlawish
- Penn Memory Center, Departments of Medicine, Medical Ethics, and Health Policy, and Neurology, University of Pennsylvania, Philadelphia
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Phoenix
| | | | - Lori Frank
- New York Academy of Medicine, New York, New York
| | | | - Angela Taylor
- Strategic Partnerships, Lewy Body Dementia Association, Lilburn, Georgia
| |
Collapse
|
23
|
van der Schaar J, Visser LNC, Ket JCF, Groot C, Pijnenburg YAL, Scheltens P, Bredenoord AL, van den Hoven MA, van der Flier WM. Impact of sharing Alzheimer's disease biomarkers with individuals without dementia: A systematic review and meta-analysis of empirical data. Alzheimers Dement 2023; 19:5773-5794. [PMID: 37496313 DOI: 10.1002/alz.13410] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/01/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION We conducted a systematic literature review and meta-analysis of empirical evidence on expected and experienced implications of sharing Alzheimer's disease (AD) biomarker results with individuals without dementia. METHODS PubMed, Embase, APA PsycInfo, and Web of Science Core Collection were searched according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Results from included studies were synthesized, and quantitative data on psychosocial impact were meta-analyzed using a random-effects model. RESULTS We included 35 publications. Most personal stakeholders expressed interest in biomarker assessment. Learning negative biomarker results led to relief and sometimes frustration, while positive biomarkers induced anxiety but also clarity. Meta-analysis of five studies including 2012 participants (elevated amyloid = 1324 [66%], asymptomatic = 1855 [92%]) showed short-term psychological impact was not significant (random-effect estimate = 0.10, standard error = 0.23, P = 0.65). Most professional stakeholders valued biomarker testing, although attitudes and practices varied considerably. DISCUSSION Interest in AD biomarker testing was high and sharing their results did not cause psychological harm. HIGHLIGHTS Most personal stakeholders expressed interest in Alzheimer's disease biomarker assessment. Personal motivations included gaining insight, improving lifestyle, or preparing for the future. There was no short-term psychological impact of sharing biomarker status, implying it can be safe. Most professional stakeholders valued biomarker testing, believing the benefits outweigh the risk. Harmonized guidelines on biomarker testing and sharing results are required.
Collapse
Affiliation(s)
- Jetske van der Schaar
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Leonie N C Visser
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Medical Psychology, Amsterdam UMC location University of Amsterdam/AMC, Amsterdam, the Netherlands
- Amsterdam Public Health, Quality of Care, Amsterdam, the Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Johannes C F Ket
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Colin Groot
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- EQT Life Sciences, Amsterdam, the Netherlands
| | - Annelien L Bredenoord
- Erasmus School of Philosophy, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | | | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Epidemiology & Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Waziry R, Williams OA. Alzheimer Disease: A Life-Limiting Disease With a Burden of a Terminal Illness. Neurol Clin Pract 2023; 13:e200208. [PMID: 37854176 PMCID: PMC10581074 DOI: 10.1212/cpj.0000000000200208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
Purpose of Review An estimated 6.5 million Americans live with Alzheimer dementia. Better understanding of advanced stages of Alzheimer disease (AD) and timely monitoring of its preventable complications would translate to improved survival and quality of life in this population. Recent Findings In this perspective review, we proposed shifting the narrative to recognizing AD as a chronic life-limiting illness instead of a terminal disease. In support of this view, we discussed the following: (1) the biochemical, cellular (preclinical), and clinical phases of AD; (2) survival after AD; (3) AD therapeutics and potential implications for the population with AD in the advanced stages. Summary On the bases of the prolonged preclinical phase in AD, promising advances in AD therapeutics and the varying survival after AD, we proposed a new classification for AD and more broadly neurodegenerative disorders to be recognized as chronic life-limiting illnesses rather than terminal diseases with important implications for patients with AD in the advanced stages given the challenges that are specific to this population.
Collapse
Affiliation(s)
- Reem Waziry
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, NY
| | - Olajide A Williams
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, NY
| |
Collapse
|
25
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
26
|
Zhao H, Wang J, Li Z, Wang S, Yu G, Wang L. Identification ferroptosis-related hub genes and diagnostic model in Alzheimer's disease. Front Mol Neurosci 2023; 16:1280639. [PMID: 37965040 PMCID: PMC10642492 DOI: 10.3389/fnmol.2023.1280639] [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: 08/21/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Background Ferroptosis is a newly defined form of programmed cell death and plays an important role in Alzheimer's disease (AD) pathology. This study aimed to integrate bioinformatics techniques to explore biomarkers to support the correlation between ferroptosis and AD. In addition, further investigation of ferroptosis-related biomarkers was conducted on the transcriptome characteristics in the asymptomatic AD (AsymAD). Methods The microarray datasets GSE118553, GSE132903, GSE33000, and GSE157239 on AD were downloaded from the GEO database. The list of ferroptosis-related genes was extracted from the FerrDb website. Differentially expressed genes (DEGs) were identified by R "limma" package and used to screen ferroptosis-related hub genes. The random forest algorithm was used to construct the diagnostic model through hub genes. The immune cell infiltration was also analyzed by CIBERSORTx. The miRNet and DGIdb database were used to identify microRNAs (miRNAs) and drugs which targeting hub genes. Results We identified 18 ferroptosis-related hub genes anomalously expressed in AD, and consistent expression trends had been observed in both AsymAD The random forest diagnosis model had good prediction results in both training set (AUC = 0.824) and validation set (AUC = 0.734). Immune cell infiltration was analyzed and the results showed that CD4+ T cells resting memory, macrophages M2 and neutrophils were significantly higher in AD. A significant correlation of hub genes with immune infiltration was observed, such as DDIT4 showed strong positive correlation with CD4+ T cells memory resting and AKR1C2 had positive correlation with Macrophages M2. Additionally, the microRNAs (miRNAs) and drugs which targeting hub genes were screened. Conclusion These results suggest that ferroptosis-related hub genes we screened played a part in the pathological progression of AD. We explored the potential of these genes as diagnostic markers and their relevance to immune cells which will help in understanding the development of AD. Targeting miRNAs and drugs provides new research clues for preventing the development of AD.
Collapse
Affiliation(s)
| | | | | | | | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Sciences, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Sciences, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
27
|
Flores-Sandoval AA, Davila-Pérez P, Buss SS, Donohoe K, O'Connor M, Shafi MM, Pascual-Leone A, Benwell CSY, Fried PJ. Spectral power ratio as a measure of EEG changes in mild cognitive impairment due to Alzheimer's disease: a case-control study. Neurobiol Aging 2023; 130:50-60. [PMID: 37459658 PMCID: PMC10614059 DOI: 10.1016/j.neurobiolaging.2023.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 08/13/2023]
Abstract
Adopting preventive strategies in individuals with subclinical Alzheimer's disease (AD) has the potential to delay dementia onset and reduce healthcare costs. Thus, it is extremely important to identify inexpensive, scalable, sensitive, and specific markers to track disease progression. The electroencephalography spectral power ratio (SPR: the fast to slow spectral power ratio), a measure of the shift in power distribution from higher to lower frequencies, holds potential for aiding clinical practice. The SPR is altered in patients with AD, correlates with cognitive functions, and can be easily implemented in clinical settings. However, whether the SPR is sensitive to pathophysiological changes in the prodromal stage of AD is unclear. We explored the SPR of individuals diagnosed with amyloid-positive amnestic mild cognitive impairment (Aβ+aMCI) and its association with both cognitive function and amyloid load. The SPR was lower in Aβ+aMCI than in the cognitively unimpaired individuals and correlated with executive function scores but not with amyloid load. Hypothesis-generating analyses suggested that aMCI participants with a lower SPR had an increased probability of a positive amyloid positron emission tomography. Future research may explore the potential of this measure to classify aMCI individuals according to their AD biomarker status.
Collapse
Affiliation(s)
- Aimee A Flores-Sandoval
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Charité - Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Paula Davila-Pérez
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Department of Clinical Neurophysiology, Hospital Universitario Rey Juan Carlos, Móstoles, Spain; Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Stephanie S Buss
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kevin Donohoe
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Margaret O'Connor
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Mouhsin M Shafi
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Hinda and Arthur Marcus Institute for Aging Research, and Deanna and Sidney Wolk Center for Memory Health, Hebrew Senior Life, Boston, MA, USA
| | - Christopher S Y Benwell
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Division of Psychology, School of Humanities, Social Sciences and Law, University of Dundee, Dundee, UK
| | - Peter J Fried
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Hendriksen HMA, van Gils AM, van Harten AC, Hartmann T, Mangialasche F, Kamondi A, Kivipelto M, Rhodius-Meester HFM, Smets EMA, van der Flier WM, Visser LNC. Communication about diagnosis, prognosis, and prevention in the memory clinic: perspectives of European memory clinic professionals. Alzheimers Res Ther 2023; 15:131. [PMID: 37543608 PMCID: PMC10404377 DOI: 10.1186/s13195-023-01276-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/19/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND The paradigm shift towards earlier Alzheimer's disease (AD) stages and personalized medicine creates new challenges for clinician-patient communication. We conducted a survey among European memory clinic professionals to identify opinions on communication about (etiological) diagnosis, prognosis, and prevention, and inventory needs for augmenting communication skills. METHODS Memory clinic professionals (N = 160) from 21 European countries completed our online survey (59% female, 14 ± 10 years' experience, 73% working in an academic hospital). We inventoried (1) opinions on communication about (etiological) diagnosis, prognosis, and prevention using 11 statements; (2) current communication practices in response to five hypothetical cases (AD dementia, mild cognitive impairment (MCI), subjective cognitive decline (SCD), with ( +) or without ( -) abnormal AD biomarkers); and (3) needs for communication support regarding ten listed communication skills. RESULTS The majority of professionals agreed that communication on diagnosis, prognosis, and prevention should be personalized to the individual patient. In response to the hypothetical patient cases, disease stage influenced the inclination to communicate an etiological AD diagnosis: 97% would explicitly mention the presence of AD to the patient with AD dementia, 68% would do so in MCI + , and 29% in SCD + . Furthermore, 58% would explicitly rule out AD in case of MCI - when talking to patients, and 69% in case of SCD - . Almost all professionals (79-99%) indicated discussing prognosis and prevention with all patients, of which a substantial part (48-86%) would personalize their communication to patients' diagnostic test results (39-68%) or patients' anamnestic information (33-82%). The majority of clinicians (79%) would like to use online tools, training, or both to support them in communicating with patients. Topics for which professionals desired support most were: stimulating patients' understanding of information, and communicating uncertainty, dementia risk, remotely/online, and with patients not (fluently) speaking the language of the country of residence. CONCLUSIONS In a survey of European memory clinic professionals, we found a strong positive attitude towards communication with patients about (etiological) diagnosis, prognosis, and prevention, and personalization of communication to characteristics and needs of individual patients. In addition, professionals expressed a need for supporting tools and skills training to further improve their communication with patients.
Collapse
Affiliation(s)
- Heleen M A Hendriksen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.
| | - Aniek M van Gils
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Tobias Hartmann
- Experimental Neurology, Saarland University, 66424, Homburg, Germany
- Deutsches Institut Für DemenzPrävention, Saarland University, 66424, Homburg, Germany
| | - Francesca Mangialasche
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Medical Unit Aging, Theme Inflammation and Aging, Stockholm, Sweden
| | - Anita Kamondi
- Department of Neurology, Neurology and Neurosurgery, National Institute of Mental Health, Budapest, Hungary
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Medical Unit Aging, Theme Inflammation and Aging, Stockholm, Sweden
- Ageing and Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Helsinki, Finland
| | - Hanneke F M Rhodius-Meester
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, Oslo, Norway
- Internal Medicine, Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Ellen M A Smets
- Medical Psychology, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Quality of Care, Personalized Medicine, , Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Leonie N C Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Medical Psychology, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Quality of Care, Personalized Medicine, , Amsterdam, The Netherlands
| |
Collapse
|
29
|
Valverde-Salazar V, Ruiz-Gabarre D, García-Escudero V. Alzheimer's Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:1460. [PMID: 37507998 PMCID: PMC10376369 DOI: 10.3390/antiox12071460] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress caused by an imbalance between free radicals and antioxidants is considered one of the main risk factors for AD, since it can result in protein, lipid and nucleic acid damage and exacerbate Aβ and tau pathology. To date, there is a lack of successful pharmacological approaches to cure or even ameliorate the terrible impact of this disease. Due to this, dietary compounds with antioxidative and anti-inflammatory properties acquire special relevance as potential therapeutic agents. In this context, green tea, and its main bioactive compound, epigallocatechin-3-gallate (EGCG), have been targeted as a plausible option for the modulation of AD. Specifically, EGCG acts as an antioxidant by regulating inflammatory processes involved in neurodegeneration such as ferroptosis and microglia-induced cytotoxicity and by inducing signalling pathways related to neuronal survival. Furthermore, it reduces tau hyperphosphorylation and aggregation and promotes the non-amyloidogenic route of APP processing, thus preventing the formation of Aβ and its subsequent accumulation. Taken together, these results suggest that EGCG may be a suitable candidate in the search for potential therapeutic compounds for neurodegenerative disorders involving inflammation and oxidative stress, including Alzheimer's disease.
Collapse
Affiliation(s)
- Víctor Valverde-Salazar
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Daniel Ruiz-Gabarre
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Vega García-Escudero
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, 28031 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
30
|
Høilund-Carlsen PF, Revheim ME, Costa T, Kepp KP, Castellani RJ, Perry G, Alavi A, Barrio JR. FDG-PET versus Amyloid-PET Imaging for Diagnosis and Response Evaluation in Alzheimer's Disease: Benefits and Pitfalls. Diagnostics (Basel) 2023; 13:2254. [PMID: 37443645 DOI: 10.3390/diagnostics13132254] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
In June 2021, the US Federal Drug and Food Administration (FDA) granted accelerated approval for the antibody aducanumab and, in January 2023, also for the antibody lecanemab, based on a perceived drug-induced removal of cerebral amyloid-beta as assessed by amyloid-PET and, in the case of lecanemab, also a presumption of limited clinical efficacy. Approval of the antibody donanemab is awaiting further data. However, published trial data indicate few, small and uncertain clinical benefits, below what is considered "clinically meaningful" and similar to the effect of conventional medication. Furthermore, a therapy-related decrease in the amyloid-PET signal may also reflect increased cell damage rather than simply "amyloid removal". This interpretation is more consistent with increased rates of amyloid-related imaging abnormalities and brain volume loss in treated patients, relative to placebo. We also challenge the current diagnostic criteria for AD based on amyloid-PET imaging biomarkers and recommend that future anti-AD therapy trials apply: (1) diagnosis of AD based on the co-occurrence of cognitive decline and decreased cerebral metabolism assessed by FDA-approved FDG-PET, (2) therapy efficacy determined by favorable effect on cognitive ability, cerebral metabolism by FDG-PET, and brain volumes by MRI, and (3) neuropathologic examination of all deaths occurring in these trials.
Collapse
Affiliation(s)
- Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Research Unit of Clinical Physiology and Nuclear Medicine, Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
| | - Mona-Elisabeth Revheim
- The Intervention Centre, Division of Technology and Innovation, Oslo University Hospital, 0372 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0313 Oslo, Norway
| | - Tommaso Costa
- GDS, Department of Psychology, Koelliker Hospital, University of Turin, 10124 Turin, Italy
- FOCUS Lab, Department of Psychology, University of Turin, 10124 Turin, Italy
| | - Kasper P Kepp
- Section of Biophysical and Biomedicinal Chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Rudolph J Castellani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Abass Alavi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
31
|
Yadollahikhales G, Rojas JC. Anti-Amyloid Immunotherapies for Alzheimer's Disease: A 2023 Clinical Update. Neurotherapeutics 2023; 20:914-931. [PMID: 37490245 PMCID: PMC10457266 DOI: 10.1007/s13311-023-01405-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
The amyloid cascade hypothesis is a useful framework for therapeutic development in Alzheimer's disease (AD). Amyloid b1-42 (Aβ) has been the main target of experimental therapies, based on evidence of the neurotoxic effects of Aβ, and of the potential adverse effects of brain Aβ burden detected in humans in vivo by positron emission tomography (PET). Progress on passive anti-amyloid immunotherapy research includes identification of antibodies that facilitate microglial activation, catalytical disaggregation, and increased flow of Aβ from cerebrospinal fluid (CSF) to plasma, thus decreasing the neurotoxic effects of Aβ. Recently completed phase 2 and 3 trials of 3rd generation anti-amyloid immunotherapies are supportive of their clinical efficacy in reducing brain Aβ burden and preventing cognitive decline. Data from recent trials implicate these agents as the first effective disease-modifying therapies against AD and has led to the US Food and Drug Administration (FDA) recent approval of aducanumab and lecanemab, under an accelerated approval pathway. The clinical effects of these agents are modest, however, and associated with amyloid-related imaging abnormalities (ARIA). Testing the effects of anti-Aβ immunotherapies in pre-symptomatic populations and identification of more potent and safer agents is the scope of ongoing and future research. Innovations in clinical trial design will be the key for the efficient and equitable development of novel anti-Aβ immunotherapies. The progress in the field of AD therapeutics will bring new clinical, logistical, and ethical challenges, which pose to revolutionize the practice of neurology, dementia care, and preventive cognitive healthcare.
Collapse
Affiliation(s)
- Golnaz Yadollahikhales
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, 1551 4th Street, 411G, San Francisco, CA, 94158, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, 1551 4th Street, 411G, San Francisco, CA, 94158, USA.
| |
Collapse
|
32
|
van der Flier WM, de Vugt ME, Smets EMA, Blom M, Teunissen CE. Towards a future where Alzheimer's disease pathology is stopped before the onset of dementia. NATURE AGING 2023; 3:494-505. [PMID: 37202515 DOI: 10.1038/s43587-023-00404-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) is a major healthcare challenge with no curative treatment at present. To address this challenge, we need a paradigm shift, where we focus on pre-dementia stages of AD. In this Perspective, we outline a strategy to move towards a future with personalized medicine for AD by preparing for and investing in effective and patient-orchestrated diagnosis, prediction and prevention of the dementia stage. While focusing on AD, this Perspective also discusses studies that do not specify the cause of dementia. Future personalized prevention strategies encompass multiple components, including tailored combinations of disease-modifying interventions and lifestyle. By empowering the public and patients to be more actively engaged in the management of their health and disease and by developing improved strategies for diagnosis, prediction and prevention, we can pave the way for a future with personalized medicine, in which AD pathology is stopped to prevent or delay the onset of dementia.
Collapse
Affiliation(s)
- Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands.
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
| | - Marjolein E de Vugt
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Ellen M A Smets
- Medical Psychology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| | - Marco Blom
- Alzheimer Nederland, Amersfoort, Utrecht, the Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Hakami ZH, Khamjan NA, Saad HM, Batiha GES, De Waard M. A Potential Link Between Visceral Obesity and Risk of Alzheimer's Disease. Neurochem Res 2023; 48:745-766. [PMID: 36409447 DOI: 10.1007/s11064-022-03817-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia characterized by the deposition of amyloid beta (Aβ) plaques and tau-neurofibrillary tangles in the brain. Visceral obesity (VO) is usually associated with low-grade inflammation due to higher expression of pro-inflammatory cytokines by adipose tissue. The objective of the present review was to evaluate the potential link between VO and the development of AD. Tissue hypoxia in obesity promotes tissue injury, production of adipocytokines, and release of pro-inflammatory cytokines leading to an oxidative-inflammatory loop with induction of insulin resistance. Importantly, brain insulin signaling is involved in the pathogenesis of AD and lower cognitive function. Obesity and enlargement of visceral adipose tissue are associated with the deposition of Aβ. All of this is consonant with VO increasing the risk of AD through the dysregulation of adipocytokines which affect the development of AD. The activated nuclear factor kappa B (NF-κB) pathway in VO might be a potential link in the development of AD. Likewise, the higher concentration of advanced glycation end-products in VO could be implicated in the pathogenesis of AD. Taken together, different inflammatory signaling pathways are activated in VO that all have a negative impact on the cognitive function and progression of AD except hypoxia-inducible factor 1 which has beneficial and neuroprotective effects in mitigating the progression of AD. In addition, VO-mediated hypoadiponectinemia and leptin resistance may promote the progression of Aβ formation and tau phosphorylation with the development of AD. In conclusion, VO-induced AD is mainly mediated through the induction of oxidative stress, inflammatory changes, leptin resistance, and hypoadiponectinemia that collectively trigger Aβ formation and neuroinflammation. Thus, early recognition of VO by visceral adiposity index with appropriate management could be a preventive measure against the development of AD in patients with VO.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120, Saint-Egrève, France.,L'institut du thorax, INSERM, CNRS, UNIV NANTES, 44007, Nantes, France.,LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
34
|
Zhao J, Huai J. Role of primary aging hallmarks in Alzheimer´s disease. Theranostics 2023; 13:197-230. [PMID: 36593969 PMCID: PMC9800733 DOI: 10.7150/thno.79535] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which severely threatens the health of the elderly and causes significant economic and social burdens. The causes of AD are complex and include heritable but mostly aging-related factors. The primary aging hallmarks include genomic instability, telomere wear, epigenetic changes, and loss of protein stability, which play a dominant role in the aging process. Although AD is closely associated with the aging process, the underlying mechanisms involved in AD pathogenesis have not been well characterized. This review summarizes the available literature about primary aging hallmarks and their roles in AD pathogenesis. By analyzing published literature, we attempted to uncover the possible mechanisms of aberrant epigenetic markers with related enzymes, transcription factors, and loss of proteostasis in AD. In particular, the importance of oxidative stress-induced DNA methylation and DNA methylation-directed histone modifications and proteostasis are highlighted. A molecular network of gene regulatory elements that undergoes a dynamic change with age may underlie age-dependent AD pathogenesis, and can be used as a new drug target to treat AD.
Collapse
|
35
|
Kwan ATH, Arfaie S, Therriault J, Azizi Z, Lussier FZ, Tissot C, Chamoun M, Bezgin G, Servaes S, Stevenon J, Rahmouni N, Pallen V, Gauthier S, Rosa-Neto P. Medial temporal tau predicts memory decline in cognitively unimpaired elderly. Brain Commun 2022; 5:fcac325. [PMID: 36627889 PMCID: PMC9814120 DOI: 10.1093/braincomms/fcac325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/05/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease can be detected in living people using in vivo biomarkers of amyloid-β and tau, even in the absence of cognitive impairment during the preclinical phase. [18F]-MK-6420 is a high-affinity PET tracer that quantifies tau neurofibrillary tangles, but its ability to predict cognitive changes associated with early Alzheimer's disease symptoms, such as memory decline, is unclear. Here, we assess the prognostic accuracy of baseline [18F]-MK-6420 tau-PET for predicting longitudinal memory decline in asymptomatic elderly individuals. In a longitudinal observational study, we evaluated a cohort of cognitively normal elderly participants (n = 111) from the translational biomarkers in ageing and dementia study (data collected between October 2017 and July 2020, with a follow-up period of 12 months). All participants underwent tau-PET with [18F]-MK-6420 and amyloid-β PET with [18F]-AZD-4694. The exclusion criteria included the presence of head trauma, stroke or other neurological disorders. There were 111 eligible participants selected based on the availability of amyloid-β PET, tau-PET, MRI and APOEɛ4 genotyping. Among these participants, the mean standard deviation age was 70.1 (8.6) years; 20 (18%) were tau-PET-positive and 71 of 111 (63.9%) were women. A significant association between the baseline Braak Stages I-II [18F]-MK-6240 standardized uptake value ratio positivity and change in composite memory score were observed at the 12-month follow-up, after correcting for age, sex and years of education [logical memory and Rey Auditory Verbal Learning Test, standardized beta = -0.52 (-0.82-0.21), P < 0.001, for dichotomized tau-PET and -1.22 (-1.84-(-0.61)], P < 0.0001, for continuous tau-PET]. Moderate cognitive decline was observed for A + T + over the follow-up period, whereas no significant change was observed for A-T+, A + T- and A-T-, although it should be noted that the A-T + group was small. Our results indicate that baseline tau neurofibrillary tangle pathology is associated with longitudinal changes in memory function, supporting the use of [18F]-MK-6420 PET to predict the likelihood of asymptomatic elderly individuals experiencing future memory decline. Overall, [18F]-MK-6420 PET is a promising tool for predicting memory decline in older adults without cognitive impairment at baseline. This is of critical relevance as the field is shifting towards a biological model of Alzheimer's disease defined by the aggregation of pathologic tau. Therefore, early detection of tau pathology using [18F]-MK-6420 PET provides us with hope that living patients with Alzheimer's disease may be diagnosed during the preclinical phase before it is too late.
Collapse
Affiliation(s)
- Angela T H Kwan
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
| | - Saman Arfaie
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC H3G 2M1, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Zahra Azizi
- Department of Medicine, McGill University Health Centre, Montreal, QC H3G 2M1, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Jenna Stevenon
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
- Department of Medicine, McGill University Health Centre, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
36
|
Statins Use in Alzheimer Disease: Bane or Boon from Frantic Search and Narrative Review. Brain Sci 2022; 12:brainsci12101290. [PMID: 36291224 PMCID: PMC9599431 DOI: 10.3390/brainsci12101290] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) was used to describe pre-senile dementia to differentiate it from senile dementia, which develops in the adult age group of more than 65 years. AD is characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. The neuropathological changes in AD are related to the deposition of amyloid plaques, neurofibrillary tangles, and progression of neuroinflammation, neuronal mitochondrial dysfunction, autophagy dysfunction, and cholinergic synaptic dysfunction. Statins are one of the main cornerstone drugs for the management of cardiovascular disorders regardless of dyslipidemia status. Increasing the use of statins, mainly in the elderly groups for primary and secondary prevention of cardiovascular diseases, may affect their cognitive functions. Extensive and prolonged use of statins may affect cognitive functions in healthy subjects and dementia patients. Statins-induced cognitive impairments in both patients and health providers had been reported according to the post-marketing survey. This survey depends mainly on sporadic cases, and no cognitive measures were used. Evidence from prospective and observational studies gives no robust conclusion regarding the beneficial or detrimental effects of statins on cognitive functions in AD patients. Therefore, this study is a narrative review aimed with evidences to the beneficial, detrimental, and neutral effects of statins on AD.
Collapse
|
37
|
Wang P, Yu L, Gong J, Xiong J, Zi S, Xie H, Zhang F, Mao Z, Liu Z, Kim JS. An Activity‐Based Fluorescent Probe for Imaging Fluctuations of Peroxynitrite (ONOO
−
) in the Alzheimer's Disease Brain. Angew Chem Int Ed Engl 2022; 61:e202206894. [DOI: 10.1002/anie.202206894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Pengzhan Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Le Yu
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Jiankang Gong
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Jianhua Xiong
- College of Chemistry and Molecular Sciences Wuhan University Wuhan Hubei 430072 China
| | - Soyu Zi
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Hua Xie
- School of Water Resources and Hydropower Wuhan University Wuhan Hubei 430072 China
| | - Fan Zhang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Zhiqiang Mao
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Zhihong Liu
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
- College of Chemistry and Molecular Sciences Wuhan University Wuhan Hubei 430072 China
| | - Jong Seung Kim
- Department of Chemistry Korea University Seoul 02841 Korea
| |
Collapse
|
38
|
Dulewicz M, Kulczyńska-Przybik A, Mroczko P, Kornhuber J, Lewczuk P, Mroczko B. Biomarkers for the Diagnosis of Alzheimer’s Disease in Clinical Practice: The Role of CSF Biomarkers during the Evolution of Diagnostic Criteria. Int J Mol Sci 2022; 23:ijms23158598. [PMID: 35955728 PMCID: PMC9369334 DOI: 10.3390/ijms23158598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive condition and the most common cause of dementia worldwide. The neuropathological changes characteristic of the disorder can be successfully detected before the development of full-blown AD. Early diagnosis of the disease constitutes a formidable challenge for clinicians. CSF biomarkers are the in vivo evidence of neuropathological changes developing in the brain of dementia patients. Therefore, measurement of their concentrations allows for improved accuracy of clinical diagnosis. Moreover, AD biomarkers may provide an indication of disease stage. Importantly, the CSF biomarkers of AD play a pivotal role in the new diagnostic criteria for the disease, and in the recent biological definition of AD by the National Institute on Aging, NIH and Alzheimer’s Association. Due to the necessity of collecting CSF by lumbar puncture, the procedure seems to be an important issue not only from a medical, but also a legal, viewpoint. Furthermore, recent technological advances may contribute to the automation of AD biomarkers measurement and may result in the establishment of unified cut-off values and reference limits. Moreover, a group of international experts in the field of AD biomarkers have developed a consensus and guidelines on the interpretation of CSF biomarkers in the context of AD diagnosis. Thus, technological advancement and expert recommendations may contribute to a more widespread use of these diagnostic tests in clinical practice to support a diagnosis of mild cognitive impairment (MCI) or dementia due to AD. This review article presents up-to-date data regarding the usefulness of CSF biomarkers in routine clinical practice and in biomarkers research.
Collapse
Affiliation(s)
- Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Correspondence:
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
| | - Piotr Mroczko
- Department of Criminal Law and Criminology, Faculty of Law, University of Bialystok, 15-213 Bialystok, Poland;
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Piotr Lewczuk
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
39
|
Wang P, Yu L, Gong J, Xiong J, Zi S, Xie H, Zhang F, Mao Z, Liu Z, Kim JS. An Activity‐Based Fluorescent Probe for Imaging Fluctuations of Peroxynitrite (ONOO‐) in the Alzheimer's Disease Brain. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202206894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Pengzhan Wang
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Hubei University, Wuhan College of chemistry and chemical engineering 430062 CHINA
| | - Le Yu
- Korea university, Seoul Chemistry KOREA, REPUBLIC OF
| | - Jiankang Gong
- Ministry of education key laboratory for the synthesis and application of organic functional molecules College of chemistry and chemical engineering 430062 CHINA
| | - Jianhua Xiong
- Wuhan university, Wuhan College of chemistry and molecular science CHINA
| | - Soyu Zi
- Korea university, Seoul Chemistry KOREA, REPUBLIC OF
| | - Hua Xie
- Wuhan University, Wuhan School of water resources and hydropower CHINA
| | - Fan Zhang
- Ministry of educational key laboratory for the synthesis and application of organic functional molecules, Hubei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Zhiqiang Mao
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Huibei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Zhihong Liu
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Huibei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Jong Seung Kim
- Korea University Department of Chemistry Anamdong 02841 Seoul KOREA, REPUBLIC OF
| |
Collapse
|
40
|
van der Schaar J, Visser LNC, Bouwman FH, Ket JCF, Scheltens P, Bredenoord AL, van der Flier WM. Considerations regarding a diagnosis of Alzheimer's disease before dementia: a systematic review. Alzheimers Res Ther 2022; 14:31. [PMID: 35144684 PMCID: PMC8829985 DOI: 10.1186/s13195-022-00971-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/31/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND The NIA-AA research framework proposes a purely biological definition of Alzheimer's disease (AD). This implies that AD can be diagnosed based on biomarker abnormalities, irrespective of clinical manifestation. While this brings opportunities, it also raises challenges. We aimed to provide an overview of considerations regarding the disclosure of AD pathology before the onset of dementia. METHODS A systematic literature review was conducted and reported according to PRISMA guidelines. We searched PubMed, Embase, APA PsycINFO, and Web of Science Core Collection (on 10 December 2020) for references on conveying AD biomarker results to individuals without dementia. Our query combined variations on the terms Alzheimer's disease, disclosure, or diagnosis, preclinical or prodromal, and biomarkers. Two reviewers independently screened the resulting 6860 titles and abstracts for eligibility and examined 162 full-text records for relevance. We included theoretical articles in English, on communicating amyloid and/or tau results to individuals with mild cognitive impairment, subjective cognitive decline, or normal cognition. MAXQDA-software was used for inductive data analysis. RESULTS We included 27 publications. From these, we extracted 26 unique considerations, which we grouped according to their primary relevance to a clinical, personal, or societal context. Clinical considerations included (lack of) validity, utility, and disclosure protocols. Personal considerations covered psychological and behavioral implications, as well as the right to (not) know. Finally, societal considerations comprised the risk of misconception, stigmatization, and discrimination. Overall, views were heterogeneous and often contradictory, with emphasis on harmful effects. CONCLUSIONS We found 26 diverse and opposing considerations, related to a clinical, personal, or societal context, which are relevant to diagnosing AD before dementia. The theoretical literature tended to focus on adverse impact and rely on common morality, while the motivation for and implications of biomarker testing are deeply personal. Our findings provide a starting point for clinicians to discuss biomarker-based diagnosis with their patients, which will become even more relevant in light of the conditional approval of a first disease-modifying drug for AD.
Collapse
Affiliation(s)
- Jetske van der Schaar
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.
| | - Leonie N C Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Femke H Bouwman
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands
| | - Annelien L Bredenoord
- Erasmus School of Philosophy, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands
- Department of Epidemiology & Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Barisano G, Montagne A, Kisler K, Schneider JA, Wardlaw JM, Zlokovic BV. Blood-brain barrier link to human cognitive impairment and Alzheimer's Disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:108-115. [PMID: 35450117 PMCID: PMC9017393 DOI: 10.1038/s44161-021-00014-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/21/2021] [Indexed: 01/18/2023]
Abstract
Vascular dysfunction is frequently seen in disorders associated with cognitive impairment, dementia and Alzheimer's disease (AD). Recent advances in neuroimaging and fluid biomarkers suggest that vascular dysfunction is not an innocent bystander only accompanying neuronal dysfunction. Loss of cerebrovascular integrity, often referred to as breakdown in the blood-brain barrier (BBB), has recently shown to be an early biomarker of human cognitive dysfunction and possibly underlying mechanism of age-related cognitive decline. Damage to the BBB may initiate or further invoke a range of tissue injuries causing synaptic and neuronal dysfunction and cognitive impairment that may contribute to AD. Therefore, better understanding of how vascular dysfunction caused by BBB breakdown interacts with amyloid-β and tau AD biomarkers to confer cognitive impairment may lead to new ways of thinking about pathogenesis, and possibly treatment and prevention of early cognitive impairment, dementia and AD, for which we still do not have effective therapies.
Collapse
Affiliation(s)
- Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- These authors contributed equally: Giuseppe Barisano and Axel Montagne
| | - Axel Montagne
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- These authors contributed equally: Giuseppe Barisano and Axel Montagne
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julie A. Schneider
- Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
42
|
O'Connor A, Karikari TK, Poole T, Ashton NJ, Lantero Rodriguez J, Khatun A, Swift I, Heslegrave AJ, Abel E, Chung E, Weston PSJ, Pavisic IM, Ryan NS, Barker S, Rossor MN, Polke JM, Frost C, Mead S, Blennow K, Zetterberg H, Fox NC. Plasma phospho-tau181 in presymptomatic and symptomatic familial Alzheimer's disease: a longitudinal cohort study. Mol Psychiatry 2021; 26:5967-5976. [PMID: 32665603 PMCID: PMC7612227 DOI: 10.1038/s41380-020-0838-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 01/05/2023]
Abstract
Blood biomarkers have great potential to advance clinical care and accelerate trials in Alzheimer's disease (AD). Plasma phospho-tau181 (p-tau181) is a promising blood biomarker however, it is unknown if levels increase in presymptomatic AD. Therefore, we investigated the timing of p-tau181 changes using 153 blood samples from 70 individuals in a longitudinal study of familial AD (FAD). Plasma p-tau181 was measured, using an in-house single molecule array assay. We compared p-tau181 between symptomatic carriers, presymptomatic carriers, and non-carriers, adjusting for age and sex. We examined the relationship between p-tau181 and neurofilament light and estimated years to/from symptom onset (EYO), as well as years to/from actual onset in a symptomatic subgroup. In addition, we studied associations between p-tau181 and clinical severity, as well testing for differences between genetic subgroups. Twenty-four were presymptomatic carriers (mean baseline EYO -9.6 years) while 27 were non-carriers. Compared with non-carriers, plasma p-tau181 concentration was higher in both symptomatic (p < 0.001) and presymptomatic mutation carriers (p < 0.001). Plasma p-tau181 showed considerable intra-individual variability but individual values discriminated symptomatic (AUC 0.93 [95% CI 0.85-0.98]) and presymptomatic (EYO ≥ -7 years) (AUC 0.86 [95% CI 0.72-0.94]) carriers from non-carriers of the same age and sex. From a fitted model there was evidence (p = 0.050) that p-tau181 concentrations were higher in mutation carriers than non-carriers from 16 years prior to estimated symptom onset. Our finding that plasma p-tau181 concentration is increased in symptomatic and presymptomatic FAD suggests potential utility as an easily accessible biomarker of AD pathology.
Collapse
Affiliation(s)
- Antoinette O'Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Teresa Poole
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia, South London & Maudsley NHS Foundation, London, UK
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ayesha Khatun
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Imogen Swift
- UK Dementia Research Institute at UCL, London, UK
| | | | - Emily Abel
- UK Dementia Research Institute at UCL, London, UK
| | - Elisha Chung
- UK Dementia Research Institute at UCL, London, UK
| | - Philip S J Weston
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Ivanna M Pavisic
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Suzie Barker
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Martin N Rossor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - James M Polke
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon Mead
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK.
- UK Dementia Research Institute at UCL, London, UK.
| |
Collapse
|
43
|
Frank L, Jennings LA, Petersen RC, Majid T, Gilmore-Bykovskyi A, Schicker L, Karlawish J. Participation of persons with dementia and their caregivers in research. J Am Geriatr Soc 2021; 69:1784-1792. [PMID: 34245586 DOI: 10.1111/jgs.17340] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND The National Institute on Aging, in conjunction with the Department of Health and Human Services as part of the National Alzheimer's Project Act, convened a 2020 National Research Summit on Care, Services, and Supports for Persons with Dementia and their Caregivers. This review article addresses research participation by persons living with dementia (PLWD) and their care partners in two different ways: as research participants with input on outcomes studied and as engaged research partners. RESULTS This article summarizes each of the topics presented at this Summit session, followed by reflection from the session panelists. Lee Jennings examined collection of outcomes directly from PLWD and the potential for individualized outcomes to enhance measurement in intervention trials. Ron Petersen discussed the impact of nomenclature on research and clinical care, and how and why investigators should be mindful of the connection between dementia nomenclature and the conduct of dementia research. Tabassum Majid examined strategies for engagement in research, including specific examples of involving PLWD and their care partners (including staff in assisted living and skilled nursing facilities), and the potential for this research engagement to improve our understanding of interventions in dementia. CONCLUSIONS Research participation by PLWD and their care partners is evolving. This review summarizes three areas of opportunity and steps for researchers to work with PLWD and their care partners to design and conduct research that enhances knowledge based on what we learn from PLWD and their care partners, and creates knowledge with them.
Collapse
Affiliation(s)
- Lori Frank
- RAND Corporation, Santa Monica, California, USA
| | - Lee A Jennings
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | | | | | | | - Jason Karlawish
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
44
|
Tian Hui Kwan A, Arfaie S, Therriault J, Rosa-Neto P, Gauthier S. Lessons Learnt from the Second Generation of Anti-Amyloid Monoclonal Antibodies Clinical Trials. Dement Geriatr Cogn Disord 2021; 49:334-348. [PMID: 33321511 DOI: 10.1159/000511506] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 09/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a chronic neurodegenerative disorder with complex pathophysiology that affects over 50 million people worldwide. Most drug therapies, to date, have focused on targeting the amyloid-beta (Aβ) pathway, but clinical outcomes of anti-Aβ antibodies have been unsuccessful and unable to meet their primary endpoints. Similar trends have also been observed in treatments that target the tau pathway. SUMMARY This paper reviews recent anti-Aβ passive monotherapies, since Bapineuzumab, that have progressed to phase 3 clinical trials. Specifically, we discuss the 4 clinical trial programs of Solanezumab (targets Aβ monomers), Aducanumab (targets Aβ oligomers and plaques), Crenezumab (targets Aβ oligomers), and Gantenerumab (targets Aβ fibrils) which are all exogenous monoclonal antibodies. We conclude with potential reasons for why they have not met their primary endpoints and discuss lessons learnt from these trials. Key Message: Future disease-modifying trials (DMTs) for AD should be conducted in asymptomatic, Aβ-positive individuals. Moreover, potential additive and/or synergistic benefits focusing on anti-Aβ and anti-tau drug combinations merit further investigation.
Collapse
Affiliation(s)
- Angela Tian Hui Kwan
- Department of Chemical & Physical Sciences, University of Toronto, Toronto, Ontario, Canada.,Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | - Saman Arfaie
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada.,Faculty of Medicine, McGill University, Montreal, Québec, Canada.,Department of Molecular Cell Biology, University of California, Berkeley, California, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada.,Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada.,Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada, .,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada, .,Department of Psychiatry, McGill University, Montreal, Québec, Canada,
| |
Collapse
|
45
|
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer's disease. Lancet 2021; 397:1577-1590. [PMID: 33667416 PMCID: PMC8354300 DOI: 10.1016/s0140-6736(20)32205-4] [Citation(s) in RCA: 2356] [Impact Index Per Article: 589.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/21/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
In this Seminar, we highlight the main developments in the field of Alzheimer's disease. The most recent data indicate that, by 2050, the prevalence of dementia will double in Europe and triple worldwide, and that estimate is 3 times higher when based on a biological (rather than clinical) definition of Alzheimer's disease. The earliest phase of Alzheimer's disease (cellular phase) happens in parallel with accumulating amyloid β, inducing the spread of tau pathology. The risk of Alzheimer's disease is 60-80% dependent on heritable factors, with more than 40 Alzheimer's disease-associated genetic risk loci already identified, of which the APOE alleles have the strongest association with the disease. Novel biomarkers include PET scans and plasma assays for amyloid β and phosphorylated tau, which show great promise for clinical and research use. Multidomain lifestyle-based prevention trials suggest cognitive benefits in participants with increased risk of dementia. Lifestyle factors do not directly affect Alzheimer's disease pathology, but can still contribute to a positive outcome in individuals with Alzheimer's disease. Promising pharmacological treatments are poised at advanced stages of clinical trials and include anti-amyloid β, anti-tau, and anti-inflammatory strategies.
Collapse
Affiliation(s)
- Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Life Science Partners, Amsterdam, Netherlands.
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurology, Leuven, Belgium; Dementia Research Institute, University College London, London, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Ageing and Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Henne Holstege
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gael Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Groupement d'Intérêt Public Cyceron, Caen, France
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, University of Nevada, Las Vegas, NV, USA; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Wiesje M van der Flier
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Epidemiology and Datascience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
46
|
Therriault J, Pascoal TA, Benedet AL, Tissot C, Savard M, Chamoun M, Lussier F, Kang MS, Berzgin G, Wang T, Fernandes-Arias J, Massarweh G, Soucy JP, Vitali P, Saha-Chaudhuri P, Gauthier S, Rosa-Neto P. Frequency of Biologically Defined Alzheimer Disease in Relation to Age, Sex, APOE ε4, and Cognitive Impairment. Neurology 2021; 96:e975-e985. [PMID: 33443136 PMCID: PMC8055338 DOI: 10.1212/wnl.0000000000011416] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To assess the frequency of biologically defined Alzheimer disease (AD) in relation to age, sex, APOE ε4, and clinical diagnosis in a prospective cohort study evaluated with amyloid-PET and tau-PET. METHODS We assessed cognitively unimpaired (CU) elderly (n = 166), patients with amnestic mild cognitive impairment (n = 77), and patients with probable AD dementia (n = 62) who underwent evaluation by dementia specialists and neuropsychologists in addition to amyloid-PET with [18F]AZD4694 and tau-PET with [18F]MK6240. Individuals were grouped according to their AD biomarker profile. Positive predictive value for biologically defined AD was assessed in relation to clinical diagnosis. Frequency of AD biomarker profiles was assessed using logistic regressions with odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The clinical diagnosis of probable AD dementia demonstrated good agreement with biologically defined AD (positive predictive value 85.2%). A total of 7.88% of CU were positive for both amyloid-PET and tau-PET. Frequency of biologically defined AD increased with age (OR 1.14; p < 0.0001) and frequency of APOE ε4 allele carriers (single ε4: OR 3.82; p < 0.0001; double ε4: OR 17.55, p < 0.0001). CONCLUSION Whereas we observed strong, but not complete, agreement between clinically defined probable AD dementia and biomarker positivity for both β-amyloid and tau, we also observed that biologically defined AD was not rare in CU elderly. Abnormal tau-PET was almost exclusively observed in individuals with abnormal amyloid-PET. Our results highlight that even in tertiary care memory clinics, detailed evaluation by dementia specialists systematically underestimates the frequency of biologically defined AD and related entities. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that biologically defined AD (abnormal amyloid PET and tau PET) was observed in 85.2% of people with clinically defined AD and 7.88% of CU elderly.
Collapse
Affiliation(s)
- Joseph Therriault
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Tharick A Pascoal
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Andrea L Benedet
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Cecile Tissot
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Melissa Savard
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Mira Chamoun
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Firoza Lussier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Min Su Kang
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Gleb Berzgin
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Tina Wang
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Jaime Fernandes-Arias
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Gassan Massarweh
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Jean-Paul Soucy
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Paolo Vitali
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Paramita Saha-Chaudhuri
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Serge Gauthier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Pedro Rosa-Neto
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada.
| |
Collapse
|
47
|
Petretto DR, Carrogu GP, Gaviano L, Pili L, Pili R. Dementia and Major Neurocognitive Disorders: Some Lessons Learned One Century after the first Alois Alzheimer's Clinical Notes. Geriatrics (Basel) 2021; 6:5. [PMID: 33440669 PMCID: PMC7838901 DOI: 10.3390/geriatrics6010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 11/30/2022] Open
Abstract
Over 100 years ago, Alois Alzheimer presented the clinical signs and symptoms of what has been later called "Alzheimer Dementia" in a young woman whose name was Augustine Deter [...].
Collapse
Affiliation(s)
- Donatella Rita Petretto
- Department of Education, Psychology and Philosophy, University of Cagliari, Via Is Mirrionis 1, 09127 Cagliari, Italy; (G.P.C.); (L.G.); (L.P.)
| | - Gian Pietro Carrogu
- Department of Education, Psychology and Philosophy, University of Cagliari, Via Is Mirrionis 1, 09127 Cagliari, Italy; (G.P.C.); (L.G.); (L.P.)
- Global Community on Longevity, Comunità Mondiale della Longevità, Selargius 09047, Italy; IERFOP Onlus, Cagliari, 09134
| | - Luca Gaviano
- Department of Education, Psychology and Philosophy, University of Cagliari, Via Is Mirrionis 1, 09127 Cagliari, Italy; (G.P.C.); (L.G.); (L.P.)
- Global Community on Longevity, Comunità Mondiale della Longevità, Selargius 09047, Italy; IERFOP Onlus, Cagliari, 09134
| | - Lorenzo Pili
- Department of Education, Psychology and Philosophy, University of Cagliari, Via Is Mirrionis 1, 09127 Cagliari, Italy; (G.P.C.); (L.G.); (L.P.)
- Global Community on Longevity, Comunità Mondiale della Longevità, Selargius 09047, Italy; IERFOP Onlus, Cagliari, 09134
| | - Roberto Pili
- Global Community on Longevity, Comunità Mondiale della Longevità, Selargius 09047, Italy; IERFOP Onlus, Cagliari, 09134
| |
Collapse
|
48
|
Seibel R, Schneider RH, Gottlieb MGV. Effects of Spices (Saffron, Rosemary, Cinnamon, Turmeric and Ginger) in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:347-357. [PMID: 34279199 DOI: 10.2174/1567205018666210716122034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 01/14/2021] [Accepted: 03/17/2021] [Indexed: 12/06/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent dementia in the elderly, causing disability, physical, psychological, social, and economic damage to the individual, their families, and caregivers. Studies have shown some spices, such as saffron, rosemary, cinnamon, turmeric, and ginger, have antioxidant and anti-inflammatory properties that act in inhibiting the aggregation of acetylcholinesterase and amyloid in AD. For this reason, spices have been studied as beneficial sources against neurodegenerative diseases, including AD. In this sense, this study aims to present a review of some spices (Saffron, Rosemary, Cinnamon, Turmeric and Ginger) and their bioactive compounds, most consumed and investigated in the world regarding AD. In this article, scientific evidence is compiled in clinical trials in adults, the elderly, animals, and in vitro, on properties considered neuroprotective, having no or negative effects on neuroprotection of these spices and their bioactive compounds. The importance of this issue is based on the pharmacological treatment for AD that is still not very effective. In addition, the recommendations and prescriptions of these spices are still permeated by questioning and lack of robust evidence of their effects on neurodegeneration. The literature search suggests all spices included in this article have bioactive compounds with anti-inflammatory and antioxidant actions associated with neuroprotection. To date, the amounts of spice ingestion in humans are not uniform, and there is no consensus on its indication and chronic consumption guarantees safety and efficacy in neuroprotection. Therefore, clinical evidence on this topic is necessary to become a formal adjuvant treatment for AD.
Collapse
Affiliation(s)
- Raquel Seibel
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| | - Rodolfo H Schneider
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| | - Maria G V Gottlieb
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre/RS, Brazil
| |
Collapse
|
49
|
Ribeiro VT, de Souza LC, Simões E Silva AC. Renin-Angiotensin System and Alzheimer's Disease Pathophysiology: From the Potential Interactions to Therapeutic Perspectives. Protein Pept Lett 2020; 27:484-511. [PMID: 31886744 DOI: 10.2174/0929866527666191230103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022]
Abstract
New roles of the Renin-Angiotensin System (RAS), apart from fluid homeostasis and Blood Pressure (BP) regulation, are being progressively unveiled, since the discoveries of RAS alternative axes and local RAS in different tissues, including the brain. Brain RAS is reported to interact with pathophysiological mechanisms of many neurological and psychiatric diseases, including Alzheimer's Disease (AD). Even though AD is the most common cause of dementia worldwide, its pathophysiology is far from elucidated. Currently, no treatment can halt the disease course. Successive failures of amyloid-targeting drugs have challenged the amyloid hypothesis and increased the interest in the inflammatory and vascular aspects of AD. RAS compounds, both centrally and peripherally, potentially interact with neuroinflammation and cerebrovascular regulation. This narrative review discusses the AD pathophysiology and its possible interaction with RAS, looking forward to potential therapeutic approaches. RAS molecules affect BP, cerebral blood flow, neuroinflammation, and oxidative stress. Angiotensin (Ang) II, via angiotensin type 1 receptors may promote brain tissue damage, while Ang-(1-7) seems to elicit neuroprotection. Several studies dosed RAS molecules in AD patients' biological material, with heterogeneous results. The link between AD and clinical conditions related to classical RAS axis overactivation (hypertension, heart failure, and chronic kidney disease) supports the hypothesized role of this system in AD. Additionally, RAStargeting drugs as Angiotensin Converting Enzyme inhibitors (ACEis) and Angiotensin Receptor Blockers (ARBs) seem to exert beneficial effects on AD. Results of randomized controlled trials testing ACEi or ARBs in AD are awaited to elucidate whether AD-RAS interaction has implications on AD therapeutics.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Internal Medicine, Service of Neurology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
50
|
Therriault J, Pascoal TA, Savard M, Benedet AL, Chamoun M, Tissot C, Lussier F, Kang MS, Thomas E, Terada T, Rej S, Massarweh G, Nasreddine Z, Vitali P, Soucy JP, Saha-Chaudhuri P, Gauthier S, Rosa-Neto P. Topographic Distribution of Amyloid-β, Tau, and Atrophy in Patients With Behavioral/Dysexecutive Alzheimer Disease. Neurology 2020; 96:e81-e92. [PMID: 33093220 PMCID: PMC7884976 DOI: 10.1212/wnl.0000000000011081] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022] Open
Abstract
Objective To determine the associations between amyloid-PET, tau-PET, and atrophy with the behavioral/dysexecutive presentation of Alzheimer disease (AD), how these differ from amnestic AD, and how they correlate to clinical symptoms. Methods We assessed 15 patients with behavioral/dysexecutive AD recruited from a tertiary care memory clinic, all of whom had biologically defined AD. They were compared with 25 patients with disease severity– and age-matched amnestic AD and a group of 131 cognitively unimpaired (CU) elderly individuals. All participants were evaluated with amyloid-PET with [18F]AZD4694, tau-PET with [18F]MK6240, MRI, and neuropsychological testing. Results Voxelwise contrasts identified patterns of frontal cortical tau aggregation in behavioral/dysexecutive AD, with peaks in medial prefrontal, anterior cingulate, and frontal insular cortices in contrast to amnestic AD. No differences were observed in the distribution of amyloid-PET or atrophy as determined by voxel-based morphometry. Voxelwise area under the receiver operating characteristic curve analyses revealed that tau-PET uptake in the medial prefrontal, anterior cingulate, and frontal insular cortices were best able to differentiate between behavioral/dysexecutive and amnestic AD (area under the curve 0.87). Voxelwise regressions demonstrated relationships between frontal cortical tau load and degree of executive dysfunction. Conclusions Our results provide evidence of frontal cortical involvement of tau pathology in behavioral/dysexecutive AD and highlight the need for consensus clinical criteria in this syndrome.
Collapse
Affiliation(s)
- Joseph Therriault
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Tharick A Pascoal
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Melissa Savard
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Andrea L Benedet
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Mira Chamoun
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Cecile Tissot
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Firoza Lussier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Min Su Kang
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Emilie Thomas
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Tatsuhiro Terada
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Soham Rej
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Gassan Massarweh
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Ziad Nasreddine
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Paolo Vitali
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Jean-Paul Soucy
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Paramita Saha-Chaudhuri
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Serge Gauthier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada
| | - Pedro Rosa-Neto
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., M.S., A.L.B., M.C., C.T., F.L., M.S.K., E.T., T.T., P.V., S.G., P.R.-N.), and Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., E.T., P.V., J.-P.S., S.G., P.R.-N.), Psychiatry (S.R., S.G.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University; Montreal Neurological Institute (J.T., T.A.P., A.L.B., M.C., C.T., F.L., M.S.K., G.M., J.-P.S., P.R.-N.), Canada; Department of Biofunctional Imaging (T.T.), Hamamatsu University School of Medicine, Japan; and MoCA Clinic and Institute (Z.N.), Montreal, Canada.
| |
Collapse
|