1
|
Maure-Blesa L, Rodríguez-Baz I, Carmona-Iragui M, Fortea J. What Can We Learn About Alzheimer's Disease from People with Down Syndrome? Curr Top Behav Neurosci 2024. [PMID: 39509049 DOI: 10.1007/7854_2024_546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Down syndrome (DS) is the most frequent cause of intellectual disability of genetic origin, estimated to affect about 1 in 700 babies born worldwide (CDC 2023). In Europe and the United States, current estimates indicate a population prevalence of 5.6 and 6.7 per 10,000 individuals, respectively, which translates to more than 200,000 people in the United States, more than 400,000 people in Europe, and approximately six million worldwide. Advances in healthcare and the treatment of accompanying conditions have significantly prolonged the lifespan of those with DS over the past 50 years. Consequently, there is a pressing need to address the challenges associated with ageing among this population, with Alzheimer's disease (AD) being the primary concern. In this chapter, we will review the significance of studying this population to understand AD biology, the insights gained on AD in DS (DSAD), and how this knowledge can help us understand the AD not only in DS but also in the general population. We will conclude by exploring the objectives that remain to be accomplished.
Collapse
Affiliation(s)
- Lucia Maure-Blesa
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Iñigo Rodríguez-Baz
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain.
| |
Collapse
|
2
|
Russell JK, Conley AC, Wilson JE, Newhouse PA. Cholinergic System Structure and Function Changes in Individuals with Down Syndrome During the Development of Alzheimer's Disease. Curr Top Behav Neurosci 2024. [PMID: 39485646 DOI: 10.1007/7854_2024_523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Adults with Down syndrome represent the population with the highest risk of developing Alzheimer's disease worldwide. The cholinergic system is known to decline in Alzheimer's disease, with this decline responsible for many of the cognitive deficits that develop. The integrity of the cholinergic system across the lifespan in individuals with Down syndrome is not well characterized. Small fetal and infant post-mortem studies suggest an intact cholinergic projection system with a potential reduction in cholinergic receptors, while post-mortem studies in adults with Down syndrome reveal an age-related decrease in cholinergic integrity. Advances in magnetic resonance imaging (MRI) and positron emission tomography (PET) over the last 20 years have allowed for studies investigating the changes in cholinergic integrity across aging and during the development of Alzheimer's disease. One large cross-sectional study demonstrated reduced cholinergic basal forebrain volume measured by MRI associated with increasing Alzheimer's disease pathology. In a small cohort of adults with Down syndrome, we have recently reported that PET measures of cholinergic integrity negatively correlated with amyloid accumulation. New disease-modifying treatments for Alzheimer's disease and treatments under development for Alzheimer's disease in Down syndrome have the potential to preserve the cholinergic system, while treatments targeting the cholinergic system directly may be used in conjunction with disease-modifying therapies to improve cognitive function further. A greater understanding of cholinergic neuronal and receptor integrity across the lifespan in individuals with Down syndrome will provide insights as to when targeting the cholinergic system is an appropriate therapeutic option and, in the future, maybe a valuable screening tool to identify individuals that would most benefit from cholinergic interventions.
Collapse
Affiliation(s)
- Jason K Russell
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander C Conley
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jo Ellen Wilson
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, USA
| | - Paul A Newhouse
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, USA.
| |
Collapse
|
3
|
Morcillo‐Nieto AO, Zsadanyi SE, Arriola‐Infante JE, Carmona‐Iragui M, Montal V, Pegueroles J, Aranha MR, Vaqué‐Alcázar L, Padilla C, Benejam B, Videla L, Barroeta I, Fernandez S, Altuna M, Giménez S, González‐Ortiz S, Bargalló N, Ribas L, Arranz J, Torres S, Iulita MF, Belbin O, Camacho V, Alcolea D, Lleó A, Fortea J, Bejanin A. Characterization of white matter hyperintensities in Down syndrome. Alzheimers Dement 2024; 20:6527-6541. [PMID: 39087352 PMCID: PMC11497714 DOI: 10.1002/alz.14146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION In Down syndrome (DS), white matter hyperintensities (WMHs) are highly prevalent, yet their topography and association with sociodemographic data and Alzheimer's disease (AD) biomarkers remain largely unexplored. METHODS In 261 DS adults and 131 euploid controls, fluid-attenuated inversion recovery magnetic resonance imaging scans were segmented and WMHs were extracted in concentric white matter layers and lobar regions. We tested associations with AD clinical stages, sociodemographic data, cerebrospinal fluid (CSF) AD biomarkers, and gray matter (GM) volume. RESULTS In DS, total WMHs arose at age 43 and showed stronger associations with age than in controls. WMH volume increased along the AD continuum, particularly in periventricular regions, and frontal, parietal, and occipital lobes. Associations were found with CSF biomarkers and temporo-parietal GM volumes. DISCUSSION WMHs increase 10 years before AD symptom onset in DS and are closely linked with AD biomarkers and neurodegeneration. This suggests a direct connection to AD pathophysiology, independent of vascular risks. HIGHLIGHTS White matter hyperintensities (WMHs) increased 10 years before Alzheimer's disease symptom onset in Down syndrome (DS). WMHs were strongly associated in DS with the neurofilament light chain biomarker. WMHs were more associated in DS with gray matter volume in parieto-temporal areas.
Collapse
|
4
|
Zsadanyi SE, Morcillo-Nieto AO, Aranha MR, Aragón I, Arriola-Infante JE, Vaqué-Alcázar L, Montal V, Pegueroles J, Arranz J, Rodríguez-Baz Í, Blesa LM, Videla L, Barroeta I, Del Hoyo Soriano L, Benejam B, Fernández S, Hernandez AS, Bargallo N, González-Ortiz S, Giménez S, Alcolea D, Belbin O, Lleó A, Fortea J, Carmona-Iragui M, Bejanin A. Associations of Microbleeds and Their Topography With Imaging and CSF Biomarkers of Alzheimer Pathology in Individuals With Down Syndrome. Neurology 2024; 103:e209676. [PMID: 39074338 PMCID: PMC11286286 DOI: 10.1212/wnl.0000000000209676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/31/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral hemorrhages are an exclusion criterion and potential adverse effect of antiamyloid agents. It is, therefore, critical to characterize the natural history of cerebral microbleeds in populations genetically predisposed to Alzheimer disease (AD), such as Down syndrome (DS). We aimed to assess microbleed emergence in adults with DS across the AD spectrum, defining their topography and associations with clinical variables, cognitive outcomes, and fluid and neuroimaging biomarkers. METHODS This cross-sectional study included participants aged 18 years or older from the Down-Alzheimer Barcelona Neuroimaging Initiative and Sant Pau Initiative on Neurodegeneration with T1-weighted and susceptibility-weighted images. Participants underwent comprehensive assessments, including apolipoprotein E (APOE) genotyping; fluid and plasma determinations of beta-amyloid, tau, and neurofilament light; cognitive outcomes (Cambridge Cognitive Examination and modified Cued Recall Test); and vascular risk factors (hypertension, diabetes mellitus, and dyslipidemia). We manually segmented microbleeds and characterized their topography. Associations between microbleed severity and AD biomarkers were explored using between-group comparisons (none vs 1 vs 2+) and multivariate linear models. RESULTS We included 276 individuals with DS and 158 healthy euploid controls (mean age = 47.8 years, 50.92% female). Individuals with DS were more likely to have microbleeds than controls (20% vs 8.9%, p < 0.001), with more severe presentation (12% with 2+ vs 1.9%). Microbleeds increased with age (12% 20-30 years vs 60% > 60 years) and AD clinical stage (12.42% asymptomatic, 27.9% prodromal, 35.09% dementia) were more common in APOEε4 carriers (26% vs 18.3% noncarriers, p = 0.008), but not associated with vascular risk factors (p > 0.05). Microbleeds were predominantly posterior (cerebellum 33.66%; occipital 14.85%; temporal 21.29%) in participants with DS. Associations with microbleed severity were found for neuroimaging and fluid AD biomarkers, but only hippocampal volumes (standardized β = -0.18 [-0.31, -0.06], p < 0.005) and CSF p-tau-181 concentrations (β = 0.26 [0.12, 0.41], p < 0.005) survived regression controlling for age and disease stage, respectively. Microbleeds had limited effect on cognitive outcomes. DISCUSSION In participants with DS, microbleeds present with a posterior, lobar predominance, are associated with disease severity, but do not affect cognitive performance. These results suggest an interplay between AD pathology and vascular lesions, implicating microbleeds as a risk factor limiting the use of antiamyloid agents in this population.
Collapse
Affiliation(s)
- Sara E Zsadanyi
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alejandra O Morcillo-Nieto
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Mateus R Aranha
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Irina Aragón
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - José E Arriola-Infante
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Victor Montal
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Jordi Pegueroles
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Javier Arranz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Íñigo Rodríguez-Baz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lucia M Blesa
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laura Videla
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Isabel Barroeta
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laura Del Hoyo Soriano
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Bessy Benejam
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Susana Fernández
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Aida S Hernandez
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Nuria Bargallo
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Sofía González-Ortiz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Sandra Giménez
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Daniel Alcolea
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Olivia Belbin
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alberto Lleó
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Juan Fortea
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Carmona-Iragui
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alexandre Bejanin
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| |
Collapse
|
5
|
Kasri A, Camporesi E, Gkanatsiou E, Boluda S, Brinkmalm G, Stimmer L, Ge J, Hanrieder J, Villain N, Duyckaerts C, Vermeiren Y, Pape SE, Nicolas G, Laquerrière A, De Deyn PP, Wallon D, Blennow K, Strydom A, Zetterberg H, Potier MC. Amyloid-β peptide signature associated with cerebral amyloid angiopathy in familial Alzheimer's disease with APPdup and Down syndrome. Acta Neuropathol 2024; 148:8. [PMID: 39026031 PMCID: PMC11258176 DOI: 10.1007/s00401-024-02756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid plaques containing amyloid-β (Aβ) peptides, intraneuronal neurofibrillary tangles, extracellular neuropil threads, and dystrophic neurites surrounding plaques composed of hyperphosphorylated tau protein (pTau). Aβ can also deposit in blood vessel walls leading to cerebral amyloid angiopathy (CAA). While amyloid plaques in AD brains are constant, CAA varies among cases. The study focuses on differences observed between rare and poorly studied patient groups with APP duplications (APPdup) and Down syndrome (DS) reported to have higher frequencies of elevated CAA levels in comparison to sporadic AD (sAD), most of APP mutations, and controls. We compared Aβ and tau pathologies in postmortem brain tissues across cases and Aβ peptides using mass spectrometry (MS). We further characterized the spatial distribution of Aβ peptides with MS-brain imaging. While intraparenchymal Aβ deposits were numerous in sAD, DS with AD (DS-AD) and AD with APP mutations, these were less abundant in APPdup. On the contrary, Aβ deposits in the blood vessels were abundant in APPdup and DS-AD while only APPdup cases displayed high Aβ deposits in capillaries. Investigation of Aβ peptide profiles showed a specific increase in Aβx-37, Aβx-38 and Aβx-40 but not Aβx-42 in APPdup cases and to a lower extent in DS-AD cases. Interestingly, N-truncated Aβ2-x peptides were particularly increased in APPdup compared to all other groups. This result was confirmed by MS-imaging of leptomeningeal and parenchymal vessels from an APPdup case, suggesting that CAA is associated with accumulation of shorter Aβ peptides truncated both at N- and C-termini in blood vessels. Altogether, this study identified striking differences in the localization and composition of Aβ deposits between AD cases, particularly APPdup and DS-AD, both carrying three genomic copies of the APP gene. Detection of specific Aβ peptides in CSF or plasma of these patients could improve the diagnosis of CAA and their inclusion in anti-amyloid immunotherapy treatments.
Collapse
Affiliation(s)
- Amal Kasri
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Elena Camporesi
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eleni Gkanatsiou
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Susana Boluda
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lev Stimmer
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Junyue Ge
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörg Hanrieder
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Nicolas Villain
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Charles Duyckaerts
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
| | - Sarah E Pape
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Gaël Nicolas
- Department of Genetics, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Peter Paul De Deyn
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - David Wallon
- Department of Neurology, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, 76000, Rouen, France
| | - Kaj Blennow
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Andre Strydom
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
- UK Dementia Research Institute at UCL, London, UK.
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China.
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA.
| | - Marie-Claude Potier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France.
| |
Collapse
|
6
|
Aranha MR, Montal V, van den Brink H, Pegueroles J, Carmona‐Iragui M, Videla L, Maure Blesa L, Benejam B, Arranz J, Valldeneu S, Barroeta I, Fernández S, Ribas L, Alcolea D, González‐Ortiz S, Bargalló N, Biessels GJ, Blesa R, Lleó A, Coutinho AM, Leite CC, Bejanin A, Fortea J. Cortical microinfarcts in adults with Down syndrome assessed with 3T-MRI. Alzheimers Dement 2024; 20:3906-3917. [PMID: 38644660 PMCID: PMC11180852 DOI: 10.1002/alz.13797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/22/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Cortical microinfarcts (CMI) were attributed to cerebrovascular disease and cerebral amyloid angiopathy (CAA). CAA is frequent in Down syndrome (DS) while hypertension is rare, yet no studies have assessed CMI in DS. METHODS We included 195 adults with DS, 63 with symptomatic sporadic Alzheimer's disease (AD), and 106 controls with 3T magnetic resonance imaging. We assessed CMI prevalence in each group and CMI association with age, AD clinical continuum, vascular risk factors, vascular neuroimaging findings, amyloid/tau/neurodegeneration biomarkers, and cognition in DS. RESULTS CMI prevalence was 11.8% in DS, 4.7% in controls, and 17.5% in sporadic AD. In DS, CMI increased in prevalence with age and the AD clinical continuum, was clustered in the parietal lobes, and was associated with lacunes and cortico-subcortical infarcts, but not hemorrhagic lesions. DISCUSSION In DS, CMI are posteriorly distributed and related to ischemic but not hemorrhagic findings suggesting they might be associated with a specific ischemic CAA phenotype. HIGHLIGHTS This is the first study to assess cortical microinfarcts (assessed with 3T magnetic resonance imaging) in adults with Down syndrome (DS). We studied the prevalence of cortical microinfarcts in DS and its relationship with age, the Alzheimer's disease (AD) clinical continuum, vascular risk factors, vascular neuroimaging findings, amyloid/tau/neurodegeneration biomarkers, and cognition. The prevalence of cortical microinfarcts was 11.8% in DS and increased with age and along the AD clinical continuum. Cortical microinfarcts were clustered in the parietal lobes, and were associated with lacunes and cortico-subcortical infarcts, but not hemorrhagic lesions. In DS, cortical microinfarcts are posteriorly distributed and related to ischemic but not hemorrhagic findings suggesting they might be associated with a specific ischemic phenotype of cerebral amyloid angiopathy.
Collapse
|
7
|
Fortea J, McGlinchey E, Espinosa JM, Rafii MS. Addressing challenges in health care and research for people with Down syndrome. Lancet 2024; 403:1830-1833. [PMID: 38521088 DOI: 10.1016/s0140-6736(24)00478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Barcelona 08025, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eimear McGlinchey
- Trinity Centre for Ageing and Intellectual Disability, Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, Trinity College Dublin, University of California San Francisco, Dublin, Ireland
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome and Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego, CA, USA
| |
Collapse
|
8
|
Altuna M, Estanga A, Garrido A, Saldias J, Cañada M, Echeverria M, Larrea JÁ, Ayo P, Fiz A, Muñoz M, Santa-Inés J, García-Landarte V, García-Sebastián M. Down Syndrome-Basque Alzheimer Initiative (DS-BAI): Clinic-Biological Cohort. J Clin Med 2024; 13:1139. [PMID: 38398452 PMCID: PMC10889106 DOI: 10.3390/jcm13041139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Down syndrome (DS) is the most common genetically determined intellectual disability. In recent decades, it has experienced an exponential increase in life expectancy, leading to a rise in age-related diseases, including Alzheimer's disease (AD). Specific health plans for the comprehensive care of the DS community are an unmet need, which is crucial for the early and accurate diagnosis of main medical comorbidities. We present the protocol of a newly created clinical and research cohort and its feasibility in real life. METHODS The Down Syndrome-Basque Alzheimer Initiative (DS-BAI) is a population-based, inclusive, multidisciplinary initiative for the clinical-assistance and clinical-biological research approach to aging in DS led by the CITA-Alzheimer Foundation (Donostia, Basque Country). It aims to achieve the following: (1) provide comprehensive care for adults with DS, (2) optimize access to rigorous and quality training for socio-family and healthcare references, and (3) create a valuable multimodal clinical-biological research platform. RESULTS During the first year, 114 adults with DS joined the initiative, with 36% of them showing symptoms indicative of AD. Furthermore, adherence to training programs for healthcare professionals and families has been high, and the willingness to collaborate in basic and translational research has been encouraging. CONCLUSION Specific health plans for DS and conducting clinical and translational research on the challenges of aging, including AD, are necessary and feasible.
Collapse
Affiliation(s)
- Miren Altuna
- Fundación CITA-Alzheimer Fundazioa, 20009 Donostia, Spain
- Debabarrena Integrated Health Organization, Osakidetza Basque Health Service, 20690 Gipuzkoa, Spain
- Department of Medicine, Faculty of Health Sciences, University of Deusto, 48007 Bilbo, Spain
| | - Ainara Estanga
- Fundación CITA-Alzheimer Fundazioa, 20009 Donostia, Spain
| | - Adolfo Garrido
- Donostialdea Integrated Health Organisation, Clinical Biochemistry Department, Osakidetza Basque Health Service, 20014 Donostia, Spain
| | - Jon Saldias
- Fundación CITA-Alzheimer Fundazioa, 20009 Donostia, Spain
| | - Marta Cañada
- Fundación CITA-Alzheimer Fundazioa, 20009 Donostia, Spain
| | - Maitane Echeverria
- Donostialdea Integrated Health Organisation, Clinical Biochemistry Department, Osakidetza Basque Health Service, 20014 Donostia, Spain
| | - José Ángel Larrea
- Department of Medicine, Faculty of Health Sciences, University of Deusto, 48007 Bilbo, Spain
- Donostialdea Integrated Health Organisation, Radiology Department, Osakidetza Basque Health Service, 20014 Donostia, Spain
| | | | | | - María Muñoz
- Fundación Goyeneche de San Sebastián, 20018 Donostia, Spain
| | | | | | | |
Collapse
|
9
|
Enfermedad de Alzheimer y síndrome de Down. Semergen 2023; 49:101872. [DOI: 10.1016/j.semerg.2022.101872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/15/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022]
|
10
|
Levin J, Hasan A, Alejandre IA, Lorenzi I, Mall V, Rohrer TR. Diseases affecting middle-aged and elderly individuals with trisomy 21. DEUTSCHES ÄRZTEBLATT INTERNATIONAL 2023. [DOI: 10.3238/arztebl.m2022.03711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
11
|
de Souza MM, Cenci AR, Teixeira KF, Machado V, Mendes Schuler MCG, Gonçalves AE, Paula Dalmagro A, André Cazarin C, Gomes Ferreira LL, de Oliveira AS, Andricopulo AD. DYRK1A Inhibitors and Perspectives for the Treatment of Alzheimer's Disease. Curr Med Chem 2023; 30:669-688. [PMID: 35726411 DOI: 10.2174/0929867329666220620162018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/22/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most common form of dementia, especially in the elderly. Due to the increase in life expectancy, in recent years, there has been an excessive growth in the number of people affected by this disease, causing serious problems for health systems. In recent years, research has been intensified to find new therapeutic approaches that prevent the progression of the disease. In this sense, recent studies indicate that the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) gene, which is located on chromosome 21q22.2 and overexpressed in Down syndrome (DS), may play a significant role in developmental brain disorders and early onset neurodegeneration, neuronal loss and dementia in DS and AD. Inhibiting DYRK1A may serve to stop the phenotypic effects of its overexpression and, therefore, is a potential treatment strategy for the prevention of ageassociated neurodegeneration, including Alzheimer-type pathology. OBJECTIVE In this review, we investigate the contribution of DYRK1A inhibitors as potential anti-AD agents. METHODS A search in the literature to compile an in vitro dataset including IC50 values involving DYRK1A was performed from 2014 to the present day. In addition, we carried out structure-activity relationship studies based on in vitro and in silico data. RESULTS molecular modeling and enzyme kinetics studies indicate that DYRK1A may contribute to AD pathology through its proteolytic process, reducing its kinase specificity. CONCLUSION further evaluation of DYRK1A inhibitors may contribute to new therapeutic approaches for AD.
Collapse
Affiliation(s)
- Márcia Maria de Souza
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Arthur Ribeiro Cenci
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | - Kerolain Faoro Teixeira
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | - Valkiria Machado
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | | | - Ana Elisa Gonçalves
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Ana Paula Dalmagro
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Camila André Cazarin
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Leonardo Luiz Gomes Ferreira
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| | - Aldo Sena de Oliveira
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| | - Adriano Defini Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| |
Collapse
|
12
|
Altered spontaneous brain activity in Down syndrome and its relation with cognitive outcome. Sci Rep 2022; 12:15410. [PMID: 36104362 PMCID: PMC9474876 DOI: 10.1038/s41598-022-19627-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractAlthough Down syndrome (DS) is the most common genetic cause of neurodevelopmental delay, few neuroimaging studies have explored this population. This investigation aimed to study whole-brain resting-state spontaneous brain activity using fractional amplitude of low-frequency fluctuation (fALFF) and regional homogeneity (ReHo) strategies to find differences in spontaneous brain activity among young people with DS and controls and to correlate these results with cognitive outcomes. The sample comprised 18 persons with DS (age mean = 28.67, standard deviation = 4.18) and 18 controls (age mean = 28.56, standard deviation = 4.26). fALFF and ReHo analyses were performed, and the results were correlated with other cognitive variables also collected (KBIT-2 and verbal fluency test). Increased activity was found in DS using fALFF in areas involving the frontal and temporal lobes and left cerebellum anterior lobe. Decreased activity in DS was found in the left parietal and occipital lobe, the left limbic lobe and the left cerebellum posterior lobe. ReHo analysis showed increased activity in certain DS areas of the left frontal lobe and left rectus, as well as the inferior temporal lobe. The areas with decreased activity in the DS participants were regions of the frontal lobe and the right limbic lobe. Altered fALFF and ReHo were found in the DS population, and this alteration could predict the cognitive abilities of the participants. To our knowledge, this is the first study to explore regional spontaneous brain activity in a population with DS. Moreover, this study suggests the possibility of using fALFF and ReHo as biomarkers of cognitive function, which is highly important given the difficulties in cognitively evaluating this population to assess dementia. More research is needed, however, to demonstrate its utility.
Collapse
|
13
|
Moon JE, Lawrence JB. Chromosome silencing in vitro reveals trisomy 21 causes cell-autonomous deficits in angiogenesis and early dysregulation in Notch signaling. Cell Rep 2022; 40:111174. [PMID: 35947952 PMCID: PMC9505374 DOI: 10.1016/j.celrep.2022.111174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/24/2021] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Despite the prevalence of Down syndrome (DS), little is known regarding the specific cell pathologies that underlie this multi-system disorder. To understand which cell types and pathways are more directly affected by trisomy 21 (T21), we used an inducible-XIST system to silence one chromosome 21 in vitro. T21 caused the dysregulation of Notch signaling in iPSCs, potentially affecting cell-type programming. Further analyses identified dysregulation of pathways important for two cell types: neurogenesis and angiogenesis. Angiogenesis is essential to many bodily systems, yet is understudied in DS; therefore, we focused next on whether T21 affects endothelial cells. An in vitro assay for microvasculature formation revealed a cellular pathology involving delayed tube formation in response to angiogenic signals. Parallel transcriptomic analysis of endothelia further showed deficits in angiogenesis regulators. Results indicate a direct cell-autonomous impact of T21 on endothelial function, highlighting the importance of angiogenesis, with wide-reaching implications for development and disease progression. Moon and Lawrence examine the immediate effects of trisomy 21 silencing and find angiogenesis and neurogenesis pathways, including Notch signaling, affected as early as pluripotency. In endothelial cells, functional analyses show that trisomy delays the angiogenic response for microvessel formation and transcriptomics show a parallel impact on angiogenic regulators and signal-response and cytoskeleton processes.
Collapse
Affiliation(s)
- Jennifer E Moon
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeanne B Lawrence
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
14
|
Martini AC, Gross TJ, Head E, Mapstone M. Beyond amyloid: Immune, cerebrovascular, and metabolic contributions to Alzheimer disease in people with Down syndrome. Neuron 2022; 110:2063-2079. [PMID: 35472307 PMCID: PMC9262826 DOI: 10.1016/j.neuron.2022.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
People with Down syndrome (DS) have increased risk of Alzheimer disease (AD), presumably conferred through genetic predispositions arising from trisomy 21. These predispositions necessarily include triplication of the amyloid precursor protein (APP), but also other Ch21 genes that confer risk directly or through interactions with genes on other chromosomes. We discuss evidence that multiple genes on chromosome 21 are associated with metabolic dysfunction in DS. The resulting dysregulated pathways involve the immune system, leading to chronic inflammation; the cerebrovascular system, leading to disruption of the blood brain barrier (BBB); and cellular energy metabolism, promoting increased oxidative stress. In combination, these disruptions may produce a precarious biological milieu that, in the presence of accumulating amyloid, drives the pathophysiological cascade of AD in people with DS. Critically, mechanistic drivers of this dysfunction may be targetable in future clinical trials of pharmaceutical and/or lifestyle interventions.
Collapse
Affiliation(s)
- Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas J Gross
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
15
|
Aranha MR, Fortea J, Carmona-Iragui M. Cerebral Amyloid Angiopathy-Related Inflammation in Down Syndrome-Related Alzheimer Disease. Neurology 2022; 98:1021-1022. [PMID: 35470138 DOI: 10.1212/wnl.0000000000200704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Mateus Rozalem Aranha
- From the Memory Unit (M.R.A., J.F., M.C.-I.), Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau-Universitat Autònoma de Barcelona; Fundació Catalana Síndrome de Down (J.F., M.C.-I.), Barcelona Down Medical Center; and Center of Biomedical Investigation Network for Neurodegenerative Diseases (J.F., M.C.-I.), Madrid, Spain
| | - Juan Fortea
- From the Memory Unit (M.R.A., J.F., M.C.-I.), Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau-Universitat Autònoma de Barcelona; Fundació Catalana Síndrome de Down (J.F., M.C.-I.), Barcelona Down Medical Center; and Center of Biomedical Investigation Network for Neurodegenerative Diseases (J.F., M.C.-I.), Madrid, Spain
| | - Maria Carmona-Iragui
- From the Memory Unit (M.R.A., J.F., M.C.-I.), Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau-Universitat Autònoma de Barcelona; Fundació Catalana Síndrome de Down (J.F., M.C.-I.), Barcelona Down Medical Center; and Center of Biomedical Investigation Network for Neurodegenerative Diseases (J.F., M.C.-I.), Madrid, Spain.
| |
Collapse
|
16
|
Wegiel J, Flory M, Kuchna I, Nowicki K, Wegiel J, Ma SY, Zhong N, Bobrowicz TW, de Leon M, Lai F, Silverman WP, Wisniewski T. Developmental deficits and staging of dynamics of age associated Alzheimer's disease neurodegeneration and neuronal loss in subjects with Down syndrome. Acta Neuropathol Commun 2022; 10:2. [PMID: 34983655 PMCID: PMC8728914 DOI: 10.1186/s40478-021-01300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
The increased life expectancy of individuals with Down syndrome (DS) is associated with increased prevalence of trisomy 21-linked early-onset Alzheimer's disease (EOAD) and dementia. The aims of this study of 14 brain regions including the entorhinal cortex, hippocampus, basal ganglia, and cerebellum in 33 adults with DS 26-72 years of age were to identify the magnitude of brain region-specific developmental neuronal deficits contributing to intellectual deficits, to apply this baseline to identification of the topography and magnitude of neurodegeneration and neuronal and volume losses caused by EOAD, and to establish age-based staging of the pattern of genetically driven neuropathology in DS. Both DS subject age and stage of dementia, themselves very strongly correlated, were strong predictors of an AD-associated decrease of the number of neurons, considered a major contributor to dementia. The DS cohort was subclassified by age as pre-AD stage, with 26-41-year-old subjects with a full spectrum of developmental deficit but with very limited incipient AD pathology, and 43-49, 51-59, and 61-72-year-old groups with predominant prevalence of mild, moderately severe, and severe dementia respectively. This multiregional study revealed a 28.1% developmental neuronal deficit in DS subjects 26-41 years of age and 11.9% AD-associated neuronal loss in DS subjects 43-49 years of age; a 28.0% maximum neuronal loss at 51-59 years of age; and a 11.0% minimum neuronal loss at 61-72 years of age. A total developmental neuronal deficit of 40.8 million neurons and AD-associated neuronal loss of 41.6 million neurons reflect a comparable magnitude of developmental neuronal deficit contributing to intellectual deficits, and AD-associated neuronal loss contributing to dementia. This highly predictable pattern of pathology indicates that successful treatment of DS subjects in the fourth decade of life may prevent AD pathology and functional decline.
Collapse
Affiliation(s)
- Jerzy Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Michael Flory
- New York State Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY USA
| | - Izabela Kuchna
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Krzysztof Nowicki
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Jarek Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Shuang Yong Ma
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Nanbert Zhong
- New York State Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY USA
| | | | - Mony de Leon
- Department of Radiology, Weill Cornell Medicine, New York, NY USA
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Wayne P. Silverman
- Department of Pediatrics, Irvine Medical Center, University of California, Irvine, CA USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, NYU Grossman School of Medicine, New York, NY 10016 USA
| |
Collapse
|
17
|
Chan AWS, Cho IK, Li CX, Zhang X, Patel S, Rusnak R, Raper J, Bachevalier J, Moran SP, Chi T, Cannon KH, Hunter CE, Martin RC, Xiao H, Yang SH, Gumber S, Herndon JG, Rosen RF, Hu WT, Lah JJ, Levey AI, Smith Y, Walker LC. Cerebral Aβ deposition in an Aβ-precursor protein-transgenic rhesus monkey. AGING BRAIN 2022; 2:100044. [PMID: 36589695 PMCID: PMC9802652 DOI: 10.1016/j.nbas.2022.100044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
With the ultimate goal of developing a more representative animal model of Alzheimer's disease (AD), two female amyloid-β-(Aβ) precursor protein-transgenic (APPtg) rhesus monkeys were generated by lentiviral transduction of the APP gene into rhesus oocytes, followed by in vitro fertilization and embryo transfer. The APP-transgene included the AD-associated Swedish K670N/M671L and Indiana V717F mutations (APPSWE/IND) regulated by the human polyubiquitin-C promoter. Overexpression of APP was confirmed in lymphocytes and brain tissue. Upon sacrifice at 10 years of age, one of the monkeys had developed Aβ plaques and cerebral Aβ-amyloid angiopathy in the occipital, parietal, and caudal temporal neocortices. The induction of Aβ deposition more than a decade prior to its usual emergence in the rhesus monkey supports the feasibility of creating a transgenic nonhuman primate model for mechanistic analyses and preclinical testing of treatments for Alzheimer's disease and cerebrovascular amyloidosis.
Collapse
Affiliation(s)
- Anthony W S Chan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - In Ki Cho
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Chun-Xia Li
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Xiaodong Zhang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sudeep Patel
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca Rusnak
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jessica Raper
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jocelyne Bachevalier
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Psychology, Emory College, Atlanta, GA 30322, USA
| | - Sean P Moran
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Tim Chi
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Katherine H Cannon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Carissa E Hunter
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ryan C Martin
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shang-Hsun Yang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjeev Gumber
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - James G Herndon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca F Rosen
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lary C Walker
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Yamada K, Yoshimura J, Watanabe M, Suzuki K. Application of 7 tesla magnetic resonance imaging for pediatric neurological disorders: Early clinical experience. J Clin Imaging Sci 2021; 11:65. [PMID: 34992941 PMCID: PMC8720429 DOI: 10.25259/jcis_185_2021] [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: 09/29/2021] [Accepted: 11/07/2021] [Indexed: 11/04/2022] Open
Abstract
Ultra-high field magnetic resonance imaging (MRI) has been introduced for use in pediatric developmental neurology. While higher magnetic fields have certain advantages, optimized techniques with specific considerations are required to ensure rational and safe use in children and those with pediatric neurological disorders (PNDs). Here, we summarize our initial experience with clinical translational studies that utilized 7 tesla (T)-MRI in the fields of developmental neurology. T2-reversed images and three-dimensional anisotropy contrast imaging enabled the depiction of targeted pathological brain structures with better spatial resolution. Diffusion imaging and susceptibility-weighted imaging enabled visualization of intracortical, subcortical, and intratumoral microstructures in vivo within highly limited scan times appropriate for patients with PNDs. 7T-MRI appears to have significant potential to enhance the depiction of the structural and functional properties of the brain, particularly those associated with atypical brain development.
Collapse
Affiliation(s)
- Kenichi Yamada
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Junichi Yoshimura
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Watanabe
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kiyotaka Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
19
|
Fortea J, Zaman SH, Hartley S, Rafii MS, Head E, Carmona-Iragui M. Alzheimer's disease associated with Down syndrome: a genetic form of dementia. Lancet Neurol 2021; 20:930-942. [PMID: 34687637 PMCID: PMC9387748 DOI: 10.1016/s1474-4422(21)00245-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 02/03/2023]
Abstract
Adults with Down syndrome develop the neuropathological hallmarks of Alzheimer's disease and are at very high risk of developing early-onset dementia, which is now the leading cause of death in this population. Diagnosis of dementia remains a clinical challenge because of the lack of validated diagnostic criteria in this population, and because symptoms are overshadowed by the intellectual disability associated with Down syndrome. In people with Down syndrome, fluid and imaging biomarkers have shown good diagnostic performances and a strikingly similar temporality of changes with respect to sporadic and autosomal dominant Alzheimer's disease. Most importantly, there are no treatments to prevent Alzheimer's disease, even though adults with Down syndrome could be an optimal population in whom to conduct Alzheimer's disease prevention trials. Unprecedented research activity in Down syndrome is rapidly changing this bleak scenario that will translate into disease-modifying therapies that could benefit other populations.
Collapse
Affiliation(s)
- Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu y Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain.
| | - Shahid H Zaman
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK; Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, UK
| | - Sigan Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu y Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| |
Collapse
|
20
|
Rujeedawa T, Carrillo Félez E, Clare ICH, Fortea J, Strydom A, Rebillat AS, Coppus A, Levin J, Zaman SH. The Clinical and Neuropathological Features of Sporadic (Late-Onset) and Genetic Forms of Alzheimer's Disease. J Clin Med 2021; 10:4582. [PMID: 34640600 PMCID: PMC8509365 DOI: 10.3390/jcm10194582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to compare and highlight the clinical and pathological aspects of genetic versus acquired Alzheimer's disease: Down syndrome-associated Alzheimer's disease in (DSAD) and Autosomal Dominant Alzheimer's disease (ADAD) are compared with the late-onset form of the disease (LOAD). DSAD and ADAD present in a younger population and are more likely to manifest with non-amnestic (such as dysexecutive function features) in the prodromal phase or neurological features (such as seizures and paralysis) especially in ADAD. The very large variety of mutations associated with ADAD explains the wider range of phenotypes. In the LOAD, age-associated comorbidities explain many of the phenotypic differences.
Collapse
Affiliation(s)
- Tanzil Rujeedawa
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Eva Carrillo Félez
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Isabel C. H. Clare
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, 08029 Barcelona, Spain
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK;
- South London and the Maudsley NHS Foundation Trust, The LonDowns Consortium, London SE5 8AZ, UK
| | | | - Antonia Coppus
- Department for Primary and Community Care, Department of Primary and Community Care (149 ELG), Radboud University Nijmegen Medical Center, P.O. Box 9101, 6525 GA Nijmegen, The Netherlands;
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Shahid H. Zaman
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| |
Collapse
|
21
|
Marano M, Pompucci A, Motolese F, Rossi M, Coletta E, Di Lazzaro V, Fasano A, Petrella G. Normal Pressure Hydrocephalus in Down Syndrome: The Report of Two Cases. J Alzheimers Dis 2021; 77:979-984. [PMID: 32804139 DOI: 10.3233/jad-200409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Down syndrome (DS) is the most common cause of intellectual disability in infants and has a well-known relationship with the Alzheimer's disease. The association between DS and the other pathologies of senescence, such as normal pressure hydrocephalus (NPH), has been poorly investigated. This series included two DS patients with NPH. In both cases, NPH symptoms were initially misdiagnosed as DS associated senescence. Patients were treated with ventricular-peritoneal shunt, showing a sustained improvement (1 and 4 years of follow-up). To our knowledge, this is the first description of the occurrence of NPH in adult patients with DS and surgical outcomes.
Collapse
Affiliation(s)
- Massimo Marano
- Neurology, Neurophysiology and Neurobiology unit, Department of Medicine, University Campus Bio-Medico of Rome, Italy
| | - Angelo Pompucci
- Department of Neurosurgery, S. Maria Goretti Hospital, Latina, Italy
| | - Francesco Motolese
- Neurology, Neurophysiology and Neurobiology unit, Department of Medicine, University Campus Bio-Medico of Rome, Italy
| | - Mariagrazia Rossi
- Neurology, Neurophysiology and Neurobiology unit, Department of Medicine, University Campus Bio-Medico of Rome, Italy
| | | | - Vincenzo Di Lazzaro
- Neurology, Neurophysiology and Neurobiology unit, Department of Medicine, University Campus Bio-Medico of Rome, Italy
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre and the, Toronto Western Hospital, UHN, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada.,Krembil Brain Institute, Toronto, Ontario, Canada.,CenteR for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, Canada
| | | |
Collapse
|
22
|
Toshikawa H, Ikenaka A, Li L, Nishinaka-Arai Y, Niwa A, Ashida A, Kazuki Y, Nakahata T, Tamai H, Russell DW, Saito MK. N-Acetylcysteine prevents amyloid-β secretion in neurons derived from human pluripotent stem cells with trisomy 21. Sci Rep 2021; 11:17377. [PMID: 34462463 PMCID: PMC8405674 DOI: 10.1038/s41598-021-96697-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
Down syndrome (DS) is caused by the trisomy of chromosome 21. Among the many disabilities found in individuals with DS is an increased risk of early-onset Alzheimer's disease (AD). Although higher oxidative stress and an upregulation of amyloid β (Aβ) peptides from an extra copy of the APP gene are attributed to the AD susceptibility, the relationship between the two factors is unclear. To address this issue, we established an in vitro cellular model using neurons differentiated from DS patient-derived induced pluripotent stem cells (iPSCs) and isogenic euploid iPSCs. Neurons differentiated from DS patient-derived iPSCs secreted more Aβ compared to those differentiated from the euploid iPSCs. Treatment of the neurons with an antioxidant, N-acetylcysteine, significantly suppressed the Aβ secretion. These findings suggest that oxidative stress has an important role in controlling the Aβ level in neurons differentiated from DS patient-derived iPSCs and that N-acetylcysteine can be a potential therapeutic option to ameliorate the Aβ secretion.
Collapse
Affiliation(s)
- Hiromitsu Toshikawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Social Welfare Organization "SAISEIKAI" Imperial Gift Foundation Inc., Saiseikai Suita Hospital, Suita, 5640013, Japan
| | - Akihiro Ikenaka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Li Li
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Yoko Nishinaka-Arai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Ashida
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan.,Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Tatsutoshi Nakahata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Tamai
- Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan.,Institute for Developmental Brain Research, Osaka Medical and Pharmaceutical University, Takatsuki, 5690801, Japan
| | - David W Russell
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
23
|
Montoliu-Gaya L, Strydom A, Blennow K, Zetterberg H, Ashton NJ. Blood Biomarkers for Alzheimer's Disease in Down Syndrome. J Clin Med 2021; 10:3639. [PMID: 34441934 PMCID: PMC8397053 DOI: 10.3390/jcm10163639] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Epidemiological evidence suggests that by the age of 40 years, all individuals with Down syndrome (DS) have Alzheimer's disease (AD) neuropathology. Clinical diagnosis of dementia by cognitive assessment is complex in these patients due to the pre-existing and varying intellectual disability, which may mask subtle declines in cognitive functioning. Cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers, although accurate, are expensive, invasive, and particularly challenging in such a vulnerable population. The advances in ultra-sensitive detection methods have highlighted blood biomarkers as a valuable and realistic tool for AD diagnosis. Studies with DS patients have proven the potential blood-based biomarkers for sporadic AD (amyloid-β, tau, phosphorylated tau, and neurofilament light chain) to be useful in this population. In addition, biomarkers related to other pathologies that could aggravate dementia progression-such as inflammatory dysregulation, energetic imbalance, or oxidative stress-have been explored. This review serves to provide a brief overview of the main findings from the limited neuroimaging and CSF studies, outline the current state of blood biomarkers to diagnose AD in patients with DS, discuss possible past limitations of the research, and suggest considerations for developing and validating blood-based biomarkers in the future.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London WC2R 2LS, UK;
- South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- London Down Syndrome Consortium (LonDowns), London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Nicholas James Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden; (K.B.); (H.Z.); (N.J.A.)
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RT, UK
- NIHR Biomedical Research Centre for Mental Health, Biomedical Research Unit for Dementia at South London, Maudsley NHS Foundation, London SE5 8AF, UK
| |
Collapse
|
24
|
Fernández M, de Coo A, Quintela I, García E, Diniz-Freitas M, Limeres J, Diz P, Blanco J, Carracedo Á, Cruz R. Genetic Susceptibility to Periodontal Disease in Down Syndrome: A Case-Control Study. Int J Mol Sci 2021; 22:ijms22126274. [PMID: 34200970 PMCID: PMC8230717 DOI: 10.3390/ijms22126274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/26/2022] Open
Abstract
Severe periodontitis is prevalent in Down syndrome (DS). This study aimed to identify genetic variations associated with periodontitis in individuals with DS. The study group was distributed into DS patients with periodontitis (n = 50) and DS patients with healthy periodontium (n = 36). All samples were genotyped with the “Axiom Spanish Biobank” array, which contains 757,836 markers. An association analysis at the individual marker level using logistic regression, as well as at the gene level applying the sequence kernel association test (SKAT) was performed. The most significant genes were included in a pathway analysis using the free DAVID software. C12orf74 (rs4315121, p = 9.85 × 10−5, OR = 8.84), LOC101930064 (rs4814890, p = 9.61 × 10−5, OR = 0.13), KBTBD12 (rs1549874, p = 8.27 × 10−5, OR = 0.08), PIWIL1 (rs11060842, p = 7.82 × 10−5, OR = 9.05) and C16orf82 (rs62030877, p = 8.92 × 10−5, OR = 0.14) showed a higher probability in the individual analysis. The analysis at the gene level highlighted PIWIL, MIR9-2, LHCGR, TPR and BCR. At the signaling pathway level, PI3K-Akt, long-term depression and FoxO achieved nominal significance (p = 1.3 × 10−2, p = 5.1 × 10−3, p = 1.2 × 10−2, respectively). In summary, various metabolic pathways are involved in the pathogenesis of periodontitis in DS, including PI3K-Akt, which regulates cell proliferation and inflammatory response.
Collapse
Affiliation(s)
- María Fernández
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
| | - Alicia de Coo
- Grupo de Medicina Xenómica, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.d.C.); (I.Q.); (Á.C.); (R.C.)
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.d.C.); (I.Q.); (Á.C.); (R.C.)
- Centro Nacional de Genotipado, Plataforma de Recursos Biomoleculares, Instituto de Salud Carlos III (CeGen-PRB3-ISCIII), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Eliane García
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
| | - Márcio Diniz-Freitas
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
- Correspondence: ; Tel.: +34-981563100 (ext. 12344)
| | - Jacobo Limeres
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
| | - Pedro Diz
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
| | - Juan Blanco
- Grupo de Investigación en Odontología Médico-Quirúrgica (OMEQUI), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.F.); (E.G.); (J.L.); (P.D.); (J.B.)
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.d.C.); (I.Q.); (Á.C.); (R.C.)
- Centro Nacional de Genotipado, Plataforma de Recursos Biomoleculares, Instituto de Salud Carlos III (CeGen-PRB3-ISCIII), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), CIBERER-Instituto de Salud Carlos III, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica—SERGAS, 15706 Santiago de Compostela, Spain
| | - Raquel Cruz
- Grupo de Medicina Xenómica, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.d.C.); (I.Q.); (Á.C.); (R.C.)
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), CIBERER-Instituto de Salud Carlos III, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
25
|
Hendrix JA, Amon A, Abbeduto L, Agiovlasitis S, Alsaied T, Anderson HA, Bain LJ, Baumer N, Bhattacharyya A, Bogunovic D, Botteron KN, Capone G, Chandan P, Chase I, Chicoine B, Cieuta-Walti C, DeRuisseau LR, Durand S, Esbensen A, Fortea J, Giménez S, Granholm AC, Hahn LJ, Head E, Hillerstrom H, Jacola LM, Janicki MP, Jasien JM, Kamer AR, Kent RD, Khor B, Lawrence JB, Lemonnier C, Lewanda AF, Mobley W, Moore PE, Nelson LP, Oreskovic NM, Osorio RS, Patterson D, Rasmussen SA, Reeves RH, Roizen N, Santoro S, Sherman SL, Talib N, Tapia IE, Walsh KM, Warren SF, White AN, Wong GW, Yi JS. Opportunities, barriers, and recommendations in down syndrome research. TRANSLATIONAL SCIENCE OF RARE DISEASES 2021; 5:99-129. [PMID: 34268067 PMCID: PMC8279178 DOI: 10.3233/trd-200090] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent advances in medical care have increased life expectancy and improved the quality of life for people with Down syndrome (DS). These advances are the result of both pre-clinical and clinical research but much about DS is still poorly understood. In 2020, the NIH announced their plan to update their DS research plan and requested input from the scientific and advocacy community. OBJECTIVE The National Down Syndrome Society (NDSS) and the LuMind IDSC Foundation worked together with scientific and medical experts to develop recommendations for the NIH research plan. METHODS NDSS and LuMind IDSC assembled over 50 experts across multiple disciplines and organized them in eleven working groups focused on specific issues for people with DS. RESULTS This review article summarizes the research gaps and recommendations that have the potential to improve the health and quality of life for people with DS within the next decade. CONCLUSIONS This review highlights many of the scientific gaps that exist in DS research. Based on these gaps, a multidisciplinary group of DS experts has made recommendations to advance DS research. This paper may also aid policymakers and the DS community to build a comprehensive national DS research strategy.
Collapse
Affiliation(s)
| | - Angelika Amon
- Deceased. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA
| | | | - Tarek Alsaied
- Heart Institute Department of Pediatrics Cincinnati Children’s Hospital Medical Center University of Cincinnati, Cincinnati, OH, USA
| | | | | | - Nicole Baumer
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA; Down Syndrome Program, Developmental Medicine Center, Boston Children’s Hospital, Boston, MA, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Dusan Bogunovic
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Department of Pediatrics, Icahn School of Medicine at Mt. Sinai, New York, NY; Precision Immunology Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Kelly N. Botteron
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Priya Chandan
- Department of Neurosurgery, Division of Physical Medicine and Rehabilitation, University of Louisville School of Medicine, Louisville, KY, USA
| | - Isabelle Chase
- Department of Pediatric Dentistry, Boston Children’s Hospital, Boston, MA, USA
| | - Brian Chicoine
- Advocate Medical Group Adult Down Syndrome Center, Park Ridge, IL, USA
| | | | | | | | - Anna Esbensen
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Juan Fortea
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain; Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Sandra Giménez
- Multidisciplinary Sleep Unit, Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Laura J. Hahn
- Department of Speech and Hearing Science, University of Illinois Urbana Champaign, Champaign, IL, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, UC Irvine School of Medicine, Orange, CA, USA
| | | | - Lisa M. Jacola
- Department of Psychology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Joan M. Jasien
- Division of Pediatric Neurology, Duke University Health System, Durham, NC, USA
| | - Angela R. Kamer
- Department of Periodontology and Implant Dentistry, New York University, College of Dentistry, New York, NY, USA
| | - Raymond D. Kent
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Bernard Khor
- Benaroy Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jeanne B. Lawrence
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Amy Feldman Lewanda
- Children s National Rare Disease Institute, Children’s National Health System, Washington, DC., USA
| | - William Mobley
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Paul E. Moore
- Division of Allergy, Immunology, and Pulmonology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Nicolas M. Oreskovic
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Department of Internal Medicine, Massachusetts General Hospital, Boston, Mass
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ricardo S. Osorio
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - David Patterson
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
- Eleanor Roosevelt Institute, University of Denver, Denver, CO, USA; Department of Biological Sciences, University of Denver, Denver, CO, USA; Molecular and Cellular Biophysics Program, University of Denver, Denver, CO, USA
| | - Sonja A. Rasmussen
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL; Department of Epidemiology, University of Florida College of Public Health and Health Professions and College of Medicine, Gainesville, FL
| | - Roger H. Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nancy Roizen
- Department of Pediatrics, UH/Rainbow Babies and Children’s Hospital and Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Stephanie Santoro
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Stephanie L. Sherman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nasreen Talib
- Division of General Pediatrics, Children’s Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA
| | - Ignacio E. Tapia
- Sleep Center, Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kyle M. Walsh
- Division of Neuro-epidemiology, Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Steven F. Warren
- Institute for Life Span Studies, University of Kansas, Lawrence, KS, USA
| | - A. Nicole White
- Research Foundation, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Guang William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John S. Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
26
|
Santoro JD, Pagarkar D, Chu DT, Rosso M, Paulsen KC, Levitt P, Rafii MS. Neurologic complications of Down syndrome: a systematic review. J Neurol 2020; 268:4495-4509. [PMID: 32920658 DOI: 10.1007/s00415-020-10179-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022]
Abstract
Down syndrome (DS) is one of the most well-recognized genetic disorders. Persons with DS are known to have a variety of co-morbid medical problems, affecting nearly all organ systems. Improved healthcare interventions and research have allowed for increased life span of persons with DS, although disorders of the neurologic system remain underexplored. The purpose of this systematic review is to provide clinically pertinent information on the neurological phenotypes of frequently occurring or clinically relevant conditions. A retrospective review of MEDLINE, Scopus, and Pubmed were used to identify sources among seventeen, clinically relevant, search categories. MeSH terms all contained the phrase "Down Syndrome" in conjunction with the topic of interest. 'Frequently-occurring' was defined as prevalent in more than 10% of persons with DS across their lifespan, whereas 'clinically-relevant' was defined as a disease condition where early diagnosis or intervention can augment the disease course. In total, 4896 sources were identified with 159 sources meeting criteria for inclusion. Seventeen clinical conditions were grouped under the following subjects: hypotonia, intellectual and learning disability, cervical instability, autism spectrum disorder, epilepsy, cerebrovascular disease, Alzheimer's disease and neuropsychiatric disease. The results of this review provide a blueprint for the clinical neurologist taking care of persons with DS across the age spectrum and indicate that there are many underrecognized and misdiagnosed co-occurring conditions in DS, highlighting the need for further research.
Collapse
Affiliation(s)
- Jonathan D Santoro
- Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA. .,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Dania Pagarkar
- Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Duong T Chu
- Faculty of Medicine, Queen's University, Kingston, ON, Canada
| | - Mattia Rosso
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Kelli C Paulsen
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Department of Pediatrics and Program in Developmental Neuroscience and Developmental Neurogenetics, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael S Rafii
- Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Alzheimer's Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Sheline YI, Snider BJ, Beer JC, Seok D, Fagan AM, Suckow RF, Lee JM, Waligorska T, Korecka M, Aselcioglu I, Morris JC, Shaw LM, Cirrito JR. Effect of escitalopram dose and treatment duration on CSF Aβ levels in healthy older adults: A controlled clinical trial. Neurology 2020; 95:e2658-e2665. [PMID: 32913021 DOI: 10.1212/wnl.0000000000010725] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 06/08/2020] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To determine whether treatment with escitalopram compared with placebo would lower CSF β-amyloid 42 (Aβ42) levels. RATIONALE Serotonin signaling suppresses Aβ42 in animal models of Alzheimer disease (AD) and young healthy humans. In a prospective study in older adults, we examined dose and treatment duration effects of escitalopram. METHODS Using lumbar punctures to sample CSF levels before and after a course of escitalopram treatment, cognitively normal older adults (n = 114) were assigned to placebo, 20 mg escitalopram × 2 weeks, 20 mg escitalopram × 8 weeks, or 30 mg escitalopram × 8 weeks; CSF sampled pretreatment and posttreatment and within-subject percent change in Aβ42 was used as the primary outcome in subsequent analyses. RESULTS An overall 9.4% greater reduction in CSF Aβ42 was found in escitalopram-treated compared with placebo-treated groups (p < 0.001, 95% confidence interval [CI] 4.9%-14.2%, d = 0.81). Positive baseline Aβ status (CSF Aβ42 levels <250 pg/mL) was associated with smaller Aβ42 reduction (p = 0.006, 95% CI -16.7% to 0.5%, d = -0.52) compared with negative baseline amyloid status (CSF Aβ42 levels >250 pg/mL). CONCLUSIONS Short-term longitudinal doses of escitalopram decreased CSF Aβ42 in cognitively normal older adults, the target group for AD prevention. CLINICALTRIALSGOV IDENTIFIER NCT02161458. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that for cognitively normal older adults, escitalopram decreases CSF Aβ42.
Collapse
Affiliation(s)
- Yvette I Sheline
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York.
| | - B Joy Snider
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Joanne C Beer
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Darsol Seok
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Anne M Fagan
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Raymond F Suckow
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Jin-Moo Lee
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Teresa Waligorska
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Magdalena Korecka
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Irem Aselcioglu
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - John C Morris
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - Leslie M Shaw
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| | - John R Cirrito
- From the Center for Neuromodulation in Depression and Stress, Department of Psychiatry (Y.I.S., D.S., I.A.), and Departments of Radiology (Y.I.S.), Neurology (Y.I.S.), Biostatistics, Epidemiology and Bioinformatics (J.C.B.), and Pathology (T.W., M.K., L.M.S.), University of Pennsylvania, Philadelphia; Department of Neurology, The Knight Alzheimer Disease Research Center, Hope Center for Neurological Disorders (B.J.S., A.M.F., J.-M.L., J.C.M., J.R.C.), Washington University School of Medicine, St Louis MO; and New York State Psychiatric Institute and Department of Psychiatry (R.F.S.), Columbia University, New York
| |
Collapse
|
28
|
Snyder HM, Bain LJ, Brickman AM, Carrillo MC, Esbensen AJ, Espinosa JM, Fernandez F, Fortea J, Hartley SL, Head E, Hendrix J, Kishnani PS, Lai F, Lao P, Lemere C, Mobley W, Mufson EJ, Potter H, Zaman SH, Granholm AC, Rosas HD, Strydom A, Whitten MS, Rafii MS. Further understanding the connection between Alzheimer's disease and Down syndrome. Alzheimers Dement 2020; 16:1065-1077. [PMID: 32544310 PMCID: PMC8865308 DOI: 10.1002/alz.12112] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/25/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Improved medical care of individuals with Down syndrome (DS) has led to an increase in life expectancy to over the age of 60 years. In conjunction, there has been an increase in age-related co-occurring conditions including Alzheimer's disease (AD). Understanding the factors that underlie symptom and age of clinical presentation of dementia in people with DS may provide insights into the mechanisms of sporadic and DS-associated AD (DS-AD). In March 2019, the Alzheimer's Association, Global Down Syndrome Foundation and the LuMind IDSC Foundation partnered to convene a workshop to explore the state of the research on the intersection of AD and DS research; to identify research gaps and unmet needs; and to consider how best to advance the field. This article provides a summary of discussions, including noting areas of emerging science and discovery, considerations for future studies, and identifying open gaps in our understanding for future focus.
Collapse
Affiliation(s)
- Heather M. Snyder
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Lisa J. Bain
- Independent Science Writer, Elverson, Pennsylvania, USA
| | - Adam M. Brickman
- Department of Neurology, College of Physicians and Surgeons, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Maria C. Carrillo
- Alzheimer’s Association, Medical & Scientific Relations, Chicago, Illinois, USA
| | - Anna J. Esbensen
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center & University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Joaquin M. Espinosa
- Department of Pharmacology, Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Fabian Fernandez
- Departments of Psychology and Neurology, BIO5 Institute, and The Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Juan Fortea
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autonoma de Barcelona, CIBERNED, Barcelona, Spain
- Down Medical Center, Catalan Down Syndrome Foundation, Barcelona, Spain
| | - Sigan L. Hartley
- Department of Human Development and Family Studies, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, California, USA
| | - James Hendrix
- LuMind IDSC Foundation, Burlington, Massachusetts, USA
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Florence Lai
- Department of Neurology, Harvard University/Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick Lao
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Cynthia Lemere
- Department of Neurology, Brigham & Women’s Hospital and Harvard University, Boston, Massachusetts, USA
| | - William Mobley
- Department of Neurosciences, University of California, San Diego, San Diego, California, USA
| | | | - Huntington Potter
- Rocky Mountain Alzheimer’s Disease Center and Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Shahid H. Zaman
- Cambridge Intellectual & Developmental Disability Research Group, Department of Psychiatry University of Cambridge, Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, UK
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, USA
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - H. Diana Rosas
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Psychology and Neuroscience, King’s College London, South London and the Maudsley NHS Foundation Trust, LonDowns Consortium, Institute of Psychiatry, London, UK
| | | | - Michael S. Rafii
- Alzheimer’s Therapeutics Research Institute and Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
29
|
Molton IR, Ordway A. Aging With Disability: Populations, Programs, and the New Paradigm An Introduction to the Special Issue. J Aging Health 2020; 31:3S-20S. [PMID: 31718415 DOI: 10.1177/0898264319880120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: The purpose of this article is to introduce a special issue dedicated to research at the intersection of aging and disability. Method: We provide some context for the importance of cross-disciplinary collaboration among aging and disability researchers and summarize the nine articles in this issue. Results: Articles in the special issue are centered around several overarching themes. These include meaningful social and community participation, goals and values in the context of disability, and the reach and effectiveness of programs and policies on rehabilitation and service utilization. Conclusion: As care models continue to merge aging and disability services, collaboration among traditional aging and disability research networks can lead to improved outcomes for adults aging with long-term disability.
Collapse
|
30
|
White matter hyperintensities are associated with subthreshold amyloid accumulation. Neuroimage 2020; 218:116944. [PMID: 32445880 DOI: 10.1016/j.neuroimage.2020.116944] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 11/20/2022] Open
Abstract
The association between white matter hyperintensities (WMH) and amyloid accumulation over time in cognitively normal, amyloid-negative elderly people remains largely unexplored. In order to study whether baseline WMH were associated with longitudinal subthreshold amyloid accumulation, 159 cognitively normal participants from the Alzheimer's Disease Neuroimaging Initiative who were amyloid-negative at baseline were examined. All the participants underwent a T1 and a Fluid-Attenuated Inversion Recovery MRI scan at baseline. Amyloid PET imaging was performed at baseline and follow-up visits in 2-year intervals for up to 8 years. Partial volume correction was applied for quantifying cortical Standardised Uptake Value Ratios (SUVR). The associations between global and regional WMH burden and amyloid accumulation were assessed using linear mixed models adjusted by demographic characteristics and baseline SUVR. Partial volume correction increased the measured annual rate of change (+2.4%) compared to that obtained from non-corrected data (+0.5%). There were no significant correlations between baseline WMHs and baseline subthreshold cortical amyloid uptake. In a longitudinal analysis, increased baseline cortical SUVR and increased baseline burden of global (p = 0.006), frontal (p = 0.006), and parietal WMH (p = 0.003) were associated with faster amyloid accumulation. WMH-related amyloid accumulation occurred in parietal, frontal, and, to a lesser extent, cingulate cortices. These results remained unchanged after a sensitivity analysis excluding participants with the highest cortical SUVRs. This is the first study to identify a specific spatial distribution of WMH which is associated with future amyloid accumulation in cognitively normal elderly subjects without PET-detectable amyloid pathology. These findings may have important implications in prevention trials for the early identification of amyloid accumulation.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW People with Down syndrome represent the world's largest population with a genetic risk for Alzheimer's disease. This review will provide a short summary of what is known and will include recent findings from the field. RECENT FINDINGS There has been an increasing focus on biomarker research in this population, with a number of studies presenting findings on promising new markers - Neurofilament Light (NfL) appears to be one such promising marker that has emerged. Imaging studies have increased our knowledge on the progression of Alzheimer's disease in this population. SUMMARY The inclusion of people with Down syndrome in dementia research is vital from a scientific and an equity perspective. Recent advances in the field can have further impact with multisite, cross country collaborative efforts. For this to happen, instruments need to be validated across language and cultures.
Collapse
|
32
|
Rueda N, Flórez J, Dierssen M, Martínez-Cué C. Translational validity and implications of pharmacotherapies in preclinical models of Down syndrome. PROGRESS IN BRAIN RESEARCH 2019; 251:245-268. [PMID: 32057309 DOI: 10.1016/bs.pbr.2019.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders are challenging to study in the laboratory, and despite a large investment, few novel treatments have been developed in the last decade. While animal models have been valuable in elucidating disease mechanisms and in providing insights into the function of specific genes, the predictive validity of preclinical models to test potential therapies has been questioned. In the last two decades, diverse new murine models of Down syndrome (DS) have been developed and numerous studies have demonstrated neurobiological alterations that could be responsible for the cognitive and behavioral phenotypes found in this syndrome. In many cases, similar alterations were found in murine models and in individuals with DS, although several phenotypes shown in animals have yet not been confirmed in the human condition. Some of the neurobiological alterations observed in mice have been proposed to account for their changes in cognition and behavior, and have received special attention because of being putative therapeutic targets. Those include increased oxidative stress, altered neurogenesis, overexpression of the Dyrk1A gene, GABA-mediated overinhibition and Alzheimer's disease-related neurodegeneration. Subsequently, different laboratories have tested the efficacy of pharmacotherapies targeting these alterations. Unfortunately, animal models are limited in their ability to mimic the extremely complex process of human neurodevelopment and neuropathology. Therefore, the safety and efficacy identified in animal studies are not always translated to humans, and most of the drugs tested have not demonstrated any positive effect or very limited efficacy in clinical trials. Despite their limitations, though, animal trials give us extremely valuable information for developing and testing drugs for human use that cannot be obtained from molecular or cellular experiments alone. This chapter reviews some of these therapeutic approaches and discusses some reasons that could account for the discrepancy between the findings in mouse models of DS and in humans, including: (i) the incomplete resemble of the genetic alterations of available mouse models of DS and human trisomy 21, (ii) the lack of evidence that some of the phenotypic alterations found in mice (e.g., GABA-mediated overinhibition, and alterations in adult neurogenesis) are also present in DS individuals, and (iii) the inaccuracy and/or inadequacy of the methods used in clinical trials to detect changes in the cognitive and behavioral functions of people with DS. Despite the shortcomings of animal models, animal experimentation remains an invaluable tool in developing drugs. Thus, we will also discuss how to increase predictive validity of mouse models.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, University of Cantabria, Santander, Spain
| | - Jesús Flórez
- Department of Physiology and Pharmacology, University of Cantabria, Santander, Spain
| | - Mara Dierssen
- Cellular and Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, Spain.
| |
Collapse
|
33
|
Affiliation(s)
- Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, California
| | - Joseph H Lee
- Department of Epidemiology, Department of Neurology, Columbia University Medical Center, Sergievsky Center, Taub Institute, New York, New York
| |
Collapse
|