1
|
O'Connor A, Ryan NS, Belder CRS, Lynch DS, Lahiri N, Houlden H, Rohrer JD, Fox NC, O'Dowd S. Genetic testing in dementia. Pract Neurol 2025; 25:127-136. [PMID: 39288984 DOI: 10.1136/pn-2024-004241] [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] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
There is growing public awareness and concern regarding dementia risk. In addition, genetic testing is increasingly accessible and is at the point of being integrated into routine clinical practice. As a result, there is a pressing need for treating clinicians to have the appropriate knowledge base to request and consent for diagnostic genetic testing in cognitive clinics. We outline our approach to genetic testing in patients with Alzheimer's disease, frontotemporal dementia, dementia with Lewy bodies and vascular cognitive impairment. We discuss when to consider testing, the consenting process, and the interpretation and communication of genetic test results.
Collapse
Affiliation(s)
- Antoinette O'Connor
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Tallaght Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Christopher R S Belder
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- The University of Adelaide, Adelaide, South Australia, Australia
| | - David S Lynch
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and National Hospital for Neurology & Neurosurgery, London, UK
| | - Nayana Lahiri
- St. George's, University of London & St George's University Hospitals NHS Foundation Trust, Cardiovascular and Genomics Institute, London, UK
| | - Henry Houlden
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and National Hospital for Neurology & Neurosurgery, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Sean O'Dowd
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
- National Dementia Services, Health Services Executive, Ireland
| |
Collapse
|
2
|
Kedia S, Simons M. Oligodendrocytes in Alzheimer's disease pathophysiology. Nat Neurosci 2025; 28:446-456. [PMID: 39881195 DOI: 10.1038/s41593-025-01873-x] [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: 04/20/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Our understanding of Alzheimer's disease (AD) has transformed from a purely neuronal perspective to one that acknowledges the involvement of glial cells. Despite remarkable progress in unraveling the biology of microglia, astrocytes and vascular elements, the exploration of oligodendrocytes in AD is still in its early stages. Contrary to the traditional notion of oligodendrocytes as passive bystanders in AD pathology, emerging evidence indicates their active participation in and reaction to amyloid and tau pathology. Oligodendrocytes undergo a functional transition to a disease-associated state, engaging in immune modulation, stress responses and cellular survival. Far from being inert players, they appear to serve a dual role in AD pathogenesis, potentially offering defense mechanisms against pathology while also contributing to disease progression. This Review explores recent advancements in understanding the roles of oligodendrocytes and their myelin sheaths in the context of AD, shedding light on their complex interactions within the disease pathology.
Collapse
Affiliation(s)
- Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
3
|
Yoshimoto T, Yamagami H, Matsumaru Y. Recent Advances in Stroke Genetics-Unraveling the Complexity of Cerebral Infarction: A Brief Review. Genes (Basel) 2025; 16:59. [PMID: 39858606 PMCID: PMC11764629 DOI: 10.3390/genes16010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND/OBJECTIVES Recent advances in stroke genetics have substantially enhanced our understanding of the complex genetic architecture underlying cerebral infarction and other stroke subtypes. As knowledge in this field expands, healthcare providers must remain informed about these latest developments. This review aims to provide a comprehensive overview of recent advances in stroke genetics, with a focus on cerebral infarction, and discuss their potential impact on patient care and future research directions. METHODS We reviewed recent literature about advances in stroke genetics, focusing on cerebral infarction, and discussed their potential impact on patient care and future research directions. Key developments include the identification of monogenic stroke syndromes, such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, and cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy caused by mutations in the NOTCH3 and HTRA1 genes, respectively. In addition, the role of RNF213 in moyamoya disease and other cerebrovascular disorders, particularly in East Asian populations, has been elucidated. The development of polygenic risk scores for assessing genetic predisposition to stroke has demonstrated the potential to improve risk prediction beyond traditional factors. Genetic studies have also elucidated the distinct genetic architecture of stroke subtypes, including large artery atherosclerosis, small vessel disease, and cardioembolic stroke. Furthermore, the investigation of epigenetic modifications influencing stroke risk and its outcomes has revealed new research avenues, while advancements in pharmacogenomics highlight the potential for personalized stroke treatment based on individual genetic profiles. CONCLUSIONS These genetic discoveries have important clinical implications, including improved risk stratification, targeted prevention strategies, and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Takeshi Yoshimoto
- Department of Stroke and Cerebrovascular Diseases, University of Tsukuba Hospital, Tsukuba 305-8576, Japan;
- Division of Stroke Prevention and Treatment, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Hiroshi Yamagami
- Division of Stroke Prevention and Treatment, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Yuji Matsumaru
- Department of Stroke and Cerebrovascular Diseases, University of Tsukuba Hospital, Tsukuba 305-8576, Japan;
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| |
Collapse
|
4
|
Volloch V, Rits-Volloch S. Production of Amyloid-β in the Aβ-Protein-Precursor Proteolytic Pathway Is Discontinued or Severely Suppressed in Alzheimer's Disease-Affected Neurons: Contesting the 'Obvious'. Genes (Basel) 2025; 16:46. [PMID: 39858593 PMCID: PMC11764795 DOI: 10.3390/genes16010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
A notion of the continuous production of amyloid-β (Aβ) via the proteolysis of Aβ-protein-precursor (AβPP) in Alzheimer's disease (AD)-affected neurons constitutes both a cornerstone and an article of faith in the Alzheimer's research field. The present Perspective challenges this assumption. It analyses the relevant empirical data and reaches an unexpected conclusion, namely that in AD-afflicted neurons, the production of AβPP-derived Aβ is either discontinued or severely suppressed, a concept that, if proven, would fundamentally change our understanding of the disease. This suppression, effectively self-suppression, occurs in the context of the global inhibition of the cellular cap-dependent protein synthesis as a consequence of the neuronal integrated stress response (ISR) elicited by AβPP-derived intraneuronal Aβ (iAβ; hence self-suppression) upon reaching certain levels. Concurrently with the suppression of the AβPP proteolytic pathway, the neuronal ISR activates in human neurons, but not in mouse neurons, the powerful AD-driving pathway generating the C99 fragment of AβPP independently of AβPP. The present study describes molecular mechanisms potentially involved in these phenomena, propounds novel approaches to generate transgenic animal models of AD, advocates for the utilization of human neuronal cells-based models of the disease, makes verifiable predictions, suggests experiments designed to validate the proposed concept, and considers its potential research and therapeutic implications. Remarkably, it opens up the possibility that the conventional production of AβPP, BACE enzymes, and γ-secretase components is also suppressed under the neuronal ISR conditions in AD-affected neurons, resulting in the dyshomeostasis of AβPP. It follows that whereas conventional AD is triggered by AβPP-derived iAβ accumulated to the ISR-eliciting levels, the disease, in its both conventional and unconventional (triggered by the neuronal ISR-eliciting stressors distinct from iAβ) forms, is driven not (or not only) by iAβ produced in the AβPP-independent pathway, as we proposed previously, but mainly, possibly exclusively, by the C99 fragment generated independently of AβPP and not cleaved at the γ-site due to the neuronal ISR-caused deficiency of γ-secretase (apparently, the AD-driving "substance X" predicted in our previous study), a paradigm consistent with a dictum by George Perry that Aβ is "central but not causative" in AD. The proposed therapeutic strategies would not only deplete the driver of the disease and abrogate the AβPP-independent production of C99 but also reverse the neuronal ISR and ameliorate the AβPP dyshomeostasis, a potentially significant contributor to AD pathology.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Volloch V, Rits-Volloch S. Quintessential Synergy: Concurrent Transient Administration of Integrated Stress Response Inhibitors and BACE1 and/or BACE2 Activators as the Optimal Therapeutic Strategy for Alzheimer's Disease. Int J Mol Sci 2024; 25:9913. [PMID: 39337400 PMCID: PMC11432332 DOI: 10.3390/ijms25189913] [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: 08/14/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The present study analyzes two potential therapeutic approaches for Alzheimer's disease (AD). One is the suppression of the neuronal integrated stress response (ISR). Another is the targeted degradation of intraneuronal amyloid-beta (iAβ) via the activation of BACE1 (Beta-site Aβ-protein-precursor Cleaving Enzyme) and/or BACE2. Both approaches are rational. Both are promising. Both have substantial intrinsic limitations. However, when combined in a carefully orchestrated manner into a composite therapy they display a prototypical synergy and constitute the apparently optimal, potentially most effective therapeutic strategy for AD.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
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
|
7
|
Iaccarino L, Llibre-Guerra JJ, McDade E, Edwards L, Gordon B, Benzinger T, Hassenstab J, Kramer JH, Li Y, Miller BL, Miller Z, Morris JC, Mundada N, Perrin RJ, Rosen HJ, Soleimani-Meigooni D, Strom A, Tsoy E, Wang G, Xiong C, Allegri R, Chrem P, Vazquez S, Berman SB, Chhatwal J, Masters CL, Farlow MR, Jucker M, Levin J, Salloway S, Fox NC, Day GS, Gorno-Tempini ML, Boxer AL, La Joie R, Bateman R, Rabinovici GD. Molecular neuroimaging in dominantly inherited versus sporadic early-onset Alzheimer's disease. Brain Commun 2024; 6:fcae159. [PMID: 38784820 PMCID: PMC11114609 DOI: 10.1093/braincomms/fcae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 03/14/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Approximately 5% of Alzheimer's disease patients develop symptoms before age 65 (early-onset Alzheimer's disease), with either sporadic (sporadic early-onset Alzheimer's disease) or dominantly inherited (dominantly inherited Alzheimer's disease) presentations. Both sporadic early-onset Alzheimer's disease and dominantly inherited Alzheimer's disease are characterized by brain amyloid-β accumulation, tau tangles, hypometabolism and neurodegeneration, but differences in topography and magnitude of these pathological changes are not fully elucidated. In this study, we directly compared patterns of amyloid-β plaque deposition and glucose hypometabolism in sporadic early-onset Alzheimer's disease and dominantly inherited Alzheimer's disease individuals. Our analysis included 134 symptomatic sporadic early-onset Alzheimer's disease amyloid-Positron Emission Tomography (PET)-positive cases from the University of California, San Francisco, Alzheimer's Disease Research Center (mean ± SD age 59.7 ± 5.6 years), 89 symptomatic dominantly inherited Alzheimer's disease cases (age 45.8 ± 9.3 years) and 102 cognitively unimpaired non-mutation carriers from the Dominantly Inherited Alzheimer Network study (age 44.9 ± 9.2). Each group underwent clinical and cognitive examinations, 11C-labelled Pittsburgh Compound B-PET and structural MRI. 18F-Fluorodeoxyglucose-PET was also available for most participants. Positron Emission Tomography scans from both studies were uniformly processed to obtain a standardized uptake value ratio (PIB50-70 cerebellar grey reference and FDG30-60 pons reference) images. Statistical analyses included pairwise global and voxelwise group comparisons and group-independent component analyses. Analyses were performed also adjusting for covariates including age, sex, Mini-Mental State Examination, apolipoprotein ε4 status and average composite cortical of standardized uptake value ratio. Compared with dominantly inherited Alzheimer's disease, sporadic early-onset Alzheimer's disease participants were older at age of onset (mean ± SD, 54.8 ± 8.2 versus 41.9 ± 8.2, Cohen's d = 1.91), with more years of education (16.4 ± 2.8 versus 13.5 ± 3.2, d = 1) and more likely to be apolipoprotein ε4 carriers (54.6% ε4 versus 28.1%, Cramer's V = 0.26), but similar Mini-Mental State Examination (20.6 ± 6.1 versus 21.2 ± 7.4, d = 0.08). Sporadic early-onset Alzheimer's disease had higher global cortical Pittsburgh Compound B-PET binding (mean ± SD standardized uptake value ratio, 1.92 ± 0.29 versus 1.58 ± 0.44, d = 0.96) and greater global cortical 18F-fluorodeoxyglucose-PET hypometabolism (mean ± SD standardized uptake value ratio, 1.32 ± 0.1 versus 1.39 ± 0.19, d = 0.48) compared with dominantly inherited Alzheimer's disease. Fully adjusted comparisons demonstrated relatively higher Pittsburgh Compound B-PET standardized uptake value ratio in the medial occipital, thalami, basal ganglia and medial/dorsal frontal regions in dominantly inherited Alzheimer's disease versus sporadic early-onset Alzheimer's disease. Sporadic early-onset Alzheimer's disease showed relatively greater 18F-fluorodeoxyglucose-PET hypometabolism in Alzheimer's disease signature temporoparietal regions and caudate nuclei, whereas dominantly inherited Alzheimer's disease showed relatively greater hypometabolism in frontal white matter and pericentral regions. Independent component analyses largely replicated these findings by highlighting common and unique Pittsburgh Compound B-PET and 18F-fluorodeoxyglucose-PET binding patterns. In summary, our findings suggest both common and distinct patterns of amyloid and glucose hypometabolism in sporadic and dominantly inherited early-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jorge J Llibre-Guerra
- The Dominantly Inherited Alzheimer Network (DIAN), St Louis, MO 63108, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Eric McDade
- The Dominantly Inherited Alzheimer Network (DIAN), St Louis, MO 63108, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Brian Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Jason Hassenstab
- The Dominantly Inherited Alzheimer Network (DIAN), St Louis, MO 63108, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yan Li
- Department of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Zachary Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - John C Morris
- The Dominantly Inherited Alzheimer Network (DIAN), St Louis, MO 63108, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Richard J Perrin
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - David Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Elena Tsoy
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Guoqiao Wang
- Department of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires 1428, Argentina
| | - Patricio Chrem
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires 1428, Argentina
| | - Silvia Vazquez
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires 1428, Argentina
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Colin L Masters
- Department of Neuroscience, Florey Institute, The University of Melbourne, Melbourne 3052, Australia
| | - Martin R Farlow
- Neuroscience Center, Indiana University School of Medicine at Indianapolis, Indiana, IN 46202, USA
| | - Mathias Jucker
- DZNE-German Center for Neurodegenerative Diseases, Tübingen 72076, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich 80539, Germany
- German Center for Neurodegenerative Diseases, Munich 81377, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich 81377, Germany
| | - Stephen Salloway
- Memory & Aging Program, Butler Hospital, Brown University in Providence, RI 02906, USA
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, UK
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL 33224, USA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Randall Bateman
- The Dominantly Inherited Alzheimer Network (DIAN), St Louis, MO 63108, USA
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
8
|
Joseph‐Mathurin N, Feldman RL, Lu R, Shirzadi Z, Toomer C, Saint Clair JR, Ma Y, McKay NS, Strain JF, Kilgore C, Friedrichsen KA, Chen CD, Gordon BA, Chen G, Hornbeck RC, Massoumzadeh P, McCullough AA, Wang Q, Li Y, Wang G, Keefe SJ, Schultz SA, Cruchaga C, Preboske GM, Jack CR, Llibre‐Guerra JJ, Allegri RF, Ances BM, Berman SB, Brooks WS, Cash DM, Day GS, Fox NC, Fulham M, Ghetti B, Johnson KA, Jucker M, Klunk WE, la Fougère C, Levin J, Niimi Y, Oh H, Perrin RJ, Reischl G, Ringman JM, Saykin AJ, Schofield PR, Su Y, Supnet‐Bell C, Vöglein J, Yakushev I, Brickman AM, Morris JC, McDade E, Xiong C, Bateman RJ, Chhatwal JP, Benzinger TLS. Presenilin-1 mutation position influences amyloidosis, small vessel disease, and dementia with disease stage. Alzheimers Dement 2024; 20:2680-2697. [PMID: 38380882 PMCID: PMC11032566 DOI: 10.1002/alz.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Amyloidosis, including cerebral amyloid angiopathy, and markers of small vessel disease (SVD) vary across dominantly inherited Alzheimer's disease (DIAD) presenilin-1 (PSEN1) mutation carriers. We investigated how mutation position relative to codon 200 (pre-/postcodon 200) influences these pathologic features and dementia at different stages. METHODS Individuals from families with known PSEN1 mutations (n = 393) underwent neuroimaging and clinical assessments. We cross-sectionally evaluated regional Pittsburgh compound B-positron emission tomography uptake, magnetic resonance imaging markers of SVD (diffusion tensor imaging-based white matter injury, white matter hyperintensity volumes, and microhemorrhages), and cognition. RESULTS Postcodon 200 carriers had lower amyloid burden in all regions but worse markers of SVD and worse Clinical Dementia Rating® scores compared to precodon 200 carriers as a function of estimated years to symptom onset. Markers of SVD partially mediated the mutation position effects on clinical measures. DISCUSSION We demonstrated the genotypic variability behind spatiotemporal amyloidosis, SVD, and clinical presentation in DIAD, which may inform patient prognosis and clinical trials. HIGHLIGHTS Mutation position influences Aβ burden, SVD, and dementia. PSEN1 pre-200 group had stronger associations between Aβ burden and disease stage. PSEN1 post-200 group had stronger associations between SVD markers and disease stage. PSEN1 post-200 group had worse dementia score than pre-200 in late disease stage. Diffusion tensor imaging-based SVD markers mediated mutation position effects on dementia in the late stage.
Collapse
|
9
|
Banerjee G, Schott JM, Ryan NS. Familial cerebral amyloid disorders with prominent white matter involvement. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:289-315. [PMID: 39322385 DOI: 10.1016/b978-0-323-99209-1.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Familial cerebral amyloid disorders are characterized by the accumulation of fibrillar protein aggregates, which deposit in the parenchyma as plaques and in the vasculature as cerebral amyloid angiopathy (CAA). Amyloid β (Aβ) is the most common of these amyloid proteins, accumulating in familial and sporadic forms of Alzheimer's disease and CAA. However, there are also a number of rare, hereditary, non-Aβ cerebral amyloidosis. The clinical manifestations of these familial cerebral amyloid disorders are diverse, including cognitive or neuropsychiatric presentations, intracerebral hemorrhage, seizures, myoclonus, headache, ataxia, and spasticity. Some mutations are associated with extensive white matter hyperintensities on imaging, which may or may not be accompanied by hemorrhagic imaging markers of CAA; others are associated with occipital calcification. We describe the clinical, imaging, and pathologic features of these disorders and discuss putative disease mechanisms. Familial disorders of cerebral amyloid accumulation offer unique insights into the contributions of vascular and parenchymal amyloid to pathogenesis and the pathways underlying white matter involvement in neurodegeneration. With Aβ immunotherapies now entering the clinical realm, gaining a deeper understanding of these processes and the relationships between genotype and phenotype has never been more relevant.
Collapse
Affiliation(s)
- Gargi Banerjee
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, United Kingdom
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom.
| |
Collapse
|
10
|
Millar PR, Gordon BA, Wisch JK, Schultz SA, Benzinger TL, Cruchaga C, Hassenstab JJ, Ibanez L, Karch C, Llibre-Guerra JJ, Morris JC, Perrin RJ, Supnet-Bell C, Xiong C, Allegri RF, Berman SB, Chhatwal JP, Chrem Mendez PA, Day GS, Hofmann A, Ikeuchi T, Jucker M, Lee JH, Levin J, Lopera F, Niimi Y, Sánchez-González VJ, Schofield PR, Sosa-Ortiz AL, Vöglein J, Bateman RJ, Ances BM, McDade EM. Advanced structural brain aging in preclinical autosomal dominant Alzheimer disease. Mol Neurodegener 2023; 18:98. [PMID: 38111006 PMCID: PMC10729487 DOI: 10.1186/s13024-023-00688-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND "Brain-predicted age" estimates biological age from complex, nonlinear features in neuroimaging scans. The brain age gap (BAG) between predicted and chronological age is elevated in sporadic Alzheimer disease (AD), but is underexplored in autosomal dominant AD (ADAD), in which AD progression is highly predictable with minimal confounding age-related co-pathology. METHODS We modeled BAG in 257 deeply-phenotyped ADAD mutation-carriers and 179 non-carriers from the Dominantly Inherited Alzheimer Network using minimally-processed structural MRI scans. We then tested whether BAG differed as a function of mutation and cognitive status, or estimated years until symptom onset, and whether it was associated with established markers of amyloid (PiB PET, CSF amyloid-β-42/40), phosphorylated tau (CSF and plasma pTau-181), neurodegeneration (CSF and plasma neurofilament-light-chain [NfL]), and cognition (global neuropsychological composite and CDR-sum of boxes). We compared BAG to other MRI measures, and examined heterogeneity in BAG as a function of ADAD mutation variants, APOE ε4 carrier status, sex, and education. RESULTS Advanced brain aging was observed in mutation-carriers approximately 7 years before expected symptom onset, in line with other established structural indicators of atrophy. BAG was moderately associated with amyloid PET and strongly associated with pTau-181, NfL, and cognition in mutation-carriers. Mutation variants, sex, and years of education contributed to variability in BAG. CONCLUSIONS We extend prior work using BAG from sporadic AD to ADAD, noting consistent results. BAG associates well with markers of pTau, neurodegeneration, and cognition, but to a lesser extent, amyloid, in ADAD. BAG may capture similar signal to established MRI measures. However, BAG offers unique benefits in simplicity of data processing and interpretation. Thus, results in this unique ADAD cohort with few age-related confounds suggest that brain aging attributable to AD neuropathology can be accurately quantified from minimally-processed MRI.
Collapse
Affiliation(s)
- Peter R Millar
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Stephanie A Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Tammie Ls Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- NeuroGenomics & Informatics Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Celeste Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | | | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Chengjie Xiong
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Yoshiki Niimi
- Unit for Early and Exploratory Clinical Development, The University of Tokyo Hospital, Bunkyo-Ku, Tokyo, Japan
| | - Victor J Sánchez-González
- Departamento de Clínicas, CUALTOS, Universidad de Guadalajara, Tepatitlán de Morelos, Jalisco, México
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ana Luisa Sosa-Ortiz
- Instituto Nacional de Neurologia y Neurocirugía MVS, CDMX, Ciudad de México, Mexico
| | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric M McDade
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
11
|
Banerjee G, Collinge J, Fox NC, Lashley T, Mead S, Schott JM, Werring DJ, Ryan NS. Clinical considerations in early-onset cerebral amyloid angiopathy. Brain 2023; 146:3991-4014. [PMID: 37280119 PMCID: PMC10545523 DOI: 10.1093/brain/awad193] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/16/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an important cerebral small vessel disease associated with brain haemorrhage and cognitive change. The commonest form, sporadic amyloid-β CAA, usually affects people in mid- to later life. However, early-onset forms, though uncommon, are increasingly recognized and may result from genetic or iatrogenic causes that warrant specific and focused investigation and management. In this review, we firstly describe the causes of early-onset CAA, including monogenic causes of amyloid-β CAA (APP missense mutations and copy number variants; mutations of PSEN1 and PSEN2) and non-amyloid-β CAA (associated with ITM2B, CST3, GSN, PRNP and TTR mutations), and other unusual sporadic and acquired causes including the newly-recognized iatrogenic subtype. We then provide a structured approach for investigating early-onset CAA, and highlight important management considerations. Improving awareness of these unusual forms of CAA amongst healthcare professionals is essential for facilitating their prompt diagnosis, and an understanding of their underlying pathophysiology may have implications for more common, late-onset, forms of the disease.
Collapse
Affiliation(s)
- Gargi Banerjee
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - John Collinge
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, London, W1 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Simon Mead
- MRC Prion Unit at University College London (UCL), Institute of Prion Diseases, UCL, London, W1W 7FF, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - David J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| |
Collapse
|
12
|
Liang Z, Wu Y, Li C, Liu Z. Clinical and genetic characteristics in a central-southern Chinese cohort of early-onset Alzheimer's disease. Front Neurol 2023; 14:1119326. [PMID: 37051054 PMCID: PMC10084792 DOI: 10.3389/fneur.2023.1119326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
BackgroundMutations in the presenilin-1 (PSEN1), presenilin-2 (PSEN2), and amyloid precursor protein (APP) genes have been commonly identified in early-onset Alzheimer's disease (EOAD). Some of the mutations in the three causative genes, especially the PSEN1 gene, result in variable phenotypes and exhibit clinical heterogeneity among EOAD families.MethodsUsing next-generation sequencing (NGS), we performed genetic screening in a Chinese cohort of 18 patients with EOAD, consisting of five familial EOAD and 13 sporadic cases.ResultsWe identified two likely pathogenic PSEN1 mutations (one novel) and a novel APP mutation in three cases of EOAD, where two are familial and one is sporadic, respectively. In addition, we detected a few variants of uncertain significance (VUS) in several genes, including not only the two known variants in PSEN2 (p.H169N and p.V214L) but also genes causal of other types of dementia or previously identified as risk factors for AD, suggesting the possible involvement of multiple genes in the etiopathology of AD. The patients carrying PSEN1 mutations had an earlier mean age at the onset than those with PSEN2 or APP variants. The initial symptoms varied greatly among patients in the EOAD cohort, from progressive memory impairment and epilepsy to uncommon motor symptoms such as involuntary tremors in the upper extremities.ConclusionsIn conclusion, our study provides further evidence of the genetic profile of patients with EOAD from China and expands the mutation spectrum of both PSEN1 and APP. In addition, our results highlight the clinical heterogeneity in patients with EOAD and mutations in PSEN1, PSEN2, and APP and suggest strong effects of genetic variants on clinical phenotypes. Future functional studies are needed to clarify the interaction between AD-causative gene mutations and phenotypic heterogeneity.
Collapse
Affiliation(s)
- Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chuanzhou Li
| | - Zhijun Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhijun Liu
| |
Collapse
|
13
|
Andjelkovic AV, Situ M, Citalan-Madrid AF, Stamatovic SM, Xiang J, Keep RF. Blood-Brain Barrier Dysfunction in Normal Aging and Neurodegeneration: Mechanisms, Impact, and Treatments. Stroke 2023; 54:661-672. [PMID: 36848419 PMCID: PMC9993074 DOI: 10.1161/strokeaha.122.040578] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Cerebral endothelial cells and their linking tight junctions form a unique, dynamic and multi-functional interface, the blood-brain barrier (BBB). The endothelium is regulated by perivascular cells and components forming the neurovascular unit. This review examines BBB and neurovascular unit changes in normal aging and in neurodegenerative disorders, particularly focusing on Alzheimer disease, cerebral amyloid angiopathy and vascular dementia. Increasing evidence indicates BBB dysfunction contributes to neurodegeneration. Mechanisms underlying BBB dysfunction are outlined (endothelium and neurovascular unit mediated) as is the BBB as a therapeutic target including increasing the uptake of systemically delivered therapeutics across the BBB, enhancing clearance of potential neurotoxic compounds via the BBB, and preventing BBB dysfunction. Finally, a need for novel biomarkers of BBB dysfunction is addressed.
Collapse
Affiliation(s)
- Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor MI, USA
| | | | | | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor MI, USA
| |
Collapse
|
14
|
Morris JC, Weiner M, Xiong C, Beckett L, Coble D, Saito N, Aisen PS, Allegri R, Benzinger TLS, Berman SB, Cairns NJ, Carrillo MC, Chui HC, Chhatwal JP, Cruchaga C, Fagan AM, Farlow M, Fox NC, Ghetti B, Goate AM, Gordon BA, Graff-Radford N, Day GS, Hassenstab J, Ikeuchi T, Jack CR, Jagust WJ, Jucker M, Levin J, Massoumzadeh P, Masters CL, Martins R, McDade E, Mori H, Noble JM, Petersen RC, Ringman JM, Salloway S, Saykin AJ, Schofield PR, Shaw LM, Toga AW, Trojanowski JQ, Vöglein J, Weninger S, Bateman RJ, Buckles VD. Autosomal dominant and sporadic late onset Alzheimer's disease share a common in vivo pathophysiology. Brain 2022; 145:3594-3607. [PMID: 35580594 PMCID: PMC9989348 DOI: 10.1093/brain/awac181] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
The extent to which the pathophysiology of autosomal dominant Alzheimer's disease corresponds to the pathophysiology of 'sporadic' late onset Alzheimer's disease is unknown, thus limiting the extrapolation of study findings and clinical trial results in autosomal dominant Alzheimer's disease to late onset Alzheimer's disease. We compared brain MRI and amyloid PET data, as well as CSF concentrations of amyloid-β42, amyloid-β40, tau and tau phosphorylated at position 181, in 292 carriers of pathogenic variants for Alzheimer's disease from the Dominantly Inherited Alzheimer Network, with corresponding data from 559 participants from the Alzheimer's Disease Neuroimaging Initiative. Imaging data and CSF samples were reprocessed as appropriate to guarantee uniform pipelines and assays. Data analyses yielded rates of change before and after symptomatic onset of Alzheimer's disease, allowing the alignment of the ∼30-year age difference between the cohorts on a clinically meaningful anchor point, namely the participant age at symptomatic onset. Biomarker profiles were similar for both autosomal dominant Alzheimer's disease and late onset Alzheimer's disease. Both groups demonstrated accelerated rates of decline in cognitive performance and in regional brain volume loss after symptomatic onset. Although amyloid burden accumulation as determined by PET was greater after symptomatic onset in autosomal dominant Alzheimer's disease than in late onset Alzheimer's disease participants, CSF assays of amyloid-β42, amyloid-β40, tau and p-tau181 were largely overlapping in both groups. Rates of change in cognitive performance and hippocampal volume loss after symptomatic onset were more aggressive for autosomal dominant Alzheimer's disease participants. These findings suggest a similar pathophysiology of autosomal dominant Alzheimer's disease and late onset Alzheimer's disease, supporting a shared pathobiological construct.
Collapse
Affiliation(s)
- John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Weiner
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Laurel Beckett
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Dean Coble
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Naomi Saito
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Paul S Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nigel J Cairns
- College of Medicine and Health and the Living Systems Institute, University of Exeter, Exeter, UK
| | | | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Martin Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nick C Fox
- Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Institute of Neurology, London, UK
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Mathias Jucker
- Cell Biology of Neurological Diseases Group, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Johannes Levin
- DZNE Munich, Munich Cluster of Systems Neurology (SyNergy) and Ludwig-Maximilians-Universität, Munich, Germany
| | - Parinaz Massoumzadeh
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- Florey Institute, University of Melbourne, Melbourne, Australia
| | - Ralph Martins
- Sir James McCusker Alzheimer’s Disease Research Unit, Edith Cowan University, Nedlands, Australia
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hiroshi Mori
- Department of Neuroscience, Osaka City University Medical School, Osaka City, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | | | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Salloway
- Department of Neurology, Butler Hospital and Alpert Medical School of Brown University, Providence, RI, 02906, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter R Schofield
- Neuroscience Research Australia and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE) and Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Virginia D Buckles
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
15
|
Jiang L, Qin Y, Zhao YW, Zeng Q, Pan HX, Liu ZH, Sun QY, Xu Q, Tan JQ, Yan XX, Li JC, Tang BS, Guo JF. PSEN1 G417S mutation in a Chinese pedigree causing early-onset parkinsonism with cognitive impairment. Neurobiol Aging 2022; 115:70-76. [DOI: 10.1016/j.neurobiolaging.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/03/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022]
|
16
|
Vemuri P, Decarli CS, Duering M. Imaging Markers of Vascular Brain Health: Quantification, Clinical Implications, and Future Directions. Stroke 2022; 53:416-426. [PMID: 35000423 PMCID: PMC8830603 DOI: 10.1161/strokeaha.120.032611] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cerebrovascular disease (CVD) manifests through a broad spectrum of mechanisms that negatively impact brain and cognitive health. Oftentimes, CVD changes (excluding acute stroke) are insufficiently considered in aging and dementia studies which can lead to an incomplete picture of the etiologies contributing to the burden of cognitive impairment. Our goal with this focused review is 3-fold. First, we provide a research update on the current magnetic resonance imaging methods that can measure CVD lesions as well as early CVD-related brain injury specifically related to small vessel disease. Second, we discuss the clinical implications and relevance of these CVD imaging markers for cognitive decline, incident dementia, and disease progression in Alzheimer disease, and Alzheimer-related dementias. Finally, we present our perspective on the outlook and challenges that remain in the field. With the increased research interest in this area, we believe that reliable CVD imaging biomarkers for aging and dementia studies are on the horizon.
Collapse
Affiliation(s)
| | - Charles S. Decarli
- Departments of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, California, USA
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany
- Medical Image Analysis Center (MIAC AG) and qbig, Department of Biomedical Engineering, University of Basel, Switzerland
| |
Collapse
|
17
|
Eger SJ, Le Guen Y, Khan RR, Hall JN, Kennedy G, Zaharchuk G, Couthouis J, Brooks WS, Velakoulis D, Napolioni V, Belloy ME, Dalgard CL, Mormino EC, Gitler AD, Greicius MD. Confirming Pathogenicity of the F386L PSEN1 Variant in a South Asian Family With Early-Onset Alzheimer Disease. Neurol Genet 2021; 8:e647. [PMID: 34901437 PMCID: PMC8655848 DOI: 10.1212/nxg.0000000000000647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/20/2021] [Indexed: 11/15/2022]
Abstract
Objectives The F386L PSEN1 variant has been reported in 1 Japanese family with limited clinical information. We aimed to prove that F386L is pathogenic by demonstrating that it segregates with early-onset Alzheimer disease (AD). Methods Eight individuals in a South Asian family provided DNA for genetic testing and underwent a neurologic examination. Results The female proband was diagnosed with AD at age 45 years and died at age 49 years. She had a CSF biomarker profile consistent with AD, and her florbetaben PET scan was amyloid positive with high uptake in the striatum. Her MRI showed no prominent white matter disease. Her affected relatives had an age at onset range of 38–57 years and had imaging and biomarker profiles similar to hers. Discussion The results presented here, in conjunction with the prior report, confirm the pathogenicity of F386L. Furthermore, our study highlights the importance of studying families from underrepresented populations to identify or confirm the pathogenicity of rare variants that may be specific to certain genetic ancestries.
Collapse
Affiliation(s)
- Sarah J Eger
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Raiyan R Khan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Jacob N Hall
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Gabriel Kennedy
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Greg Zaharchuk
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Julien Couthouis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - William S Brooks
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Dennis Velakoulis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Valerio Napolioni
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Michaël E Belloy
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Clifton L Dalgard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Aaron D Gitler
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, (S.J.E., Y.L.G., G.K., M.E.B., E.C.M., M.D.G.); Department of Computer Science, Columbia University, New York, NY (R.R.K.) the Neurology Center of Southern California, Temecula, CA (J.N.H.); Department of Radiology, Stanford University School of Medicine, Stanford, CA (G.Z.) Department of Genetics, Stanford University School of Medicine, Stanford, CA (J.C., A.D.G.); Neuroscience Research Australia, Randwick NSW 2031, Australia (W.S.B); the University of New South Wales, Sydney NSW 2052, Australia (W.S.B.); Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville VIC 3050, Australia (D.V.); School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy (V.N); Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.); the American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD (C.L.D.)
| |
Collapse
|
18
|
O'Connor A, Karikari TK, Poole T, Ashton NJ, Lantero Rodriguez J, Khatun A, Swift I, Heslegrave AJ, Abel E, Chung E, Weston PSJ, Pavisic IM, Ryan NS, Barker S, Rossor MN, Polke JM, Frost C, Mead S, Blennow K, Zetterberg H, Fox NC. Plasma phospho-tau181 in presymptomatic and symptomatic familial Alzheimer's disease: a longitudinal cohort study. Mol Psychiatry 2021; 26:5967-5976. [PMID: 32665603 PMCID: PMC7612227 DOI: 10.1038/s41380-020-0838-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 01/05/2023]
Abstract
Blood biomarkers have great potential to advance clinical care and accelerate trials in Alzheimer's disease (AD). Plasma phospho-tau181 (p-tau181) is a promising blood biomarker however, it is unknown if levels increase in presymptomatic AD. Therefore, we investigated the timing of p-tau181 changes using 153 blood samples from 70 individuals in a longitudinal study of familial AD (FAD). Plasma p-tau181 was measured, using an in-house single molecule array assay. We compared p-tau181 between symptomatic carriers, presymptomatic carriers, and non-carriers, adjusting for age and sex. We examined the relationship between p-tau181 and neurofilament light and estimated years to/from symptom onset (EYO), as well as years to/from actual onset in a symptomatic subgroup. In addition, we studied associations between p-tau181 and clinical severity, as well testing for differences between genetic subgroups. Twenty-four were presymptomatic carriers (mean baseline EYO -9.6 years) while 27 were non-carriers. Compared with non-carriers, plasma p-tau181 concentration was higher in both symptomatic (p < 0.001) and presymptomatic mutation carriers (p < 0.001). Plasma p-tau181 showed considerable intra-individual variability but individual values discriminated symptomatic (AUC 0.93 [95% CI 0.85-0.98]) and presymptomatic (EYO ≥ -7 years) (AUC 0.86 [95% CI 0.72-0.94]) carriers from non-carriers of the same age and sex. From a fitted model there was evidence (p = 0.050) that p-tau181 concentrations were higher in mutation carriers than non-carriers from 16 years prior to estimated symptom onset. Our finding that plasma p-tau181 concentration is increased in symptomatic and presymptomatic FAD suggests potential utility as an easily accessible biomarker of AD pathology.
Collapse
Affiliation(s)
- Antoinette O'Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Teresa Poole
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia, South London & Maudsley NHS Foundation, London, UK
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ayesha Khatun
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Imogen Swift
- UK Dementia Research Institute at UCL, London, UK
| | | | - Emily Abel
- UK Dementia Research Institute at UCL, London, UK
| | - Elisha Chung
- UK Dementia Research Institute at UCL, London, UK
| | - Philip S J Weston
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Ivanna M Pavisic
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Suzie Barker
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Martin N Rossor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - James M Polke
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon Mead
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK.
- UK Dementia Research Institute at UCL, London, UK.
| |
Collapse
|
19
|
Mao C, Li J, Dong L, Huang X, Lei D, Wang J, Chu S, Liu C, Peng B, Román GC, Cui L, Gao J. Clinical Phenotype and Mutation Spectrum of Alzheimer's Disease with Causative Genetic Mutation in a Chinese Cohort. Curr Alzheimer Res 2021; 18:265-272. [PMID: 34102969 PMCID: PMC8506917 DOI: 10.2174/1567205018666210608120339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/06/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Background Alzheimer’s disease with a causative genetic mutation (AD-CGM) is an uncommon form, characterized by a heterogeneous clinical phenotype and variations in the genotype of racial groups affected. Objective We aimed to systemically describe the phenotype variance and mutation spectrum in the large sample size of the Peking Union Medical College Hospital (PUMCH) cohort, Beijing, China. Methods Next-generation sequencing (NGS) was carried out in 1108 patients diagnosed with dementia. A total of 40 Han Chinese patients with three AD gene mutations were enrolled. A systemic review of all the patients was performed, including clinical history, neurocognitive assessment, brain magnetic resonance imaging, and cerebrospinal fluid (CSF) biomarkers. Results We studied the following gene mutation variants: 12 AβPP, 13 PSEN1, and 9 PSEN2, and 23 among them were novel. Most of them were early-onset, but PSEN1 mutation carriers had the youngest onset age. The commonest symptoms were similar to those of AD, including an amnestic syndrome, followed by psychiatric symptoms and movement disorder. On MRI, parietal and posterior temporal atrophy was prominent in PSEN1 and PSEN2 mutation carriers, while AβPP mutation carriers had more vascular changes. The CSF biomarkers profile was indistinguishable from sporadic AD. Conclusion We identified a small group of AD-CGM subjects representing 3.6% among more than 1000 demented patients in the PUMCH cohort. These subjects usually presented with early-onset
dementia and exhibited significant clinical and genetic heterogeneity. Identification required complete screening of genetic mutations using NGS. Although family history was usually present, we found non-familial cases of all three genetic mutations.
Collapse
Affiliation(s)
- Chenhui Mao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jie Li
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Liling Dong
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Xinying Huang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Dan Lei
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jie Wang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Shanshan Chu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Caiyan Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Bin Peng
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Gustavo C Román
- Stanley H. Appel Department of Neurology, Nantz National Alzheimer Center, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
20
|
Willumsen N, Poole T, Nicholas JM, Fox NC, Ryan NS, Lashley T. Variability in the type and layer distribution of cortical Aβ pathology in familial Alzheimer's disease. Brain Pathol 2021; 32:e13009. [PMID: 34319632 PMCID: PMC9048809 DOI: 10.1111/bpa.13009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/30/2022] Open
Abstract
Familial Alzheimer's disease (FAD) is caused by autosomal dominant mutations in the PSEN1, PSEN2 or APP genes, giving rise to considerable clinical and pathological heterogeneity in FAD. Here we investigate variability in clinical data and the type and distribution of Aβ pathologies throughout the cortical layers of different FAD mutation cases. Brain tissue from 20 FAD cases [PSEN1 pre-codon 200 (n = 10), PSEN1 post-codon 200 (n = 6), APP (n = 4)] were investigated. Frontal cortex sections were stained immunohistochemically for Aβ, and Nissl to define the cortical layers. The frequency of different amyloid-beta plaque types was graded for each cortical layer and the severity of cerebral amyloid angiopathy (CAA) was determined in cortical and leptomeningeal blood vessels. Comparisons were made between FAD mutations and APOE4 status, with associations between pathology, clinical and genetic data investigated. In this cohort, possession of an APOE4 allele was associated with increased disease duration but not with age at onset, after adjusting for mutation sub-group and sex. We found Aβ pathology to be heterogeneous between cases although Aβ load was highest in cortical layer 3 for all mutation groups and a higher Aβ load was associated with APOE4. The PSEN1 post-codon 200 group had a higher Aβ load in lower cortical layers, with a small number of this group having increased cotton wool plaque pathology in lower layers. Cotton wool plaque frequency was positively associated with the severity of CAA in the whole cohort and in the PSEN1 post-codon 200 group. Carriers of the same PSEN1 mutation can have differing patterns of Aβ deposition, potentially because of differences in risk factors. Our results highlight possible influences of APOE4 genotype, and PSEN1 mutation type on Aβ deposition, which may have effects on the clinical heterogeneity of FAD.
Collapse
Affiliation(s)
- Nanet Willumsen
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Teresa Poole
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK.,Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Jennifer M Nicholas
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK.,Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at University College London, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at University College London, London, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
21
|
Chen Y, Wang X, Guan L, Wang Y. Role of White Matter Hyperintensities and Related Risk Factors in Vascular Cognitive Impairment: A Review. Biomolecules 2021; 11:biom11081102. [PMID: 34439769 PMCID: PMC8391787 DOI: 10.3390/biom11081102] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
White matter hyperintensities (WMHs) of presumed vascular origin are one of the imaging markers of cerebral small-vessel disease, which is prevalent in older individuals and closely associated with the occurrence and development of cognitive impairment. The heterogeneous nature of the imaging manifestations of WMHs creates difficulties for early detection and diagnosis of vascular cognitive impairment (VCI) associated with WMHs. Because the underlying pathological processes and biomarkers of WMHs and their development in cognitive impairment remain uncertain, progress in prevention and treatment is lagging. For this reason, this paper reviews the status of research on the features of WMHs related to VCI, as well as mediators associated with both WMHs and VCI, and summarizes potential treatment strategies for the prevention and intervention in WMHs associated with VCI.
Collapse
Affiliation(s)
- Yiyi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Xing Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Department of Neurology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400000, China
| | - Ling Guan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Correspondence: (L.G.); (Y.W.)
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Correspondence: (L.G.); (Y.W.)
| |
Collapse
|
22
|
Schoemaker D, Velilla-Jimenez L, Zuluaga Y, Baena A, Ospina C, Bocanegra Y, Alvarez S, Ochoa-Escudero M, Guzmán-Vélez E, Martinez J, Lopera F, Arboleda-Velasquez JF, Quiroz YT. Global Cardiovascular Risk Profile and Cerebrovascular Abnormalities in Presymptomatic Individuals with CADASIL or Autosomal Dominant Alzheimer's Disease. J Alzheimers Dis 2021; 82:841-853. [PMID: 34092645 DOI: 10.3233/jad-210313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cardiovascular risk factors increase the risk of developing dementia, including Alzheimer's disease and vascular dementia. OBJECTIVE Studying individuals with autosomal dominant mutations leading to the early onset of dementia, this study examines the effect of the global cardiovascular risk profile on early cognitive and neuroimaging features of Alzheimer's disease and vascular dementia. METHODS We studied 85 non-demented and stroke-free individuals, including 20 subjects with Presenilin1 (PSEN1) E280A mutation leading to the early onset of autosomal dominant Alzheimer's disease (ADAD), 20 subjects with NOTCH3 mutations leading to cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and to the early onset of vascular dementia, and 45 non-affected family members (non-carriers). All subjects underwent clinical and neuropsychological evaluations and an MRI. The global cardiovascular risk profile was estimated using the office-based Framingham Cardiovascular Risk Profile (FCRP) score. RESULTS In individuals with CADASIL, a higher FCRP score was associated with a reduced hippocampal volume (B = -0.06, p < 0.05) and an increased severity of cerebral microbleeds (B = 0.13, p < 0.001), lacunes (B = 0.30, p < 0.001), and perivascular space enlargement in the basal ganglia (B = 0.50, p < 0.05). There was no significant association between the FCRP score and neuroimaging measures in ADAD or non-carrier subjects. While the FCRP score was related to performance in executive function in non-carrier subjects (B = 0.06, p < 0.05), it was not significantly associated with cognitive performance in individuals with CADASIL or ADAD. CONCLUSION Our results suggest that individuals with CADASIL and other forms of vascular cognitive impairment might particularly benefit from early interventions aimed at controlling cardiovascular risks.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | - Yesica Zuluaga
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Ana Baena
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Carolina Ospina
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Yamile Bocanegra
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Sergio Alvarez
- Department of Radiology, Hospital Pablo Tobon Uribe, Medellín, Colombia
| | | | - Edmarie Guzmán-Vélez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jairo Martinez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco Lopera
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Joseph F Arboleda-Velasquez
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Tortelli R, Seripa D, Zecca C, Dell’Abate MT, Bisceglia P, Barulli MR, De Blasi R, Logroscino G. A New Presenilin 1 (Psen1) Mutation (p.Cys263Trp) as a Cause of Both Early and Late-Onset Alzheimer's Disease in a Large Italian Family. Int J Mol Sci 2021; 22:ijms22126215. [PMID: 34207526 PMCID: PMC8230263 DOI: 10.3390/ijms22126215] [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: 05/13/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
Mutations in the PSEN1 gene are the most common cause of autosomal dominant Alzheimer's disease, and are characterized by a high phenotype variability. This study describes a five-generation family, with a prevalent late-onset of the disease and a high frequency of depression, in which a new missense mutation (c.789T > G, p.Cys263Trp) in exon 8 of the PSEN1 gene was found. Only the proband presented an early onset at the age of 45 with attention deficit, followed by spatial disorientation, psychiatric symptoms and parkinsonian signs. The other two cases had a late onset of the disease and a typical presentation with memory loss. Both were characterized by a high level of anxiety and depression. The disease course was different with signs of Lewy body dementia for the proband's mother, and pyramidal involvement and a shorter disease duration for the proband's maternal aunt. The other eight cases with late-onset dementia and three cases with a long history of depression have been reported in the family pedigree, underlying the high phenotype variability of PSEN1 mutations.
Collapse
Affiliation(s)
- Rosanna Tortelli
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
- Correspondence: (R.T.); (G.L.); Tel.: +39-0833773904 (R.T. & G.L.); Fax: +39-08831830670 (R.T. & G.L.)
| | - Davide Seripa
- Complex Unit of Geriatrics, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.S.); (P.B.)
- Laboratory for Advanced Hematological Diagnostics, Department of Hematology and Stem Cell Transplant, “Vito Fazzi” Hospital, 73100 Lecce, Italy
| | - Chiara Zecca
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Maria Teresa Dell’Abate
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Paola Bisceglia
- Complex Unit of Geriatrics, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.S.); (P.B.)
| | - Maria Rosaria Barulli
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Roberto De Blasi
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
- Department of Radiology, “Pia Fondazione Cardinale G. Panico”, 73039 Tricase, Italy
| | - Giancarlo Logroscino
- Department of Radiology, “Pia Fondazione Cardinale G. Panico”, 73039 Tricase, Italy
- Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
- Correspondence: (R.T.); (G.L.); Tel.: +39-0833773904 (R.T. & G.L.); Fax: +39-08831830670 (R.T. & G.L.)
| |
Collapse
|
24
|
The Role of White Matter Dysfunction and Leukoencephalopathy/Leukodystrophy Genes in the Aetiology of Frontotemporal Dementias: Implications for Novel Approaches to Therapeutics. Int J Mol Sci 2021; 22:ijms22052541. [PMID: 33802612 PMCID: PMC7961524 DOI: 10.3390/ijms22052541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 01/01/2023] Open
Abstract
Frontotemporal dementia (FTD) is a common cause of presenile dementia and is characterized by behavioural and/or language changes and progressive cognitive deficits. Genetics is an important component in the aetiology of FTD, with positive family history of dementia reported for 40% of cases. This review synthesizes current knowledge of the known major FTD genes, including C9orf72 (chromosome 9 open reading frame 72), MAPT (microtubule-associated protein tau) and GRN (granulin), and their impact on neuronal and glial pathology. Further, evidence for white matter dysfunction in the aetiology of FTD and the clinical, neuroimaging and genetic overlap between FTD and leukodystrophy/leukoencephalopathy are discussed. The review highlights the role of common variants and mutations in genes such as CSF1R (colony-stimulating factor 1 receptor), CYP27A1 (cytochrome P450 family 27 subfamily A member 1), TREM2 (triggering receptor expressed on myeloid cells 2) and TMEM106B (transmembrane protein 106B) that play an integral role in microglia and oligodendrocyte function. Finally, pharmacological and non-pharmacological approaches for enhancing remyelination are discussed in terms of future treatments of FTD.
Collapse
|
25
|
White matter hyperintensities and patterns of atrophy in early onset Alzheimer's disease with causative gene mutations. Clin Neurol Neurosurg 2021; 203:106552. [PMID: 33601235 DOI: 10.1016/j.clineuro.2021.106552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/25/2021] [Accepted: 02/06/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE White matter hyperintensities could be found in many degenerative dementias including Alzheimer's disease (AD). Pathogenesis of white matter hyperintensities in AD is complicated. We aim to identify the features of white matter hyperintensities and the atrophy pattern in early onset Alzheimer's disease with causative gene mutations. METHODS 7 AD dementia patients with causative mutations were included and the clinical history, neuropsychology, neuroimaging,APOE genotype and whole-genome sequencing (WGS) were analyzed. Axial T1-weighted images and Fluid attenuated inversion recovery (FLAIR) were analyzed with visual rating scale to examine cortical atrophy and white matter hyperintensities. RESULTS 5 female and 2 male patients with 4PSEN1, 2PSEN2 and 1APP mutation were included. The average age of onset was 46.7y/o (44-52) and the duration of disease was 28.6 months (8-60). Clinical phenotype included memory loss (100 %), visual/spatial disorder (100 %), executive dysfunction (100 %), calculation disorder (85.7 %), disorientation (85.7 %), language problem (57.1 %), personality change (28.6 %) and movement disorder (14.3 %). The grading of posterior cortex atrophy was higher than medial temporal lobe atrophy. Periventricular hyperintensities surrounding occipital and frontal horn of ventricle and sub-ventricular bands were most common, while small foci of lesions were also detected in deep white matter, sub-cortical and juxta-cortical area. Mutations carriers in the APP gene or PSEN1 gene postcodon 200 had more severe white matter hyperintensities than other mutations. CONCLUSION White matter hyperintensities were found in early onset AD with causative mutations. The severity was related to genotypes and spatial distributions. Axon degeneration following neuronal loss and ischemic injury might be the pathogenesis of white matter damage. Severer atrophy in the posterior cortex than medial temporal lobe can present in early onset AD.
Collapse
|
26
|
Koriath CAM, Kenny J, Ryan NS, Rohrer JD, Schott JM, Houlden H, Fox NC, Tabrizi SJ, Mead S. Genetic testing in dementia - utility and clinical strategies. Nat Rev Neurol 2021; 17:23-36. [PMID: 33168964 DOI: 10.1038/s41582-020-00416-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2020] [Indexed: 02/07/2023]
Abstract
Techniques for clinical genetic testing in dementia disorders have advanced rapidly but remain to be more widely implemented in practice. A positive genetic test offers a precise molecular diagnosis, can help members of an affected family to determine personal risk, provides a basis for reproductive choices and can offer options for clinical trials. The likelihood of identifying a specific genetic cause of dementia depends on the clinical condition, the age at onset and family history. Attempts to match phenotypes to single genes are mostly inadvisable owing to clinical overlap between the dementias, genetic heterogeneity, pleiotropy and concurrent mutations. Currently, the appropriate genetic test in most cases of dementia is a next-generation sequencing gene panel, though some conditions necessitate specific types of test such as repeat expansion testing. Whole-exome and whole-genome sequencing are becoming financially feasible but raise or exacerbate complex issues such as variants of uncertain significance, secondary findings and the potential for re-analysis in light of new information. However, the capacity for data analysis and counselling is already restricting the provision of genetic testing. Patients and their relatives need to be given reliable information to enable them to make informed choices about tests, treatments and data sharing; the ability of patients with dementia to make decisions must be considered when providing this information.
Collapse
Affiliation(s)
| | - Joanna Kenny
- South West Thames Regional Genetics Service, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henry Houlden
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK.
| |
Collapse
|
27
|
Schaeffer MJ, Chan L, Barber PA. The neuroimaging of neurodegenerative and vascular disease in the secondary prevention of cognitive decline. Neural Regen Res 2021; 16:1490-1499. [PMID: 33433462 PMCID: PMC8323688 DOI: 10.4103/1673-5374.303011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Structural brain changes indicative of dementia occur up to 20 years before the onset of clinical symptoms. Efforts to modify the disease process after the onset of cognitive symptoms have been unsuccessful in recent years. Thus, future trials must begin during the preclinical phases of the disease before symptom onset. Age related cognitive decline is often the result of two coexisting brain pathologies: Alzheimer’s disease (amyloid, tau, and neurodegeneration) and vascular disease. This review article highlights some of the common neuroimaging techniques used to visualize the accumulation of neurodegenerative and vascular pathologies during the preclinical stages of dementia such as structural magnetic resonance imaging, positron emission tomography, and white matter hyperintensities. We also describe some emerging neuroimaging techniques such as arterial spin labeling, diffusion tensor imaging, and quantitative susceptibility mapping. Recent literature suggests that structural imaging may be the most sensitive and cost-effective marker to detect cognitive decline, while molecular positron emission tomography is primarily useful for detecting disease specific pathology later in the disease process. Currently, the presence of vascular disease on magnetic resonance imaging provides a potential target for optimizing vascular risk reduction strategies, and the presence of vascular disease may be useful when combined with molecular and metabolic markers of neurodegeneration for identifying the risk of cognitive impairment.
Collapse
Affiliation(s)
- Morgan J Schaeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Leona Chan
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Philip A Barber
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
28
|
Pavisic IM, Nicholas JM, O'Connor A, Rice H, Lu K, Fox NC, Ryan NS. Disease duration in autosomal dominant familial Alzheimer disease: A survival analysis. NEUROLOGY-GENETICS 2020; 6:e507. [PMID: 33225064 PMCID: PMC7673285 DOI: 10.1212/nxg.0000000000000507] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/21/2020] [Indexed: 11/15/2022]
Abstract
Objective To use survival modeling to estimate disease duration in autosomal dominant familial Alzheimer disease (ADAD) and ascertain whether factors influencing age at onset also affect survival. Methods Symptomatic mutation carriers (201 presenilin 1 [PSEN1] and 55 amyloid precursor protein [APP]) from ADAD families referred to the Dementia Research Centre, between 1987 and 2019, were included. Survival was assessed with respect to age at onset, year of birth, APOE ε4 status, cognitive presentation, and sex using multilevel mixed-effects Weibull survival models. The contribution of mutation and family to variance in age at onset and duration was also assessed. Results Estimated mean survival was 11.6 (10.4–12.9) years and was similar for APP and PSEN1 mutations. Sixty-seven percent of the variance in age at onset was explained by mutation and 72% by mutation and family together. In contrast, only 6% of the variance in disease duration was explained by mutation specificity and 18% by family membership. Irrespective of gene, survival appeared longer for successive generations and in individuals with atypical presentations. Older age at onset was associated with longer duration within PSEN1 and shorter duration within APP mutation carriers. No differences in survival time were found between sexes or between mutations located before or beyond codon 200 within PSEN1. Conclusions Survival is influenced by mutation to a much lesser extent than age at onset. Survival time has increased over time and is longer in atypical presentations. These insights may inform the interpretation of disease-modifying therapy trials in ADAD.
Collapse
Affiliation(s)
- Ivanna M Pavisic
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Jennifer M Nicholas
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Antoinette O'Connor
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Helen Rice
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Kirsty Lu
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Nick C Fox
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| | - Natalie S Ryan
- Department of Neurodegenerative Diseases (I.M.P., J.M.N., A.O., H.R., K.L., N.C.F., N.S.R.), Dementia Research Centre, UCL Queen Square Institute of Neurology, London; UK Dementia Research Institute at University College London (I.M.P., A.O., H.R., N.C.F., N.S.R.); and Department of Medial Statistics (J.M.N.), London School of Hygiene and Tropical Medicine, United Kingdom
| |
Collapse
|
29
|
Zhang Q, Wu L, Du C, Xu K, Sun J, Zhang J, Li H, Li X. Effects of an APOE Promoter Polymorphism on Fronto-Parietal Functional Connectivity During Nondemented Aging. Front Aging Neurosci 2020; 12:183. [PMID: 32694990 PMCID: PMC7338603 DOI: 10.3389/fnagi.2020.00183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/26/2020] [Indexed: 01/03/2023] Open
Abstract
Background: The rs405509 polymorphism ofthe apolipoprotein E (APOE) promoter is related to Alzheimer'sdisease (AD). The T/T allele of rs405509 is known to decrease the transcription of the APOE gene and lead to impairments in specific brain structural networks with aging; thus, it is an important risk factor for AD. However, it remains unknown whether rs405509 affects brain functional connectivity (FC) in aging. Methods: We investigated the effect of the rs405509 genotype (T/T vs. G-allele) on age-related brain FC using functional magnetic resonance imaging. Forty-five elderly TT carriers and 45 elderly G-allele carriers were scanned during a working memory (WM) task. Results: We found that TT carriers showed an accelerated age-related increase in functional activation in the left postcentral gyrus compared with G-allele carriers. Furthermore, the FC between the left postcentral gyrus and some key regions during WM performance, including the right caudal and superior frontal sulcus (SFS), was differentially modulated by age across rs405509 genotype groups. Conclusions: These results demonstrate that the rs405509 T/T allele of APOE causes an age-related brain functional decline in nondemented elderly people, which may be beneficial for understanding the neural mechanisms of rs405509-related cognitive aging and AD pathogenesis.
Collapse
Affiliation(s)
- Qirui Zhang
- Institute of Criminology, People’s Public Security University of China, Beijing, China
| | - Lingli Wu
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- BABRI Centre, Beijing Normal University, Beijing, China
| | - Chao Du
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- BABRI Centre, Beijing Normal University, Beijing, China
| | - Kai Xu
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- BABRI Centre, Beijing Normal University, Beijing, China
| | - Jinping Sun
- The Affiliated Hospital of Qingdao University, Shandong, China
| | - Junying Zhang
- BABRI Centre, Beijing Normal University, Beijing, China
| | - He Li
- BABRI Centre, Beijing Normal University, Beijing, China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- BABRI Centre, Beijing Normal University, Beijing, China
| |
Collapse
|
30
|
Castillo-Barnes D, Su L, Ramírez J, Salas-Gonzalez D, Martinez-Murcia FJ, Illan IA, Segovia F, Ortiz A, Cruchaga C, Farlow MR, Xiong C, Graff-Radford NR, Schofield PR, Masters CL, Salloway S, Jucker M, Mori H, Levin J, Gorriz JM. Autosomal Dominantly Inherited Alzheimer Disease: Analysis of genetic subgroups by Machine Learning. AN INTERNATIONAL JOURNAL ON INFORMATION FUSION 2020; 58:153-167. [PMID: 32284705 PMCID: PMC7153760 DOI: 10.1016/j.inffus.2020.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Despite subjects with Dominantly-Inherited Alzheimer's Disease (DIAD) represent less than 1% of all Alzheimer's Disease (AD) cases, the Dominantly Inherited Alzheimer Network (DIAN) initiative constitutes a strong impact in the understanding of AD disease course with special emphasis on the presyptomatic disease phase. Until now, the 3 genes involved in DIAD pathogenesis (PSEN1, PSEN2 and APP) have been commonly merged into one group (Mutation Carriers, MC) and studied using conventional statistical analysis. Comparisons between groups using null-hypothesis testing or longitudinal regression procedures, such as the linear-mixed-effects models, have been assessed in the extant literature. Within this context, the work presented here performs a comparison between different groups of subjects by considering the 3 genes, either jointly or separately, and using tools based on Machine Learning (ML). This involves a feature selection step which makes use of ANOVA followed by Principal Component Analysis (PCA) to determine which features would be realiable for further comparison purposes. Then, the selected predictors are classified using a Support-Vector-Machine (SVM) in a nested k-Fold cross-validation resulting in maximum classification rates of 72-74% using PiB PET features, specially when comparing asymptomatic Non-Carriers (NC) subjects with asymptomatic PSEN1 Mutation-Carriers (PSEN1-MC). Results obtained from these experiments led to the idea that PSEN1-MC might be considered as a mixture of two different subgroups including: a first group whose patterns were very close to NC subjects, and a second group much more different in terms of imaging patterns. Thus, using a k-Means clustering algorithm it was determined both subgroups and a new classification scenario was conducted to validate this process. The comparison between each subgroup vs. NC subjects resulted in classification rates around 80% underscoring the importance of considering DIAN as an heterogeneous entity.
Collapse
Affiliation(s)
- Diego Castillo-Barnes
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
| | - Li Su
- Department of Psychiatry, University of Cambridge, Cambridge (UK)
| | - Javier Ramírez
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
| | - Diego Salas-Gonzalez
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
| | | | - Ignacio A. Illan
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
| | - Fermin Segovia
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
| | - Andres Ortiz
- Department of Communications Engineering, University of Malaga, Malaga (Spain)
| | - Carlos Cruchaga
- Department of Psychiatry and Neurology, Washington University School of Medicine, St. Louis, Missouri (USA)
| | - Martin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana (USA)
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri (USA)
| | | | - Peter R. Schofield
- Neuroscience Research Australia and School of Medical Sciences, University of New South Wales, Sydney (Australia)
| | - Colin L. Masters
- Florey Institute and University of Melbourne, Victoria (Australia)
| | | | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen (Germany)
| | - Hiroshi Mori
- Department of Clinical Neuroscience, Osaka City University Medical school, Osaka (Japan)
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University of Munich, Munich (Germany)
| | - Juan M. Gorriz
- Department of Signal Theory, Telematics and Communications, University of Granada, Granada (Spain)
- Department of Psychiatry, University of Cambridge, Cambridge (UK)
| | | |
Collapse
|
31
|
The R278I Mutation of PSEN1 in the Familial Alzheimer Disease. Dement Neurocogn Disord 2020; 19:33-35. [PMID: 32174048 PMCID: PMC7105715 DOI: 10.12779/dnd.2020.19.1.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/17/2019] [Accepted: 11/22/2019] [Indexed: 11/29/2022] Open
|
32
|
Walsh P, Sudre CH, Fiford CM, Ryan NS, Lashley T, Frost C, Barnes J. CSF amyloid is a consistent predictor of white matter hyperintensities across the disease course from aging to Alzheimer's disease. Neurobiol Aging 2020; 91:5-14. [PMID: 32305782 DOI: 10.1016/j.neurobiolaging.2020.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 01/06/2023]
Abstract
This study investigated the relationship between white matter hyperintensities (WMH) and cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers. Subjects included 180 controls, 107 individuals with a significant memory concern, 320 individuals with early mild cognitive impairment, 171 individuals with late mild cognitive impairment, and 151 individuals with AD, with 3T MRI and CSF Aβ1-42, total tau (t-tau), and phosphorylated tau (p-tau) data. Multiple linear regression models assessed the relationship between WMH and CSF Aβ1-42, t-tau, and p-tau. Directionally, a higher WMH burden was associated with lower CSF Aβ1-42 within each diagnostic group, with no evidence for a difference in the slope of the association across diagnostic groups (p = 0.4). Pooling all participants, this association was statistically significant after adjustment for t-tau, p-tau, age, diagnostic group, and APOE-ε4 status (p < 0.001). Age was the strongest predictor of WMH (partial R2~16%) compared with CSF Aβ1-42 (partial R2~5%). There was no evidence for an association with WMH and either t-tau or p-tau. These data are supportive of a link between amyloid burden and presumed vascular pathology.
Collapse
Affiliation(s)
- Phoebe Walsh
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| | - Carole H Sudre
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; Centre for Medical Image Computing, University College London, London, UK
| | - Cassidy M Fiford
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Chris Frost
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Josephine Barnes
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | |
Collapse
|
33
|
Santos-Mandujano RA, Ryan NS, Chávez-Gutiérrez L, Sánchez-Torres C, Meraz-Ríos MA. Clinical Association of White Matter Hyperintensities Localization in a Mexican Family with Spastic Paraparesis Carrying the PSEN1 A431E Mutation. J Alzheimers Dis 2020; 73:1075-1083. [DOI: 10.3233/jad-190978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Natalie S. Ryan
- Dementia Research Centre, Department of Neurodegenerative Diseases, UCL Institute of Neurology, London, UK
| | - Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Carmen Sánchez-Torres
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV), CDMX, México
| | - Marco Antonio Meraz-Ríos
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV), CDMX, México
| |
Collapse
|
34
|
Greenberg SM, Bacskai BJ, Hernandez-Guillamon M, Pruzin J, Sperling R, van Veluw SJ. Cerebral amyloid angiopathy and Alzheimer disease - one peptide, two pathways. Nat Rev Neurol 2020; 16:30-42. [PMID: 31827267 PMCID: PMC7268202 DOI: 10.1038/s41582-019-0281-2] [Citation(s) in RCA: 510] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2019] [Indexed: 12/22/2022]
Abstract
The shared role of amyloid-β (Aβ) deposition in cerebral amyloid angiopathy (CAA) and Alzheimer disease (AD) is arguably the clearest instance of crosstalk between neurodegenerative and cerebrovascular processes. The pathogenic pathways of CAA and AD intersect at the levels of Aβ generation, its circulation within the interstitial fluid and perivascular drainage pathways and its brain clearance, but diverge in their mechanisms of brain injury and disease presentation. Here, we review the evidence for and the pathogenic implications of interactions between CAA and AD. Both pathologies seem to be driven by impaired Aβ clearance, creating conditions for a self-reinforcing cycle of increased vascular Aβ, reduced perivascular clearance and further CAA and AD progression. Despite the close relationship between vascular and plaque Aβ deposition, several factors favour one or the other, such as the carboxy-terminal site of the peptide and specific co-deposited proteins. Amyloid-related imaging abnormalities that have been seen in trials of anti-Aβ immunotherapy are another probable intersection between CAA and AD, representing overload of perivascular clearance pathways and the effects of removing Aβ from CAA-positive vessels. The intersections between CAA and AD point to a crucial role for improving vascular function in the treatment of both diseases and indicate the next steps necessary for identifying therapies.
Collapse
Affiliation(s)
- Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Brian J Bacskai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mar Hernandez-Guillamon
- Neurovascular Research Laboratory, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jeremy Pruzin
- Center for Alzheimer Research and Treatment, Brigham & Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa Sperling
- Center for Alzheimer Research and Treatment, Brigham & Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Weaver NA, Doeven T, Barkhof F, Biesbroek JM, Groeneveld ON, Kuijf HJ, Prins ND, Scheltens P, Teunissen CE, van der Flier WM, Biessels GJ. Cerebral amyloid burden is associated with white matter hyperintensity location in specific posterior white matter regions. Neurobiol Aging 2019; 84:225-234. [DOI: 10.1016/j.neurobiolaging.2019.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 11/24/2022]
|
36
|
Carmona-Iragui M, Videla L, Lleó A, Fortea J. Down syndrome, Alzheimer disease, and cerebral amyloid angiopathy: The complex triangle of brain amyloidosis. Dev Neurobiol 2019; 79:716-737. [PMID: 31278851 DOI: 10.1002/dneu.22709] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/04/2019] [Accepted: 07/02/2019] [Indexed: 11/07/2022]
Abstract
Down syndrome (DS) is the main genetic cause of intellectual disability worldwide. The overexpression of the Amyloid Precursor Protein, present in chromosome 21, leads to β-amyloid deposition that results in Alzheimer disease (AD) and, in most cases, also to cerebral amyloid angiopathy (CAA) neuropathology. People with DS invariably develop the neuropathological hallmarks of AD at the age of 40, and they are at an ultra high risk for suffering AD-related cognitive impairment thereafter. In the general population, cerebrovascular disease is a significant contributor to AD-related cognitive impairment, while in DS remains understudied. This review describes the current knowledge on cerebrovascular disease in DS and reviews the potential biomarkers that could be useful in the future studies, focusing on CAA. We also discuss available evidence on sporadic AD or other genetically determined forms of AD. We highlight the urgent need of large biomarker-characterized cohorts, including neuropathological correlations, to study the exact contribution of CAA and related vascular factors that play a role in cognition and occur with aging, their characterization and interrelationships. DS represents a unique context in which to perform these studies as this population is relatively protected from some conventional vascular risk factors and they develop significant CAA, DS represents a particular atheroma-free model to study AD-related vascular pathologies. Only deepening on these underlying mechanisms, new preventive and therapeutic strategies could be designed to improve the quality of life of this population and their caregivers and lead to new avenues of treatment also in the general AD population.
Collapse
Affiliation(s)
- María Carmona-Iragui
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i 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 Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Videla
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i 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 Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i 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 Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
37
|
Slightly Symptomatic Cerebral Amyloid Angiopathy-Related Inflammation with Spontaneous Remission in Four Months. Case Rep Neurol Med 2019; 2019:5308208. [PMID: 31355028 PMCID: PMC6637666 DOI: 10.1155/2019/5308208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 11/18/2022] Open
Abstract
Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a rare variant of CAA with autoimmune inflammation. A 77-year-old female experienced light-headedness during walking and mild ataxic gait without any other objective neuropsychological deficits. Brain magnetic resonance imaging (MRI) revealed an area of abnormal signal and mild parenchymal swelling in the right parietal lobe, indicating vasogenic edema. T2⁎-weighted gradient echo imaging revealed some subcortical microbleeds in the same lesion. Based on the proposed criteria for CAA-ri, she was diagnosed with probable CAA-ri. After 4 months, the spontaneous improvement was noted in the patient's clinical and radiological findings. This report presents a rare and atypical case of CAA-ri in which the diagnosis was established after the patient underwent neuroimaging for only mild neurological symptoms, and the patient's clinical and radiological findings displayed spontaneous improvement. Despite typical and striking MRI findings of CAA-ri, this patient only presented a minimal symptom; this dissociation could highlight the significance of not misinterpreting any new neurological symptoms. Thus, increased availability of MRI and growing awareness of CAA-ri might result in more incidentally diagnosed cases in the future. Furthermore, this case suggests that it would be better to strictly monitor the clinical-radiological findings of patients with probable CAA-ri who only present with minimal symptoms without the initiation of immunosuppressive therapy.
Collapse
|
38
|
Dehury B, Tang N, Blundell TL, Kepp KP. Structure and dynamics of γ-secretase with presenilin 2 compared to presenilin 1. RSC Adv 2019; 9:20901-20916. [PMID: 35515530 PMCID: PMC9065803 DOI: 10.1039/c9ra02623a] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
Severe early-onset familial Alzheimer's disease (FAD) is caused by more than 200 different mutations in the genes coding for presenilin, the catalytic subunit of the 4-subunit protease complex γ-secretase, which cleaves the C99 fragment of the amyloid precursor protein (APP) to produce Aβ peptides. γ-Secretase exists with either of two homologues, PS1 and PS2. All cryo-electron microscopic structures and computational work has so far focused on γ-secretase with PS1, yet PS2 mutations also cause FAD. A central question is thus whether there are structural and dynamic differences between PS1 and PS2. To address this question, we use the cryo-electron microscopic data for PS1 to develop the first structural and dynamic model of PS2-γ-secretase in the catalytically relevant mature membrane-bound state at ambient temperature, equilibrated by three independent 500 ns molecular dynamics simulations. We find that the characteristic nicastrin extra-cellular domain breathing mode and major movements in the cytosolic loop between TM6 and TM7 occur in both PS2- and PS1-γ-secretase. The overall structures and conformational states are similar, suggesting similar catalytic activities. However, at the sequence level, charge-controlled membrane-anchoring is extracellular for PS1 and intracellular for PS2, which suggests different subcellular locations. The tilt angles of the TM2, TM6, TM7 and TM9 helices differ in the two forms of γ-secretase, suggesting that the two proteins have somewhat different substrate processing and channel sizes. Our MD simulations consistently indicated that PS2 retains several water molecules near the catalytic site at the bilayer, as required for catalysis. The possible reasons for the differences of PS1 and PS2 are discussed in relation to their location and function.
Collapse
Affiliation(s)
- Budheswar Dehury
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| | - Ning Tang
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge Cambridge CB2 1GA UK
| | - Kasper P Kepp
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| |
Collapse
|
39
|
Vöglein J, Paumier K, Jucker M, Preische O, McDade E, Hassenstab J, Benzinger TL, Noble JM, Berman SB, Graff-Radford NR, Ghetti B, Farlow MR, Chhatwal J, Salloway S, Xiong C, Karch CM, Cairns N, Mori H, Schofield PR, Masters CL, Goate A, Buckles V, Fox N, Rossor M, Chrem P, Allegri R, Ringman JM, Höglinger G, Steiner H, Dieterich M, Haass C, Laske C, Morris JC, Bateman RJ, Danek A, Levin J. Clinical, pathophysiological and genetic features of motor symptoms in autosomal dominant Alzheimer's disease. Brain 2019; 142:1429-1440. [PMID: 30897203 PMCID: PMC6735903 DOI: 10.1093/brain/awz050] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 12/21/2018] [Accepted: 01/13/2019] [Indexed: 11/14/2022] Open
Abstract
Owing to an early and marked deposition of amyloid-β in the basal ganglia, autosomal dominant Alzheimer's disease could distinctly involve motor symptoms. Therefore, we aimed to assess the prevalence and characteristics of motor signs in autosomal dominant Alzheimer's disease. Baseline Unified Parkinson Disease Rating Scale part three scores (UPDRS-III) from 433 participants of the Dominantly Inherited Alzheimer's Network observational study were analysed. Motor symptoms were scrutinized with respect to associations with mutation carrier status, mutation site within PSEN1, basal ganglia amyloid-β as measured by Pittsburgh compound B PET, estimated years to symptom onset and Clinical Dementia Rating Scale-Sum of Boxes. Motor findings in mutation carriers were compared to patients with sporadic Alzheimer's disease using data of the National Alzheimer's Coordination Center. Mutation carriers showed motor findings at a higher frequency (28.4% versus 12.8%; P < 0.001) and severity (mean UPDRS-III scores 2.0 versus 0.4; P < 0.001) compared to non-carriers. Eleven of the 27 UPDRS-III items were statistically more frequently affected in mutation carriers after adjustment for multiple comparisons. Ten of these 11 items were subscale components of bradykinesia. In cognitively asymptomatic mutation carriers, dysdiadochokinesia was more frequent compared to non-carriers (right hand: 3.8% versus 0%; adjusted P = 0.023; left: 4.4% versus 0.6%; adjusted P = 0.031). In this cohort, the positive predictive value for mutation carrier status in cognitively asymptomatic participants (50% a priori risk) of dysdiadochokinesia was 100% for the right and 87.5% for the left side. Mutation carriers with motor findings more frequently were basal ganglia amyloid-β positive (84% versus 63.3%; P = 0.006) and showed more basal ganglia amyloid-β deposition (Pittsburgh compound B-standardized uptake value ratio 2.472 versus 1.928; P = 0.002) than those without. Frequency and severity of motor findings were greater in post-codon 200 PSEN1 mutations (36%; mean UPDRS-III score 3.03) compared to mutations pre-codon 200 PSEN1 (19.3%, P = 0.022; 0.91, P = 0.013). In mutation carriers, motor symptom severity was significantly positively correlated with basal ganglia amyloid-β deposition, Clinical Dementia Rating scores and estimated years to symptom onset. Mutation carriers with a Clinical Dementia Rating global score of 2 exhibited more pronounced motor symptoms than sporadic Alzheimer's disease patients with the same Clinical Dementia Rating global score (mean UPDRS-III scores 20.71 versus 5.96; P < 0.001). With a prevalence of approximately 30% and increasing severity with progression of dementia, motor symptoms are proven as a clinically relevant finding in autosomal dominant Alzheimer's disease, in particular in advanced dementia stages, that correlates with deposition of amyloid-β in the basal ganglia. In a very small per cent of cognitively asymptomatic members of families with autosomal dominant Alzheimer's disease, dysdiadochokinesia may increase the chance of an individual's status as mutation carrier.
Collapse
Affiliation(s)
- Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrina Paumier
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Oliver Preische
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Eric McDade
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Jason Hassenstab
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Tammie L Benzinger
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, and Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, 710 West 168th Street Box 176, New York, NY, USA
| | - Sarah B Berman
- University of Pittsburgh, 3471 Fifth Ave #900, Pittsburgh, PA, USA
| | | | | | - Martin R Farlow
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Chengjie Xiong
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Celeste M Karch
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Nigel Cairns
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Hiroshi Mori
- Osaka City University Medical School, Asahimachi, Abenoku, Osaka 545–8585, Japan
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Colin L Masters
- Florey Institute, University of Melbourne, Level 5, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, B1065, New York, NY, USA
| | - Virginia Buckles
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Nick Fox
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | - Martin Rossor
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | | | | | - John M Ringman
- Keck School of Medicine of University of Southern California, Center for the Health Professionals, 1540 Alcazar Street, Suite 209F, Los Angeles, CA, USA
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedical Center (BMC), Metabolic Biochemistry, LMU Munich, Germany
| | - Marianne Dieterich
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Vertigo and Balance Disorders, Ludwig Maximilians University, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Biomedical Center (BMC), Metabolic Biochemistry, LMU Munich, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
| | - John C Morris
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University School of Medicine, 660 South Euclid, Saint Louis, MO, USA
| | - Adrian Danek
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | |
Collapse
|
40
|
Gu X, Chu T, Liu L, Han X. Genetic influences on white matter and metabolism abnormal change in Alzheimer's disease: Meta-analysis for neuroimaging research on presenilin 1 mutation. Clin Neurol Neurosurg 2019; 177:47-53. [PMID: 30599314 DOI: 10.1016/j.clineuro.2018.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 10/18/2018] [Accepted: 12/24/2018] [Indexed: 11/19/2022]
Abstract
Mutations in the presenilin1 (PSEN1) cause familial Alzheimer's disease (FAD), providing a special opportunity to study pre-symptomatic individuals who would be predicted to develop Alzheimer's disease (AD) in the future. However, whether presenilin1 (PSEN1) genotype and neuroimaging markers is a harbinger of AD remains controversial. We aimed to explore the association of PSEN1 genotype with neuroimaging markers of AD: white matter integrity, cerebral amyloid deposition and brain metabolism. We reviewed studies of diffusion tensor imaging (DTI), amyloid deposition and cerebral metabolism in patients with AD and control, in order to address the relative change of white matter microstructural associated with PSEN1 genotype. We performed a systematic meta-analysis and review of 11 cross-sectional studies identified in several database from 2008 to 2018 (n = 165). The pooled standard mean difference (SMD) value was calculated to estimate the association between PSEN1 and white matter change and brain metabolism. PSEN1 mutation carrier status was associated with mean diffusivity (MD) change (pooled SMD: 2.29; 95% CI 1.04 to 3.53; p < 0.001) and increased cerebral amyloid positron emission tomography tracer (pooled SMD: 3.78, 95% CI 1.04 to 6.53, p = 0.007). PSEN1 was not associated with white matter metabolism change (p = 0.069). PSEN1 was associated with mean diffusivity (MD) increase in DTI markers and decreased brain metabolism. Theses associations may suggest the potential role of the PSEN1 gene and imaging marker in Alzheimer's disease.
Collapse
Affiliation(s)
- Xiaochun Gu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China; Key Laboratory of Developmental Genes and Human Diseases, Department of Histology Embryology, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China.
| | - Tao Chu
- Nanjing Normal University Affiliated Middle School Xincheng Junior High School, 123 Huangshan Road, Nanjing 210009, China
| | - Li Liu
- Key Laboratory of Developmental Genes and Human Diseases, Department of Histology Embryology, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Xiao Han
- Key Laboratory of Developmental Genes and Human Diseases, Department of Histology Embryology, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| |
Collapse
|
41
|
Xu Y, Liu X, Shen J, Tian W, Fang R, Li B, Ma J, Cao L, Chen S, Li G, Tang H. The Whole Exome Sequencing Clarifies the Genotype- Phenotype Correlations in Patients with Early-Onset Dementia. Aging Dis 2018; 9:696-705. [PMID: 30090657 PMCID: PMC6065298 DOI: 10.14336/ad.2018.0208] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/08/2018] [Indexed: 12/23/2022] Open
Abstract
Our study aimed to identify the underlying causes in patients with early onset dementia by clinical and genetic exploration. We recruited a group of 38 patients with early-onset dementia. Firstly, hexanucleotide repeat expansions in C9ORF72 gene were screened in all subjects to exclude the possibility of copy number variation. Then, the whole exome sequencing (WES) was conducted, and the data were analyzed focusing on 89 dementia-related causing and susceptible genes. The effects of identified variants were classified according to the American College of Medical Genetics and Genomics (ACMG) standards and guidelines. There were no pathogenic expansions in C9ORF72 detected. According to the ACMG standards and guidelines, we identified five known pathogenic mutations, PSEN1 P284L, PSEN1c.857-1G>A, PSEN1 I143T, PSEN1 G209E and MAPT G389R, and one novel pathogenic mutation APP K687N. All these mutations caused dementia with the mean onset age of 38.3 (range from 27 to 51) and rapid progression. Eleven variants with uncertain significance were also detected and needed further verification. The clinical phenotypes of dementia are heterogeneous, with both onset ages and clinical features being influenced by mutation position as well as the causative gene. WES can serve as efficient diagnostic tools for different heterogeneous dementia.
Collapse
Affiliation(s)
- Yangqi Xu
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoli Liu
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,3Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Junyi Shen
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wotu Tian
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Fang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,4Department of Neurology, Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Binyin Li
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianfang Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cao
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shengdi Chen
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guanjun Li
- 2Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huidong Tang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Woollacott IOC, Bocchetta M, Sudre CH, Ridha BH, Strand C, Courtney R, Ourselin S, Cardoso MJ, Warren JD, Rossor MN, Revesz T, Fox NC, Holton JL, Lashley T, Rohrer JD. Pathological correlates of white matter hyperintensities in a case of progranulin mutation associated frontotemporal dementia. Neurocase 2018; 24:166-174. [PMID: 30112957 PMCID: PMC6168954 DOI: 10.1080/13554794.2018.1506039] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
White matter hyperintensities (WMH) are often seen on MRI brain scans in frontotemporal dementia (FTD) due to progranulin (GRN) mutations, but their pathological correlates are unknown. We examined the histological changes underlying WMH in a patient with GRN mutation associated behavioral variant FTD. In vivo and cadaveric MRI showed progressive, asymmetric frontotemporal and parietal atrophy, and asymmetrical WMH predominantly affecting frontal mid-zones. We first performed segmentation and localization analyses of WMH present on cadaveric MRI FLAIR images, then selected five different brain regions directly matched to differing severities of WMH for histological analysis. We used immunohistochemistry to assess vascular pathology, degree of spongiosis, neuronal and axonal loss, TDP-43, demyelination and astrogliosis, and microglial burden and morphology. Brain regions with significant WMH displayed severe cortical and white matter pathology, and prominent white matter microglial activation and microglial dystrophy, but only mild axonal loss and minimal vascular pathology. Our study suggests that WMH in GRN mutation carriers are not secondary to vascular pathology. Whilst cortical pathology induced axonal degeneration could contribute to white matter damage, individuals with GRN mutations could develop selective white matter vulnerability and myelin loss due to chronic, regional microglial dysfunction arising from GRN haploinsufficiency.
Collapse
Affiliation(s)
- Ione O C Woollacott
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| | - Martina Bocchetta
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| | - Carole H Sudre
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK.,b Translational Imaging Group, Centre for Medical Image Computing , University College London , London , UK
| | - Basil H Ridha
- c NIHR Queen Square Dementia Biomedical Research Unit , UCL Institute of Neurology , London , UK
| | - Catherine Strand
- d Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience , UCL Institute of Neurology , London , UK
| | - Robert Courtney
- d Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience , UCL Institute of Neurology , London , UK
| | - Sebastien Ourselin
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK.,b Translational Imaging Group, Centre for Medical Image Computing , University College London , London , UK
| | - M Jorge Cardoso
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK.,b Translational Imaging Group, Centre for Medical Image Computing , University College London , London , UK
| | - Jason D Warren
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| | - Martin N Rossor
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| | - Tamas Revesz
- d Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience , UCL Institute of Neurology , London , UK
| | - Nick C Fox
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| | - Janice L Holton
- d Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience , UCL Institute of Neurology , London , UK
| | - Tammaryn Lashley
- d Queen Square Brain Bank for Neurological Disorders, Department of Molecular Neuroscience , UCL Institute of Neurology , London , UK
| | - Jonathan D Rohrer
- a Dementia Research Centre, Department of Neurodegenerative Disease , UCL Institute of Neurology , London , UK
| |
Collapse
|
43
|
Lee S, Zimmerman ME, Narkhede A, Nasrabady SE, Tosto G, Meier IB, Benzinger TLS, Marcus DS, Fagan AM, Fox NC, Cairns NJ, Holtzman DM, Buckles V, Ghetti B, McDade E, Martins RN, Saykin AJ, Masters CL, Ringman JM, Fӧrster S, Schofield PR, Sperling RA, Johnson KA, Chhatwal JP, Salloway S, Correia S, Jack CR, Weiner M, Bateman RJ, Morris JC, Mayeux R, Brickman AM. White matter hyperintensities and the mediating role of cerebral amyloid angiopathy in dominantly-inherited Alzheimer's disease. PLoS One 2018; 13:e0195838. [PMID: 29742105 PMCID: PMC5942789 DOI: 10.1371/journal.pone.0195838] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 04/01/2018] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION White matter hyperintensity (WMH) volume on MRI is increased among presymptomatic individuals with autosomal dominant mutations for Alzheimer's disease (AD). One potential explanation is that WMH, conventionally considered a marker of cerebrovascular disease, are a reflection of cerebral amyloid angiopathy (CAA) and that increased WMH in this population is a manifestation of this vascular form of primary AD pathology. We examined whether the presence of cerebral microbleeds, a marker of CAA, mediates the relationship between WMH and estimated symptom onset in individuals with and without autosomal dominant mutations for AD. PARTICIPANTS AND METHODS Participants (n = 175, mean age = 41.1 years) included 112 with an AD mutation and 63 first-degree non-carrier controls. We calculated the estimated years from expected symptom onset (EYO) and analyzed baseline MRI data for WMH volume and presence of cerebral microbleeds. Mixed effects regression and tests of mediation were used to examine microbleed and WMH differences between carriers and non-carriers and to test the whether the association between WMH and mutation status is dependent on the presence of microbleeds. RESULTS Mutation carriers were more likely to have microbleeds than non-carriers (p<0.05) and individuals with microbleeds had higher WMH volume than those without (p<0.05). Total WMH volume was increased in mutation carriers compared with non-carriers, up to 20 years prior to EYO, after controlling for microbleed status, as we demonstrated previously. Formal testing of mediation demonstrated that 21% of the association between mutation status and WMH was mediated by presence of microbleeds (p = 0.03) but a significant direct effect of WMH remained (p = 0.02) after controlling for presence of microbleeds. DISCUSSION Although there is some co-dependency between WMH and microbleeds, the observed increases in WMH among mutation carriers does not appear to be fully mediated by this marker of CAA. The findings highlight the possibility that WMH represent a core feature of AD independent of vascular forms of beta amyloid.
Collapse
Affiliation(s)
- Seonjoo Lee
- Research Foundation for Mental Hygiene, Inc., New York, NY, United States of America
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Molly E. Zimmerman
- Psychology Department, Fordham University, Bronx, NY, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Atul Narkhede
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Sara E. Nasrabady
- Research Foundation for Mental Hygiene, Inc., New York, NY, United States of America
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Irene B. Meier
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Tammie L. S. Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Daniel S. Marcus
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Nick C. Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Nigel J. Cairns
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Virginia Buckles
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Ralph N. Martins
- Centre of Excellence of Alzheimer’s Disease Research and Care, School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Andrew J. Saykin
- Indiana Alzheimer Disease Center and Center for Neuroimaging, Department of Radiology and Imaging Science, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Colin L. Masters
- The Florey Institute, University of Melbourne, Parkville, Australia
| | - John M. Ringman
- Memory and Aging Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States of America
| | - Stefan Fӧrster
- German Center for Neurodegenerative Diseases (DZNE) München and Tübingen and Department of Nuclear Medicine, Technische Universität München (TUM), Munich, Germany
| | - Peter R. Schofield
- Neuroscience Research Australia and University of New South Wales, Sydney, Australia
| | - Reisa A. Sperling
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Keith A. Johnson
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Jasmeer P. Chhatwal
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Stephen Salloway
- Butler Hospital and Department of Neurology, Alpert Medical School, Brown University, Providence, RI, United States of America
| | - Stephen Correia
- Department of Psychiatry & Human Behavior, Alpert Medical School, Brown University, Providence, RI United States of America
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, United States of America
| | - Michael Weiner
- Department of Radiology and Biomedical Imaging, Center for Imaging of Neurodegenerative Diseases, San Francisco Veterans Affairs Medical Center and Departments of Psychiatry, Radiology, Medicine, and Neurology, University of California at San Francisco, San Francisco, CA, United States of America
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Richard Mayeux
- Research Foundation for Mental Hygiene, Inc., New York, NY, United States of America
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
- * E-mail:
| | | |
Collapse
|
44
|
Pietroboni AM, Scarioni M, Carandini T, Basilico P, Cadioli M, Giulietti G, Arighi A, Caprioli M, Serra L, Sina C, Fenoglio C, Ghezzi L, Fumagalli GG, De Riz MA, Calvi A, Triulzi F, Bozzali M, Scarpini E, Galimberti D. CSF β-amyloid and white matter damage: a new perspective on Alzheimer's disease. J Neurol Neurosurg Psychiatry 2018; 89:352-357. [PMID: 29054920 DOI: 10.1136/jnnp-2017-316603] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/13/2017] [Accepted: 09/28/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To assess the connection between amyloid pathology and white matter (WM) macrostructural and microstructural damage in demented patients compared with controls. METHODS Eighty-five participants were recruited: 65 with newly diagnosed Alzheimer's disease (AD), non-AD dementia or mild cognitive impairment and 20 age-matched and sex-matched healthy controls. β-amyloid1-42 (Aβ) levels were determined in cerebrospinal fluid (CSF) samples from all patients and five controls. Among patients, 42 had pathological CSF Aβ levels (Aβ(+)), while 23 had normal CSF Aβ levels (Aβ(-)). All participants underwent neurological examination, neuropsychological testing and brain MRI. We used T2-weighted scans to quantify WM lesion loads (LLs) and diffusion-weighted images to assess their microstructural substrate. Non-parametric statistical tests were used for between-group comparisons and multiple regression analyses. RESULTS We found an increased WM-LL in Aβ(+) compared with both, healthy controls (p=0.003) and Aβ(-) patients (p=0.02). Interestingly, CSF Aβ concentration was the best predictor of patients' WM-LL (r=-0.30, p<0.05) when using age as a covariate. Lesion apparent diffusion coefficient value was higher in all patients than in controls (p=0.0001) and correlated with WM-LL (r=0.41, p=0.001). In Aβ(+), WM-LL correlated with WM microstructural damage in the left peritrigonal WM (p<0.0001). CONCLUSIONS WM damage is crucial in AD pathogenesis. The correlation between CSF Aβ levels and WM-LL suggests a direct link between amyloid pathology and WM macrostructural and microstructural damage.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Scarioni
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziana Carandini
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Basilico
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Marcello Cadioli
- Department of Pathophysiology and Transplantation, Neuroradiology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Andrea Arighi
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Michela Caprioli
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Serra
- Neuroimaging Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Clara Sina
- Department of Pathophysiology and Transplantation, Neuroradiology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Ghezzi
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio G Fumagalli
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Milena A De Riz
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Calvi
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- Department of Pathophysiology and Transplantation, Neuroradiology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Elio Scarpini
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Department of Pathophysiology and Transplantation, Neurodegenerative Disease Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
45
|
Fiford CM, Ridgway GR, Cash DM, Modat M, Nicholas J, Manning EN, Malone IB, Biessels GJ, Ourselin S, Carmichael OT, Cardoso MJ, Barnes J. Patterns of progressive atrophy vary with age in Alzheimer's disease patients. Neurobiol Aging 2018; 63:22-32. [PMID: 29220823 PMCID: PMC5805840 DOI: 10.1016/j.neurobiolaging.2017.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/14/2017] [Accepted: 11/06/2017] [Indexed: 01/18/2023]
Abstract
Age is not only the greatest risk factor for Alzheimer's disease (AD) but also a key modifier of disease presentation and progression. Here, we investigate how longitudinal atrophy patterns vary with age in mild cognitive impairment (MCI) and AD. Data comprised serial longitudinal 1.5-T magnetic resonance imaging scans from 153 AD, 339 MCI, and 191 control subjects. Voxel-wise maps of longitudinal volume change were obtained and aligned across subjects. Local volume change was then modeled in terms of diagnostic group and an interaction between group and age, adjusted for total intracranial volume, white-matter hyperintensity volume, and apolipoprotein E genotype. Results were significant at p < 0.05 with family-wise error correction for multiple comparisons. An age-by-group interaction revealed that younger AD patients had significantly faster atrophy rates in the bilateral precuneus, parietal, and superior temporal lobes. These results suggest younger AD patients have predominantly posterior progressive atrophy, unexplained by white-matter hyperintensity, apolipoprotein E, or total intracranial volume. Clinical trials may benefit from adapting outcome measures for patient groups with lower average ages, to capture progressive atrophy in posterior cortices.
Collapse
Affiliation(s)
- Cassidy M Fiford
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Gerard R Ridgway
- FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Wellcome Trust Centre for Neuroimaging, 12 Queen Square, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Marc Modat
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - Emily N Manning
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sebastien Ourselin
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - M Jorge Cardoso
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Josephine Barnes
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| |
Collapse
|
46
|
Tiedt HO, Benjamin B, Niedeggen M, Lueschow A. Phenotypic Variability in Autosomal Dominant Familial Alzheimer Disease due to the S170F Mutation of Presenilin-1. NEURODEGENER DIS 2018; 18:57-68. [PMID: 29466804 DOI: 10.1159/000485899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/29/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND In rare cases, patients with Alzheimer disease (AD) present at an early age and with a family history suggestive of an autosomal dominant mode of inheritance. Mutations of the presenilin-1 (PSEN1) gene are the most common causes of dementia in these patients. Early-onset and particularly familial AD patients frequently present with variable non-amnestic cognitive symptoms such as visual, language or behavioural changes as well as non-cognitive, e.g. motor, symptoms. OBJECTIVE To investigate the phenotypic variability in carriers of the PSEN1 S170F mutation. METHODS We report a family with 4 patients carrying the S170F mutation of whom 2 underwent detailed clinical examinations. We discuss our current findings in the context of previously reported S170F cases. RESULTS The clinical phenotype was consistent regarding initial memory impairment and early onset in the late twenties found in all S170F patients. There were frequent non-amnestic cognitive changes and, at early stages of the disease, indications of a more pronounced disturbance of visuospatial abilities as compared to face and object recognition. Non-cognitive symptoms most often included myoclonus and cerebellar ataxia. A review of the available case reports indicates some phenotypic variability associated with the S170F mutation including different constellations of symptoms such as parkinsonism and delusions. CONCLUSION The variable clinical findings associated with the S170F mutation highlight the relevance of atypical phenotypes in the context of research and under a clinical perspective. CSF sampling and detection of Aβ species may be essential to indicate AD pathology in unclear cases presenting with cognitive and motor symptoms at a younger age.
Collapse
Affiliation(s)
- Hannes O Tiedt
- Department of Neurology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin (CBF), Berlin, Germany
| | | | | | | |
Collapse
|
47
|
Sellal F, Wallon D, Martinez-Almoyna L, Marelli C, Dhar A, Oesterlé H, Rovelet-Lecrux A, Rousseau S, Kourkoulis CE, Rosand J, DiPucchio ZY, Frosch M, Gombert C, Audoin B, Miné M, Riant F, Frebourg T, Hannequin D, Campion D, Greenberg SM, Tournier-Lasserve E, Nicolas G. APP Mutations in Cerebral Amyloid Angiopathy with or without Cortical Calcifications: Report of Three Families and a Literature Review. J Alzheimers Dis 2018; 56:37-46. [PMID: 27858710 DOI: 10.3233/jad-160709] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Specific APP mutations cause cerebral amyloid angiopathy (CAA) with or without Alzheimer's disease (AD). OBJECTIVE We aimed at reporting APP mutations associated with CAA, describe the clinical, cerebrospinal fluid AD biomarkers, and neuroimaging features, and compare them with the data from the literature. METHODS We performed a retrospective study in two French genetics laboratories by gathering all clinical and neuroimaging data from patients referred for a genetic diagnosis of CAA with an age of onset before 66 years and fulfilling the other Boston revised criteria. We studied the segregation of mutations in families and performed a comprehensive literature review of all cases reported with the same APP mutation. RESULTS We screened APP in 61 unrelated French patients. Three mutations, located in the Aβ coding region, were detected in five patients from three families: p.Ala692Gly (Flemish), p.Glu693Lys (Italian), and p.Asp694Asn (Iowa). Patients exhibited CAA and progressive cognitive impairment associated with cortical calcifications in the Iowa and Italian mutation carriers, but not the patient carrying the Flemish mutation. CONCLUSIONS This is the first evidence of cortical calcification in patients with an APP mutation other than the Iowa mutation. We discuss the radiological, cerebrospinal fluid, and clinical phenotype of patients carrying these mutations in the literature.
Collapse
Affiliation(s)
- François Sellal
- Department of Neurology and Consultation Mémoire de Ressource et de Recherche, Hôpitaux Civils de Colmar, Colmar, France.,Strasbourg University, INSERM U-1118, Faculty of Medicine, Strasbourg, France
| | - David Wallon
- Department of Neurology, Rouen University Hospital, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | | | - Cecilia Marelli
- Service de Neurologie, CMRR, CHRU Gui de Chauliac, Montpellier, France
| | - Abhinav Dhar
- Radiology Service, Hospital of Moenchsberg, Mulhouse, France
| | - Héléne Oesterlé
- Radiology Service, Hospital of Moenchsberg, Mulhouse, France
| | - Anne Rovelet-Lecrux
- Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Stéphane Rousseau
- CNR-MAJ, Rouen University Hospital, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Christina E Kourkoulis
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, USA.,Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Jon Rosand
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, USA.,Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Zora Y DiPucchio
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, USA.,Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Matthew Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Claudine Gombert
- Neurology Department, Centre Hospitalier, Aix-en-Provence, France
| | - Bertrand Audoin
- Aix-Marseille Université, CNRS, CRMBM UMR 7339, Marseille, France/APHM, Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie, Marseille, France
| | - Manuèle Miné
- AP-HP, Service de génétique moléculaire neurovasculaire, Hôpital Lariboisiére, Paris, France.,Inserm, U1161, Université Paris 7 Diderot, Paris, France
| | - Florence Riant
- AP-HP, Service de génétique moléculaire neurovasculaire, Hôpital Lariboisiére, Paris, France.,Inserm, U1161, Université Paris 7 Diderot, Paris, France
| | - Thierry Frebourg
- Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - Didier Hannequin
- Department of Neurology, Rouen University Hospital, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - Dominique Campion
- CNR-MAJ, Rouen University Hospital, Rouen, France.,Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Research, Rouvray Psychiatric Hospital, Sotteville-Lés-Rouen, France
| | - Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, USA.,Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Elisabeth Tournier-Lasserve
- AP-HP, Service de génétique moléculaire neurovasculaire, Hôpital Lariboisiére, Paris, France.,Inserm, U1161, Université Paris 7 Diderot, Paris, France
| | - Gaël Nicolas
- CNR-MAJ, Rouen University Hospital, Rouen, France.,Inserm U1079, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| |
Collapse
|
48
|
Takahashi Y, Ohta Y, Sasaki R, Tadokoro K, Sato K, Shang J, Takemoto M, Hishikawa N, Yamashita T, Haraguchi T, Ikeuchi T, Abe K. A novel presenilin 1 mutation (Leu418Trp) associated with spasticity, parkinsonism, and white matter lesion in a dominant Alzheimer's family. J Neurol Sci 2018; 387:166-169. [PMID: 29571857 DOI: 10.1016/j.jns.2018.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/30/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Affiliation(s)
- Yoshiaki Takahashi
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Yasuyuki Ohta
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Ryo Sasaki
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kou Tadokoro
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kota Sato
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Jingwei Shang
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Mami Takemoto
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Nozomi Hishikawa
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Toru Yamashita
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Takashi Haraguchi
- National Hospital Organization Minami-Okayama Medical Center, Okayama, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Koji Abe
- Departments of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan.
| |
Collapse
|
49
|
Different Hippocampus Functional Connectivity Patterns in Healthy Young Adults with Mutations of APP/Presenilin-1/2 and APOEε4. Mol Neurobiol 2017; 55:3439-3450. [PMID: 28502043 DOI: 10.1007/s12035-017-0540-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
Abstract
This study aims to explore the hippocampus-based functional connectivity patterns in young, healthy APP and/or presenilin-1/2 mutation carriers and APOE ε4 subjects. Seventy-eight healthy young adults (33 male, mean age 24.0 ± 2.2 years; 18 APP and/or presenilin1/2 mutation carriers [APP/presenilin-1/2 group], 30 APOE ε4 subjects [APOE ε4 group], and 30 subjects without the above-mentioned genes [control group]) underwent resting-state functional MR imaging and neuropsychological assessments. Bilateral hippocampus functional connectivity patterns were compared among three groups. The brain regions with statistical differences were then extracted, and correlation analyses were performed between Z values of the brain regions and neuropsychological results. Compared with control group, both APOE ε4 group and APP/presenilin-1/2 group showed increased functional connectivity in medial prefrontal cortex and precuneus for the seeds of bilateral hippocampi. The APOE ε4 group displayed increased functional connectivity from bilateral hippocampi to the left middle temporal gyrus compared with the control group. Moreover, compared with the APP/presenilin-1/2 group, the APOE ε4 group also had markedly increased functional connectivity in right hippocampus-left middle temporal gyrus. The Z values of right hippocampus-left middle temporal gyrus correlated with various neuropsychological results across all the subjects, as well as in APOE ε4 group. Young healthy adults carrying APOE ε4 and APP/presenilin-1/2 displayed different hippocampus functional connectivity patterns, which may underlie the discrepant mechanisms of gene-modulated cognitive dysfunction in Alzheimer's disease.
Collapse
|
50
|
Roseborough A, Ramirez J, Black SE, Edwards JD. Associations between amyloid β and white matter hyperintensities: A systematic review. Alzheimers Dement 2017; 13:1154-1167. [PMID: 28322203 DOI: 10.1016/j.jalz.2017.01.026] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 11/25/2022]
Abstract
INTRODUCTION This systematic review synthesizes current evidence for associations between cortical amyloid β, visualized on amyloid positron emission tomography imaging, and white matter hyperintensity (WMH) burden on magnetic resonance imaging in healthy elderly adults and individuals with cognitive impairment and dementia. METHODS Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) systematic review guidelines, we systematically searched MEDLINE, Embase, Cochrane, and PsycINFO databases from January 2000 to September 2015. RESULTS Our search returned 492 articles, 34 of which met criteria for inclusion in the final selection. Most studies reported no significant relationships between amyloid β and WMH burden across diagnostic groups. DISCUSSION Findings of this systematic review suggest that amyloid accumulation and WMH are independent but additive processes. The limited number of independent cohorts, lack of longitudinal data, and exclusion of individuals with mixed dementia limit the generalizability of these findings. Further studies are required to elucidate the putative contributions of vascular processes to neurodegenerative pathology.
Collapse
Affiliation(s)
- Austyn Roseborough
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Toronto, Ontario, Canada
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Toronto, Ontario, Canada; Department of Medicine, Neurology, University of Toronto and Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Jodi D Edwards
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Toronto, Ontario, Canada.
| |
Collapse
|