1
|
Sin MK, Cheng Y, Ahmed A, Roseman JM, Dowling NM, Zamrini E. Cerebral Amyloid Angiopathy, Dementia, and Alzheimer Neuropathologic Changes: Findings From the ACT Autopsy Cohort. Neurology 2024; 103:e210009. [PMID: 39481068 PMCID: PMC11527483 DOI: 10.1212/wnl.0000000000210009] [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: 03/29/2024] [Accepted: 09/04/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral amyloid angiopathy (CAA) is common in older adults and is associated with dementia. Less is known whether this association is mediated by Alzheimer disease (AD) neuropathologic changes, the examination of which was the objective of this study. METHODS This was a retrospective cross-sectional examination of the Kaiser Permanente Washington database of the Adult Changes in Thought (ACT) autopsy cohort with information on CAA, dementia, the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) (amyloid neuritic plaques), and Braak (tau neurofibrillary tangles). CAA was diagnosed by immunohistochemistry and dementia by ACT Consensus Diagnostic Conference. AD neuropathology was categorized by CERAD scores and Braak stages. Multivariable logistic regression models were used to estimate odds ratios (ORs) and 95% CIs of the associations of CAA with dementia, adjusting for age at death and sex, and with additional adjustments separately for CERAD scores (moderate-severe vs mild-absent), Braak stages (V-VI vs 0-IV), APOE ε4, and stroke. Formal mediation analyses were conducted to estimate age-sex-adjusted OR (95% CI) for natural indirect effects (NIEs) of CERAD scores and Braak stages. RESULTS The 848 participants had a mean age of 86.7 ± 4.6 years at death, and 57.6% were female. CAA was present in 322 participants (38.0%), of whom 152, 145, and 25 had mild, moderate, and severe CAA, respectively. Dementia was present in 384 participants (45.3%), of whom 317 had AD. Dementia was more common in those with CAA than without (53.7% vs 40.1%; age-sex-adjusted OR 1.57, 95% CI 1.18-2.10). This association remained significant after separate adjustment for other covariates but lost significance when adjusted for CERAD scores (OR 1.27, 95% CI 0.93-1.71) and Braak stages (OR 0.96, 95% CI 0.69-1.33). Findings from our formal mediation analyses show that ORs (95% CIs) for NIE of CERAD scores and Braak stages were 1.25 (1.13-1.37) and 1.63 (1.38-1.88), respectively, and CERAD scores and Braak stages mediated 53% and 111% of the total association, respectively. DISCUSSION We observed a significant association between CAA and dementia that disappeared when adjusted for CERAD or Braak stages. Findings from our mediation analyses suggest that the CAA-dementia association may be potentially mediated by AD neuropathologic changes. This hypothesis needs to be tested in future mechanistic studies in AD accounting for unmeasured confounders.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Yan Cheng
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Ali Ahmed
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Jeffrey M Roseman
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - N Maritza Dowling
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Edward Zamrini
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| |
Collapse
|
2
|
Muir RT, Callum JL, Yu AYX, Kapral MK, Swartz RH, Black SE, MacIntosh BJ, Fergusson DA, Kleinman S, Demchuk AD, Stys PK, Smith EE, Hill MD. Beta-Amyloid Related Neurodegenerative and Neurovascular Diseases: Potential Implications for Transfusion Medicine. Transfus Med Rev 2024; 38:150858. [PMID: 39413667 DOI: 10.1016/j.tmrv.2024.150858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 10/18/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a progressive cerebrovascular and neurodegenerative disorder that is caused by the aberrant accumulation of soluble beta-amyloid isoforms in the small vessel walls of the cerebral and cerebellar cortices and the leptomeninges. Vascular beta-amyloid deposition increases vulnerability to intracerebral hemorrhage (ICH). Clinically, CAA can be the underlying cause of up to half of spontaneous lobar ICHs and can also present with convexity subarachnoid hemorrhage, transient focal neurologic episodes and progressive cognitive decline leading to dementia. The majority of CAA is sporadic, with increasing prevalence with age and often coexists with Alzheimer's Disease (AD). Genetic and iatrogenic etiologies are rare. Cases of CAA and AD have been linked to the use of cadaveric pituitary hormone and later life iatrogenic CAA has also been described following early-life neurosurgical procedures with cadaveric dura grafts. Together these data suggest a capacity of beta-amyloid transmissibility. A recent study found that in over 1 million transfusion recipients from donors who later developed (i) >1 ICH or (ii) one ICH event and dementia, had an elevated risk of developing future ICH. Considering prior reports of transfusion associated variant-Creutzfeldt Jakob Disease in humans and in vivo evidence in sheep, coupled with emerging data supporting beta-amyloid's prion-like properties, raises the question of whether CAA could be transmissible by blood transfusion. This would also have implications for screening, especially in an era of emerging plasma biomarkers of cerebral amyloidosis. Given the public health concerns raised by this biologically plausible question, there is a need for future studies with well-characterized definitions - and temporal ascertainment - of CAA exposure and outcomes to examine whether CAA is transfusion-transmissible, and, if so, with what frequency and timing of onset.
Collapse
Affiliation(s)
- Ryan T Muir
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Jeannie L Callum
- Department of Pathology and Molecular Medicine, Queen's University, Ontario, Canada; Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Amy Y X Yu
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Moira K Kapral
- ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada; Department of Medicine, General Internal Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Richard H Swartz
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Sandra E Black
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Steven Kleinman
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew D Demchuk
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Peter K Stys
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Eric E Smith
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Michael D Hill
- Calgary Stroke Program, Department of Clinical Neurosciences, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
3
|
Fritzen L, Wienken K, Wagner L, Kurtyka M, Vogel K, Körbelin J, Weggen S, Fricker G, Pietrzik CU. Truncated mini LRP1 transports cargo from luminal to basolateral side across the blood brain barrier. Fluids Barriers CNS 2024; 21:74. [PMID: 39289695 PMCID: PMC11409491 DOI: 10.1186/s12987-024-00573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND The most crucial area to focus on when thinking of novel pathways for drug delivery into the CNS is the blood brain barrier (BBB). A number of nanoparticulate formulations have been shown in earlier research to target receptors at the BBB and transport therapeutics into the CNS. However, no mechanism for CNS entrance and movement throughout the CNS parenchyma has been proposed yet. Here, the truncated mini low-density lipoprotein receptor-related protein 1 mLRP1_DIV* was presented as blood to brain transport carrier, exemplified by antibodies and immunoliposomes using a systematic approach to screen the receptor and its ligands' route across endothelial cells in vitro. METHODS The use of mLRP1_DIV* as liposomal carrier into the CNS was validated based on internalization and transport assays across an in vitro model of the BBB using hcMEC/D3 and bEnd.3 cells. Trafficking routes of mLRP1_DIV* and corresponding cargo across endothelial cells were analyzed using immunofluorescence. Modulation of γ-secretase activity by immunoliposomes loaded with the γ-secretase modulator BB25 was investigated in co-cultures of bEnd.3 mLRP1_DIV* cells and CHO cells overexpressing human amyloid precursor protein (APP) and presenilin 1 (PSEN1). RESULTS We showed that while expressed in vitro, mLRP1_DIV* transports both, antibodies and functionalized immunoliposomes from luminal to basolateral side across an in vitro model of the BBB, followed by their mLRP1_DIV* dependent release of the cargo. Importantly, functionalized liposomes loaded with the γ-secretase modulator BB25 were demonstrated to effectively reduce toxic Aß42 peptide levels after mLRP1_DIV* mediated transport across a co-cultured endothelial monolayer. CONCLUSION Together, the data strongly suggest mLRP1_DIV* as a promising tool for drug delivery into the CNS, as it allows a straight transport of cargo from luminal to abluminal side across an endothelial monolayer and it's release into brain parenchyma in vitro, where it exhibits its intended therapeutic effect.
Collapse
Affiliation(s)
- Laura Fritzen
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| | - Katharina Wienken
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Lelia Wagner
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Magdalena Kurtyka
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany
| | - Katharina Vogel
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jakob Körbelin
- Department for Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Hamburg, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Gert Fricker
- Institute for Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University of Mainz, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|
4
|
Zsadanyi SE, Morcillo-Nieto AO, Aranha MR, Aragón I, Arriola-Infante JE, Vaqué-Alcázar L, Montal V, Pegueroles J, Arranz J, Rodríguez-Baz Í, Blesa LM, Videla L, Barroeta I, Del Hoyo Soriano L, Benejam B, Fernández S, Hernandez AS, Bargallo N, González-Ortiz S, Giménez S, Alcolea D, Belbin O, Lleó A, Fortea J, Carmona-Iragui M, Bejanin A. Associations of Microbleeds and Their Topography With Imaging and CSF Biomarkers of Alzheimer Pathology in Individuals With Down Syndrome. Neurology 2024; 103:e209676. [PMID: 39074338 PMCID: PMC11286286 DOI: 10.1212/wnl.0000000000209676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/31/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral hemorrhages are an exclusion criterion and potential adverse effect of antiamyloid agents. It is, therefore, critical to characterize the natural history of cerebral microbleeds in populations genetically predisposed to Alzheimer disease (AD), such as Down syndrome (DS). We aimed to assess microbleed emergence in adults with DS across the AD spectrum, defining their topography and associations with clinical variables, cognitive outcomes, and fluid and neuroimaging biomarkers. METHODS This cross-sectional study included participants aged 18 years or older from the Down-Alzheimer Barcelona Neuroimaging Initiative and Sant Pau Initiative on Neurodegeneration with T1-weighted and susceptibility-weighted images. Participants underwent comprehensive assessments, including apolipoprotein E (APOE) genotyping; fluid and plasma determinations of beta-amyloid, tau, and neurofilament light; cognitive outcomes (Cambridge Cognitive Examination and modified Cued Recall Test); and vascular risk factors (hypertension, diabetes mellitus, and dyslipidemia). We manually segmented microbleeds and characterized their topography. Associations between microbleed severity and AD biomarkers were explored using between-group comparisons (none vs 1 vs 2+) and multivariate linear models. RESULTS We included 276 individuals with DS and 158 healthy euploid controls (mean age = 47.8 years, 50.92% female). Individuals with DS were more likely to have microbleeds than controls (20% vs 8.9%, p < 0.001), with more severe presentation (12% with 2+ vs 1.9%). Microbleeds increased with age (12% 20-30 years vs 60% > 60 years) and AD clinical stage (12.42% asymptomatic, 27.9% prodromal, 35.09% dementia) were more common in APOEε4 carriers (26% vs 18.3% noncarriers, p = 0.008), but not associated with vascular risk factors (p > 0.05). Microbleeds were predominantly posterior (cerebellum 33.66%; occipital 14.85%; temporal 21.29%) in participants with DS. Associations with microbleed severity were found for neuroimaging and fluid AD biomarkers, but only hippocampal volumes (standardized β = -0.18 [-0.31, -0.06], p < 0.005) and CSF p-tau-181 concentrations (β = 0.26 [0.12, 0.41], p < 0.005) survived regression controlling for age and disease stage, respectively. Microbleeds had limited effect on cognitive outcomes. DISCUSSION In participants with DS, microbleeds present with a posterior, lobar predominance, are associated with disease severity, but do not affect cognitive performance. These results suggest an interplay between AD pathology and vascular lesions, implicating microbleeds as a risk factor limiting the use of antiamyloid agents in this population.
Collapse
Affiliation(s)
- Sara E Zsadanyi
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alejandra O Morcillo-Nieto
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Mateus R Aranha
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Irina Aragón
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - José E Arriola-Infante
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Victor Montal
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Jordi Pegueroles
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Javier Arranz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Íñigo Rodríguez-Baz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lucia M Blesa
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laura Videla
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Isabel Barroeta
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laura Del Hoyo Soriano
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Bessy Benejam
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Susana Fernández
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Aida S Hernandez
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Nuria Bargallo
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Sofía González-Ortiz
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Sandra Giménez
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Daniel Alcolea
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Olivia Belbin
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alberto Lleó
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Juan Fortea
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Carmona-Iragui
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Alexandre Bejanin
- From the Sant Pau Memory Unit (S.E.Z., A.O.M.-N., M.R.A., I.A., J.E.A.-I., L.V.-A., V.M., J.P., J.A., Í.R.-B., L.M.B., L.V., I.B., L.H.S., B.B., A.S.H., S.G., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona; Department of Medicine (L.V.-A., N.B.), Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED) (V.M., J.P., L.V., I.B., D.A., O.B., A.L., J.F., M.C.-I., A.B.), Madrid; Barcelona Down Medical Center (L.V., B.B., S.F., A.S.H., J.F., M.C.-I.), Fundació Catalana Síndrome de Down; Radiology department (N.B., S.G.-O.), Centre de Diagnostic per la Imatge. Hospital Clínic de Barcelona; and Multidisciplinary Sleep Unit. Hospital de la Santa Creu i Sant Pau (S.G.), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| |
Collapse
|
5
|
Bezerra B, Fisher M, Pirih FQ, Casarin M. The potential impact of periodontitis on cerebral small vessel disease. Mol Oral Microbiol 2024; 39:190-198. [PMID: 37929810 DOI: 10.1111/omi.12443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/11/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
Cerebral small vessel disease (CSVD) is a term used to describe abnormalities in the intracranial microvasculature affecting small arteries, arterioles, capillaries, and venules. The etiology of these conditions is not fully understood but inflammation appears to play a significant role. Periodontal diseases have been associated with conditions such as stroke and dementia, which are clinical consequences of CSVD. Periodontitis is a highly prevalent chronic multifactorial inflammatory disease regulated by the host immune response against pathogenic bacterial colonization around the teeth. The inflammatory response and the microbial dysbiosis produce pro-inflammatory cytokines that can reach the brain and promote local changes. This review will explore the potential association between periodontitis and CSVD by assessing the impact of periodontitis-induced inflammation and periodontopathogenic bacteria on the underlying mechanisms leading to CSVD. Given the association of periodontitis with stroke and dementia, which are clinical features of CSVD, it may be possible to suggest a link with CSVD. Current evidence linking periodontitis with neuroimaging findings of CSVD enforces the possible link between these conditions.
Collapse
Affiliation(s)
- Beatriz Bezerra
- Section of Periodontics, UCLA School of Dentistry, Los Angeles, California, USA
| | - Mark Fisher
- Department of Neurology, UC Irvine Medical Center, Orange, California, USA
| | - Flavia Q Pirih
- Section of Periodontics, UCLA School of Dentistry, Los Angeles, California, USA
| | - Maísa Casarin
- Section of Periodontics, UCLA School of Dentistry, Los Angeles, California, USA
- School of Dentistry, Federal University of Pelotas, Pelotas, Brazil
| |
Collapse
|
6
|
Theodorou A, Tsantzali I, Stefanou MI, Sacco S, Katsanos AH, Shoamanesh A, Karapanayiotides T, Koutroulou I, Stamati P, Werring DJ, Cordonnier C, Palaiodimou L, Zompola C, Boviatsis E, Stavrinou L, Frantzeskaki F, Steiner T, Alexandrov AV, Paraskevas GP, Tsivgoulis G. CSF and plasma biomarkers in cerebral amyloid angiopathy: A single-center study and a systematic review/meta-analysis. Eur Stroke J 2024:23969873241260538. [PMID: 38869035 DOI: 10.1177/23969873241260538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION There are limited data regarding cerebrospinal fluid (CSF) and plasma biomarkers among patients with Cerebral Amyloid Angiopathy (CAA). We sought to investigate the levels of four biomarkers [β-amyloids (Aβ42 and Aβ40), total tau (tau) and phosphorylated tau (p-tau)] in CAA patients compared to healthy controls (HC) and patients with Alzheimer Disease (AD). PATIENTS AND METHODS A systematic review and meta-analysis of published studies, including also a 5 year single-center cohort study, with available data on CSF and plasma biomarkers in symptomatic sporadic CAA versus HC and AD was conducted. Biomarkers' comparisons were investigated using random-effects models based on the ratio of mean (RoM) biomarker concentrations. RoM < 1 and RoM > 1 indicate lower and higher biomarker concentration in CAA compared to another population, respectively. RESULTS We identified nine cohorts, comprising 327 CAA patients (mean age: 71 ± 5 years; women: 45%) versus 336 HC (mean age: 65 ± 5 years; women: 45%) and 384 AD patients (mean age: 68 ± 3 years; women: 53%) with available data on CSF biomarkers. CSF Aβ42 levels [RoM: 0.47; 95% CI: 0.36-0.62; p < 0.0001], Aβ40 levels [RoM: 0.70; 95% CI: 0.63-0.79; p < 0.0001] and the ratio Aβ42/Aβ40 [RoM: 0.62; 95% CI: 0.39-0.98; p = 0.0438] differentiated CAA from HC. CSF Aβ40 levels [RoM: 0.73; 95% CI: 0.64-0.83; p = 0.0003] differentiated CAA from AD. CSF tau and p-tau levels differentiated CAA from HC [RoM: 1.71; 95% CI: 1.41-2.09; p = 0.0002 and RoM: 1.44; 95% CI: 1.20-1.73; p = 0.0014, respectively] and from AD [RoM: 0.65; 95% CI: 0.58-0.72; p < 0.0001 and RoM: 0.64; 95% CI: 0.57-0.71; p < 0.0001, respectively]. Plasma Aβ42 [RoM: 1.14; 95% CI: 0.89-1.45; p = 0.2079] and Aβ40 [RoM: 1.07; 95% CI: 0.91-1.25; p = 0.3306] levels were comparable between CAA and HC. CONCLUSIONS CAA is characterized by a distinct CSF biomarker pattern compared to HC and AD. CSF Aβ40 levels are lower in CAA compared to HC and AD, while tau and p-tau levels are higher in CAA compared to HC, but lower in comparison to AD patients.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Tsantzali
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Simona Sacco
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Italy
| | - Aristeidis H Katsanos
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Ashkan Shoamanesh
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Theodoros Karapanayiotides
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Ioanna Koutroulou
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Polyxeni Stamati
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, Larissa, Greece
| | - David J Werring
- Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Charlotte Cordonnier
- University Lille, Inserm, CHU Lille, U1172, LilNCog, Lille Neuroscience and Cognition, France
| | - Lina Palaiodimou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Zompola
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Boviatsis
- Second Department of Neurosurgery, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Lampis Stavrinou
- Second Department of Neurosurgery, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Frantzeska Frantzeskaki
- Second Critical Care Department, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Thorsten Steiner
- Departments of Neurology, Klinikum Frankfurt Höchst, Frankfurt and Heidelberg University Hospital, Heidelberg, Germany
| | - Andrei V Alexandrov
- Department of Neurology, University of Arizona, Banner University Medical Center, Phoenix
| | - Georgios P Paraskevas
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
7
|
Ohtsuki S. Insulin receptor at the blood-brain barrier: Transport and signaling. VITAMINS AND HORMONES 2024; 126:113-124. [PMID: 39029970 DOI: 10.1016/bs.vh.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
The blood-brain barrier (BBB) is a unique system of the brain microvasculature that limits the exchange between the blood and the brain. Brain microvascular endothelial cells form the BBB as part of the neurovascular unit and express insulin receptors. The insulin receptor at the BBB has been studied in two different functional aspects. These functions include (1) the supplying of blood insulin to the brain and (2) the modulation of BBB function via insulin signaling. The first function involves drug delivery to the brain, while the second function is related to the association between central nervous system diseases and type 2 diabetes through insulin resistance. This chapter summarizes recent progress in research on the function of insulin receptors at the BBB.
Collapse
Affiliation(s)
- Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
8
|
De Kort AM, Kaushik K, Kuiperij HB, Jäkel L, Li H, Tuladhar AM, Terwindt GM, Wermer MJH, Claassen JAHR, Klijn CJM, Verbeek MM, Kessels RPC, Schreuder FHBM. The relation of a cerebrospinal fluid profile associated with Alzheimer's disease with cognitive function and neuropsychiatric symptoms in sporadic cerebral amyloid angiopathy. Alzheimers Res Ther 2024; 16:99. [PMID: 38704569 PMCID: PMC11069247 DOI: 10.1186/s13195-024-01454-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/07/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Patients with sporadic cerebral amyloid angiopathy (sCAA) frequently report cognitive or neuropsychiatric symptoms. The aim of this study is to investigate whether in patients with sCAA, cognitive impairment and neuropsychiatric symptoms are associated with a cerebrospinal fluid (CSF) biomarker profile associated with Alzheimer's disease (AD). METHODS In this cross-sectional study, we included participants with sCAA and dementia- and stroke-free, age- and sex-matched controls, who underwent a lumbar puncture, brain MRI, cognitive assessments, and self-administered and informant-based-questionnaires on neuropsychiatric symptoms. CSF phosphorylated tau, total tau and Aβ42 levels were used to divide sCAA patients in two groups: CAA with (CAA-AD+) or without a CSF biomarker profile associated with AD (CAA-AD-). Performance on global cognition, specific cognitive domains (episodic memory, working memory, processing speed, verbal fluency, visuoconstruction, and executive functioning), presence and severity of neuropsychiatric symptoms, were compared between groups. RESULTS sCAA-AD+ (n=31; mean age: 72 ± 6; 42%, 61% female) and sCAA-AD- (n=23; 70 ± 5; 42% female) participants did not differ with respect to global cognition or type of affected cognitive domain(s). The number or severity of neuropsychiatric symptoms also did not differ between sCAA-AD+ and sCAA-AD- participants. These results did not change after exclusion of patients without prior ICH. CONCLUSIONS In participants with sCAA, a CSF biomarker profile associated with AD does not impact global cognition or specific cognitive domains, or the presence of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Anna M De Kort
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kanishk Kaushik
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke Jäkel
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hao Li
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marieke J H Wermer
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jurgen A H R Claassen
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Catharina J M Klijn
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Roy P C Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
- Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
9
|
Naftali J, Tsur G, Auriel E, Barnea R, Findler M, Raphaeli G, Brauner R, Pardo K, Perlow A, Weinstein G, Weiss P, Glik A, Keret O. Impact of dementia status on intravenous thrombolysis and endovascular treatment for acute ischemic stroke: Retrospective study. J Neurol Sci 2024; 459:122954. [PMID: 38461762 DOI: 10.1016/j.jns.2024.122954] [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: 12/16/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Individuals with dementia are underrepresented in interventional studies for acute ischemic stroke (AIS). This research gap creates a bias against their treatment in clinical practice. Our goal was to compare the safety and efficacy of intravenous-thrombolysis (t-PA) and endovascular treatment (EVT) in individuals with or without pre-AIS dementia. METHOD A retrospective study of AIS patients receiving t-PA or EVT between 2019 and 2022. Patients were classified as dementia on a case-by-case review of baseline assessment. Additional variables included demographic, vascular risk factors, AIS severity and treatment. Outcomes of interest were intracerebral hemorrhage, mortality in 90-days, and the difference in modified rankin scale (mRS) before AIS and in 90-days follow-up. Outcomes were compared across non-matched groups and following propensity-score matching. RESULTS Altogether, 628 patients were included, of which 68 had pre-AIS dementia. Compared to non-dementia group, dementia group were older, had a higher rate of vascular risk factors, higher pre-stroke mRS and higher baseline NIHSS. Individuals with dementia had higher rates of mortality (25% vs.11%,p < 0.01) on non-matched comparison. All cohort and restricted t-PA EVT matched analysis showed no difference in any outcome. Regression analysis confirmed that AIS severity at presentation and its treatment, not dementia, were the chief contributors to patients' outcomes. DISCUSSION Our results indicate that pre-AIS dementia does not impact the efficacy or safety of EVT or t-PA for AIS. We thus call for more inclusive research on stroke therapy with regards to baseline cognitive status. Such studies are urgently required to inform stroke guidelines and enhance care.
Collapse
Affiliation(s)
- Jonathan Naftali
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.
| | - Gal Tsur
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel
| | - Eitan Auriel
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Rani Barnea
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Michael Findler
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Interventional Neuroradiology Unit, Rabin Medical Center, Petach Tikva, Israel
| | - Guy Raphaeli
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Interventional Neuroradiology Unit, Rabin Medical Center, Petach Tikva, Israel
| | - Ran Brauner
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Interventional Neuroradiology Unit, Rabin Medical Center, Petach Tikva, Israel
| | - Keshet Pardo
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel
| | - Alain Perlow
- Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Interventional Neuroradiology Unit, Rabin Medical Center, Petach Tikva, Israel
| | | | - Penina Weiss
- Occupational Therapy Department, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Amir Glik
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Cognitive Neurology Clinic, Rabin Medical Center, Petach Tikva, Israel
| | - Ophir Keret
- Department of Neurology, Rabin Medical Center, Petach Tikva, Israel; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel; Cognitive Neurology Clinic, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
10
|
Pillai JA, Bena J, Tousi B, Rothenberg K, Keene CD, Leverenz JB. Lewy body pathology modifies risk factors for cerebral amyloid angiopathy when comorbid with Alzheimer's disease pathology. Alzheimers Dement 2024; 20:2564-2574. [PMID: 38353367 PMCID: PMC11032524 DOI: 10.1002/alz.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/28/2023] [Accepted: 12/16/2023] [Indexed: 02/18/2024]
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) often accompanies dementia-associated pathologies and is important in the context of anti-amyloid monoclonal therapies and risk of hemorrhage. METHODS We conducted a retrospective neuropathology-confirmed study of 2384 participants in the National Alzheimer Coordinating Center cohort (Alzheimer's disease [AD], n = 1175; Lewy body pathology [LBP], n = 316; and mixed AD and LBP [AD-LBP], n = 893). We used logistic regression to evaluate age, sex, education, APOE ε4, neuritic plaques, and neurofibrillary tangles (NFTs) in CAA risk. RESULTS APOE ε4 increased CAA risk in all three groups, while younger age and higher NFT stages increased risk in AD and AD-LBP. In AD-LBP, male sex and lower education were additional risk factors. The odds of APOE ε4 carrier homozygosity related to CAA was higher in LBP (25.69) and AD-LBP (9.50) than AD (3.17). DISCUSSION AD and LBPs modify risk factors for CAA and should be considered in reviewing the risk of CAA. HIGHLIGHTS Lewy body pathology modifies risk factors for cerebral amyloid angiopathy (CAA) when present along with Alzheimer's disease (AD) neuropathology. In the context of anti-amyloid monoclonal therapies and their associated risks for hemorrhage, the risk of underlying CAA in mixed dementia with Lewy body pathology needs to be considered.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
- Department of NeurologyCleveland ClinicClevelandOhioUSA
| | - James Bena
- Quantitative Health SciencesCleveland ClinicClevelandOhioUSA
| | - Babak Tousi
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
| | - Kasia Rothenberg
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
| | - C. Dirk Keene
- Department of Laboratory Medicine and PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
- Department of NeurologyCleveland ClinicClevelandOhioUSA
| |
Collapse
|
11
|
Tsai HH, Liu CJ, Lee BC, Chen YF, Yen RF, Jeng JS, Tsai LK. Cerebral tau pathology in cerebral amyloid angiopathy. Brain Commun 2024; 6:fcae086. [PMID: 38638152 PMCID: PMC11024817 DOI: 10.1093/braincomms/fcae086] [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: 11/08/2023] [Revised: 02/01/2024] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Tau, a hallmark of Alzheimer's disease, is poorly characterized in cerebral amyloid angiopathy. We aimed to assess the clinico-radiological correlations between tau positron emission tomography scans and cerebral amyloid angiopathy. We assessed cerebral amyloid and hyperphosphorylated tau in patients with probable cerebral amyloid angiopathy (n = 31) and hypertensive small vessel disease (n = 27) using 11C-Pittsburgh compound B and 18F-T807 positron emission tomography. Multivariable regression models were employed to assess radio-clinical features related to cerebral tau pathology in cerebral amyloid angiopathy. Cerebral amyloid angiopathy exhibited a higher cerebral tau burden in the inferior temporal lobe [1.25 (1.17-1.42) versus 1.08 (1.05-1.22), P < 0.001] and all Braak stage regions of interest (P < 0.05) than hypertensive small vessel disease, although the differences were attenuated after age adjustment. Cerebral tau pathology was significantly associated with cerebral amyloid angiopathy-related vascular markers, including cortical superficial siderosis (β = 0.12, 95% confidence interval 0.04-0.21) and cerebral amyloid angiopathy score (β = 0.12, 95% confidence interval 0.03-0.21) after adjustment for age, ApoE4 status and whole cortex amyloid load. Tau pathology correlated significantly with cognitive score (Spearman's ρ=-0.56, P = 0.001) and hippocampal volume (-0.49, P = 0.007), even after adjustment. In conclusion, tau pathology is more frequent in sporadic cerebral amyloid angiopathy than in hypertensive small vessel disease. Cerebral amyloid angiopathy-related vascular pathologies, especially cortical superficial siderosis, are potential markers of cerebral tau pathology suggestive of concomitant Alzheimer's disease.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chia-Ju Liu
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Bo-Ching Lee
- Department of Medical Imaging, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Ya-Fang Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| |
Collapse
|
12
|
Wheeler KV, Irimia A, Braskie MN. Using Neuroimaging to Study Cerebral Amyloid Angiopathy and Its Relationship to Alzheimer's Disease. J Alzheimers Dis 2024; 97:1479-1502. [PMID: 38306032 DOI: 10.3233/jad-230553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β aggregation in the media and adventitia of the leptomeningeal and cortical blood vessels. CAA is one of the strongest vascular contributors to Alzheimer's disease (AD). It frequently co-occurs in AD patients, but the relationship between CAA and AD is incompletely understood. CAA may drive AD risk through damage to the neurovascular unit and accelerate parenchymal amyloid and tau deposition. Conversely, early AD may also drive CAA through cerebrovascular remodeling that impairs blood vessels from clearing amyloid-β. Sole reliance on autopsy examination to study CAA limits researchers' ability to investigate CAA's natural disease course and the effect of CAA on cognitive decline. Neuroimaging allows for in vivo assessment of brain function and structure and can be leveraged to investigate CAA staging and explore its associations with AD. In this review, we will discuss neuroimaging modalities that can be used to investigate markers associated with CAA that may impact AD vulnerability including hemorrhages and microbleeds, blood-brain barrier permeability disruption, reduced cerebral blood flow, amyloid and tau accumulation, white matter tract disruption, reduced cerebrovascular reactivity, and lowered brain glucose metabolism. We present possible areas for research inquiry to advance biomarker discovery and improve diagnostics.
Collapse
Affiliation(s)
- Koral V Wheeler
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Corwin D. Denney Research Center, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| |
Collapse
|
13
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [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] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
14
|
Xu H, Luo Z, Zhang R, Golovynska I, Huang Y, Samanta S, Zhou T, Li S, Guo B, Liu L, Weng X, He J, Liao C, Wang Y, Ohulchanskyy TY, Qu J. Exploring the effect of photobiomodulation and gamma visual stimulation induced by 808 nm and visible LED in Alzheimer's disease mouse model. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 250:112816. [PMID: 38029664 DOI: 10.1016/j.jphotobiol.2023.112816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
Although photobiomodulation (PBM) and gamma visual stimulatqion (GVS) have been overwhelmingly explored in the recent time as a possible light stimulation (LS) means of Alzheimer's disease (AD) therapy, their effects have not been assessed at once. In our research, the AD mouse model was stimulated using light with various parameters [continuous wave (PBM) or 40 Hz pulsed visible LED (GVS) or 40 Hz pulsed 808 nm LED (PBM and GVS treatment)]]. The brain slices collected from the LS treated AD model mice were evaluated using (i) fluorescence microscopy to image thioflavine-S labeled amy-loid-β (Aβ) plaques (the main hallmark of AD), or (ii) two-photon excited fluorescence (TPEF) imaging of unlabeled Aβ plaques, showing that the amount of Aβ plaques was reduced after LS treatment. The imaging results correlated well with the results of Morris water maze (MWM) test, which demonstrated that the spatial learning and memory abilities of LS treated mice were noticeably higher than those of untreated mice. The LS effect was also assessed by in vivo nonlinear optical imaging, revealing that the cerebral amyloid angiopathy decreased spe-cifically as a result of 40 Hz pulsed 808 nm irradiation, on the contrary, the angiopathy reversed after visible 40 Hz pulsed light treatment. The obtained results provide useful reference for further optimization of the LS (PBM or GVS) parameters to achieve efficient phototherapy of AD.
Collapse
Affiliation(s)
- Hao Xu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Ziyi Luo
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Renlong Zhang
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Iuliia Golovynska
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Yanxia Huang
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Soham Samanta
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Ting Zhou
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Shaowei Li
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Bingang Guo
- HOLOKOOK Co. LtD, Shenzhen 518060, Guangdong Province, P.R. China
| | - Liwei Liu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Xiaoyu Weng
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Jun He
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Changrui Liao
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Yiping Wang
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China
| | - Tymish Y Ohulchanskyy
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China.
| | - Junle Qu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, Guangdong Province, P.R. China; Engineering Research Center of Optical Instrument and System, Ministry of Education, Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R. China.
| |
Collapse
|
15
|
Chen N, Jiang F, Chen X, Zhu L, Qiao N, Zhou J, Zhang Y. Associations of Lipoprotein(a) Level with Cerebral Small Vessel Disease in Patients with Alzheimer's Disease. Brain Sci 2023; 14:34. [PMID: 38248249 PMCID: PMC10813431 DOI: 10.3390/brainsci14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND This study aimed to examine the association of lipoprotein(a) [Lp(a)] level with the burden of cerebral small vessel disease (CSVD) in patients with Alzheimer's disease (AD). METHODS Data from 111 consecutive patients with AD admitted to Nanjing First Hospital from 2015 to 2022 were retrospectively analyzed in this study. Serum Lp(a) concentrations were grouped into tertiles (T1-T3). Brain magnetic resonance imaging (MRI) was rated for the presence of CSVD, including enlarged perivascular spaces (EPVS), lacunes, white-matter lesions, and cerebral microbleeds (CMBs). The CSVD burden was calculated by summing the scores of each MRI marker at baseline. A binary or ordinal logistic regression model was used to estimate the relationship of serum Lp(a) levels with CSVD burden and each MRI marker. RESULTS Patients with higher tertiles of Lp(a) levels were less likely to have any CSVD (T1, 94.6%; T2, 78.4%; T3, 66.2%; p = 0.013). Multivariable analysis found that Lp(a) levels were inversely associated with the presence of CSVD (T2 vs. T1: adjusted odds ratio [aOR] 0.132, 95% confidence interval [CI] 0.018-0.946, p = 0.044; T3 vs. T1: aOR 0.109, 95% CI 0.016-0.737, p = 0.023) and CSVD burden (T3 vs. T1: aOR 0.576, 95% CI 0.362-0.915, p = 0.019). The independent relationship between Lp(a) levels and individual CSVD features was significant for moderate-to-severe EPVS in the centrum semiovale (T2 vs. T1: aOR 0.059, 95% CI 0.006-0.542, p = 0.012; T3 vs. T1: aOR 0.029, 95% CI 0.003-0.273, p = 0.002) and CMBs (T3 vs. T1: aOR 0.144, 95% CI 0.029-0.716, p = 0.018). CONCLUSIONS In this study, serum Lp(a) level was inversely associated with CSVD in AD patients.
Collapse
Affiliation(s)
- Nihong Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
- Department of Neurology, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing 210039, China;
| | - Fuping Jiang
- Department of Geriatrics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China;
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Na Qiao
- Department of Neurology, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing 210039, China;
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| |
Collapse
|
16
|
Wu W, Huang J, Han P, Zhang J, Wang Y, Jin F, Zhou Y. Research Progress on Natural Plant Molecules in Regulating the Blood-Brain Barrier in Alzheimer's Disease. Molecules 2023; 28:7631. [PMID: 38005352 PMCID: PMC10674591 DOI: 10.3390/molecules28227631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder. With the aging population and the continuous development of risk factors associated with AD, it will impose a significant burden on individuals, families, and society. Currently, commonly used therapeutic drugs such as Cholinesterase inhibitors, N-methyl-D-aspartate antagonists, and multiple AD pathology removal drugs have been shown to have beneficial effects on certain pathological conditions of AD. However, their clinical efficacy is minimal and they are associated with certain adverse reactions. Furthermore, the underlying pathological mechanism of AD remains unclear, posing a challenge for drug development. In contrast, natural plant molecules, widely available, offer multiple targeting pathways and demonstrate inherent advantages in modifying the typical pathologic features of AD by influencing the blood-brain barrier (BBB). We provide a comprehensive review of recent in vivo and in vitro studies on natural plant molecules that impact the BBB in the treatment of AD. Additionally, we analyze their specific mechanisms to offer novel insights for the development of safe and effective targeted drugs as well as guidance for experimental research and the clinical application of drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Weidong Wu
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Jiahao Huang
- Department of Chinese Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Pengfei Han
- Science and Education Section, Zhangjiakou First Hospital, Zhangjiakou 075041, China;
| | - Jian Zhang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Yuxin Wang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Fangfang Jin
- Department of Internal Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yanyan Zhou
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| |
Collapse
|
17
|
Hampel H, Elhage A, Cho M, Apostolova LG, Nicoll JAR, Atri A. Amyloid-related imaging abnormalities (ARIA): radiological, biological and clinical characteristics. Brain 2023; 146:4414-4424. [PMID: 37280110 PMCID: PMC10629981 DOI: 10.1093/brain/awad188] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/08/2023] Open
Abstract
Excess accumulation and aggregation of toxic soluble and insoluble amyloid-β species in the brain are a major hallmark of Alzheimer's disease. Randomized clinical trials show reduced brain amyloid-β deposits using monoclonal antibodies that target amyloid-β and have identified MRI signal abnormalities called amyloid-related imaging abnormalities (ARIA) as possible spontaneous or treatment-related adverse events. This review provides a comprehensive state-of-the-art conceptual review of radiological features, clinical detection and classification challenges, pathophysiology, underlying biological mechanism(s) and risk factors/predictors associated with ARIA. We summarize the existing literature and current lines of evidence with ARIA-oedema/effusion (ARIA-E) and ARIA-haemosiderosis/microhaemorrhages (ARIA-H) seen across anti-amyloid clinical trials and therapeutic development. Both forms of ARIA may occur, often early, during anti-amyloid-β monoclonal antibody treatment. Across randomized controlled trials, most ARIA cases were asymptomatic. Symptomatic ARIA-E cases often occurred at higher doses and resolved within 3-4 months or upon treatment cessation. Apolipoprotein E haplotype and treatment dosage are major risk factors for ARIA-E and ARIA-H. Presence of any microhaemorrhage on baseline MRI increases the risk of ARIA. ARIA shares many clinical, biological and pathophysiological features with Alzheimer's disease and cerebral amyloid angiopathy. There is a great need to conceptually link the evident synergistic interplay associated with such underlying conditions to allow clinicians and researchers to further understand, deliberate and investigate on the combined effects of these multiple pathophysiological processes. Moreover, this review article aims to better assist clinicians in detection (either observed via symptoms or visually on MRI), management based on appropriate use recommendations, and general preparedness and awareness when ARIA are observed as well as researchers in the fundamental understanding of the various antibodies in development and their associated risks of ARIA. To facilitate ARIA detection in clinical trials and clinical practice, we recommend the implementation of standardized MRI protocols and rigorous reporting standards. With the availability of approved amyloid-β therapies in the clinic, standardized and rigorous clinical and radiological monitoring and management protocols are required to effectively detect, monitor, and manage ARIA in real-world clinical settings.
Collapse
Affiliation(s)
- Harald Hampel
- Eisai Inc., Alzheimer’s Disease and Brain Health, Nutley, NJ 07110, USA
| | - Aya Elhage
- Eisai Inc., Alzheimer’s Disease and Brain Health, Nutley, NJ 07110, USA
| | - Min Cho
- Eisai Inc., Alzheimer’s Disease and Brain Health, Nutley, NJ 07110, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - James A R Nicoll
- Division of Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Alireza Atri
- Banner Sun Health Research Institute, Banner Health, Sun City, AZ 85351, USA
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Perosa V, Auger CA, Zanon Zotin MC, Oltmer J, Frosch MP, Viswanathan A, Greenberg SM, van Veluw SJ. Histopathological Correlates of Lobar Microbleeds in False-Positive Cerebral Amyloid Angiopathy Cases. Ann Neurol 2023; 94:856-870. [PMID: 37548609 DOI: 10.1002/ana.26761] [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: 03/16/2023] [Revised: 07/05/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVE A definite diagnosis of cerebral amyloid angiopathy (CAA), characterized by the accumulation of amyloid β in walls of cerebral small vessels, can only be obtained through pathological examination. A diagnosis of probable CAA during life relies on the presence of hemorrhagic markers, including lobar cerebral microbleeds (CMBs). The aim of this project was to study the histopathological correlates of lobar CMBs in false-positive CAA cases. METHODS In 3 patients who met criteria for probable CAA during life, but showed no CAA upon neuropathological examination, lobar CMBs were counted on ex vivo 3T magnetic resonance imaging (MRI) and on ex vivo 7T MRI. Areas with lobar CMBs were next sampled and cut into serial sections, on which the CMBs were then identified. RESULTS Collectively, there were 25 lobar CMBs on in vivo MRI and 22 on ex vivo 3T MRI of the analyzed hemispheres. On ex vivo MRI, we targeted 12 CMBs for sampling, and definite histopathological correlates were retrieved for 9 of them, of which 7 were true CMBs. No CAA was found on any of the serial sections. The "culprit vessels" associated with the true CMBs instead showed moderate to severe arteriolosclerosis. Furthermore, CMBs in false-positive CAA cases tended to be located more often in the juxtacortical or subcortical white matter than in the cortical ribbon. INTERPRETATION These findings suggest that arteriolosclerosis can generate lobar CMBs and that more detailed investigations into the exact localization of CMBs with respect to the cortical ribbon could potentially aid the diagnosis of CAA during life. ANN NEUROL 2023;94:856-870.
Collapse
Affiliation(s)
- Valentina Perosa
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Corinne A Auger
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Maria Clara Zanon Zotin
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Imaging Sciences and Medical Physics, Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Jan Oltmer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Matthew P Frosch
- Department of Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
19
|
Jucker M, Walker LC. Alzheimer's disease: From immunotherapy to immunoprevention. Cell 2023; 186:4260-4270. [PMID: 37729908 PMCID: PMC10578497 DOI: 10.1016/j.cell.2023.08.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Recent Aβ-immunotherapy trials have yielded the first clear evidence that removing aggregated Aβ from the brains of symptomatic patients can slow the progression of Alzheimer's disease. The clinical benefit achieved in these trials has been modest, however, highlighting the need for both a deeper understanding of disease mechanisms and the importance of intervening early in the pathogenic cascade. An immunoprevention strategy for Alzheimer's disease is required that will integrate the findings from clinical trials with mechanistic insights from preclinical disease models to select promising antibodies, optimize the timing of intervention, identify early biomarkers, and mitigate potential side effects.
Collapse
Affiliation(s)
- Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.
| | - Lary C Walker
- Department of Neurology and Emory National Primate Research Center, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
20
|
Zedde M, Grisendi I, Assenza F, Vandelli G, Napoli M, Moratti C, Lochner P, Seiffge DJ, Piazza F, Valzania F, Pascarella R. The Venular Side of Cerebral Amyloid Angiopathy: Proof of Concept of a Neglected Issue. Biomedicines 2023; 11:2663. [PMID: 37893037 PMCID: PMC10604278 DOI: 10.3390/biomedicines11102663] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Small vessel diseases (SVD) is an umbrella term including several entities affecting small arteries, arterioles, capillaries, and venules in the brain. One of the most relevant and prevalent SVDs is cerebral amyloid angiopathy (CAA), whose pathological hallmark is the deposition of amyloid fragments in the walls of small cortical and leptomeningeal vessels. CAA frequently coexists with Alzheimer's Disease (AD), and both are associated with cerebrovascular events, cognitive impairment, and dementia. CAA and AD share pathophysiological, histopathological and neuroimaging issues. The venular involvement in both diseases has been neglected, although both animal models and human histopathological studies found a deposition of amyloid beta in cortical venules. This review aimed to summarize the available information about venular involvement in CAA, starting from the biological level with the putative pathomechanisms of cerebral damage, passing through the definition of the peculiar angioarchitecture of the human cortex with the functional organization and consequences of cortical arteriolar and venular occlusion, and ending to the hypothesized links between cortical venular involvement and the main neuroimaging markers of the disease.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Ilaria Grisendi
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Federica Assenza
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Gabriele Vandelli
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Manuela Napoli
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Claudio Moratti
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - David J. Seiffge
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| |
Collapse
|
21
|
Tian M, Zuo C, Civelek AC, Carrio I, Watanabe Y, Kang KW, Murakami K, Garibotto V, Prior JO, Barthel H, Guan Y, Lu J, Zhou R, Jin C, Wu S, Zhang X, Zhong Y, Zhang H. International Nuclear Medicine Consensus on the Clinical Use of Amyloid Positron Emission Tomography in Alzheimer's Disease. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:375-389. [PMID: 37589025 PMCID: PMC10425321 DOI: 10.1007/s43657-022-00068-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 08/18/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia, with its diagnosis and management remaining challenging. Amyloid positron emission tomography (PET) has become increasingly important in medical practice for patients with AD. To integrate and update previous guidelines in the field, a task group of experts of several disciplines from multiple countries was assembled, and they revised and approved the content related to the application of amyloid PET in the medical settings of cognitively impaired individuals, focusing on clinical scenarios, patient preparation, administered activities, as well as image acquisition, processing, interpretation and reporting. In addition, expert opinions, practices, and protocols of prominent research institutions performing research on amyloid PET of dementia are integrated. With the increasing availability of amyloid PET imaging, a complete and standard pipeline for the entire examination process is essential for clinical practice. This international consensus and practice guideline will help to promote proper clinical use of amyloid PET imaging in patients with AD.
Collapse
Affiliation(s)
- Mei Tian
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
- Human Phenome Institute, Fudan University, Shanghai, 201203 China
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
- National Center for Neurological Disorders and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Ali Cahid Civelek
- Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins Medicine, Baltimore, 21287 USA
| | - Ignasi Carrio
- Department of Nuclear Medicine, Hospital Sant Pau, Autonomous University of Barcelona, Barcelona, 08025 Spain
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047 Japan
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Korea
| | - Koji Murakami
- Department of Radiology, Juntendo University Hospital, Tokyo, 113-8431 Japan
| | - Valentina Garibotto
- Diagnostic Department, University Hospitals of Geneva and NIMTlab, University of Geneva, Geneva, 1205 Switzerland
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, 1011 Switzerland
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig, 04103 Germany
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
| | - Jiaying Lu
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Shuang Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009 China
- The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, 310007 China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310007 China
| | - Molecular Imaging-Based Precision Medicine Task Group of A3 (China-Japan-Korea) Foresight Program
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
- Human Phenome Institute, Fudan University, Shanghai, 201203 China
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 China
- National Center for Neurological Disorders and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins Medicine, Baltimore, 21287 USA
- Department of Nuclear Medicine, Hospital Sant Pau, Autonomous University of Barcelona, Barcelona, 08025 Spain
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047 Japan
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Korea
- Department of Radiology, Juntendo University Hospital, Tokyo, 113-8431 Japan
- Diagnostic Department, University Hospitals of Geneva and NIMTlab, University of Geneva, Geneva, 1205 Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, 1011 Switzerland
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig, 04103 Germany
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009 China
- The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, 310007 China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310007 China
| |
Collapse
|
22
|
Pinho J, Almeida FC, Araújo JM, Machado Á, Costa AS, Silva F, Francisco A, Quintas-Neves M, Ferreira C, Soares-Fernandes JP, Oliveira TG. Sex-Specific Patterns of Cerebral Atrophy and Enlarged Perivascular Spaces in Patients with Cerebral Amyloid Angiopathy and Dementia. AJNR Am J Neuroradiol 2023; 44:792-798. [PMID: 37290817 PMCID: PMC10337609 DOI: 10.3174/ajnr.a7900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/07/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral amyloid angiopathy is characterized by amyloid β deposition in leptomeningeal and superficial cortical vessels. Cognitive impairment is common and may occur independent of concomitant Alzheimer disease neuropathology. It is still unknown which neuroimaging findings are associated with dementia in cerebral amyloid angiopathy and whether they are modulated by sex. This study compared MR imaging markers in patients with cerebral amyloid angiopathy with dementia or mild cognitive impairment or who are cognitively unimpaired and explored sex-specific differences. MATERIALS AND METHODS We studied 58 patients with cerebral amyloid angiopathy selected from the cerebrovascular and memory outpatient clinics. Clinical characteristics were collected from clinical records. Cerebral amyloid angiopathy was diagnosed on MR imaging on the basis of the Boston criteria. Visual rating scores for atrophy and other imaging features were independently assessed by 2 senior neuroradiologists. RESULTS Medial temporal lobe atrophy was higher for those with cerebral amyloid angiopathy with dementia versus those cognitively unimpaired (P = .015), but not for those with mild cognitive impairment. This effect was mainly driven by higher atrophy in men with dementia, compared with women with and without dementia (P = .034, P = .012; respectively) and with men without dementia (P = .012). Enlarged perivascular spaces in the centrum semiovale were more frequent in women with dementia versus men with and without dementia (P = .021, P = .011; respectively) and women without dementia (P = .011). CONCLUSIONS Medial temporal lobe atrophy was more prominent in men with dementia, whereas women showed a higher number of enlarged perivascular spaces in the centrum semiovale. Overall, this finding suggests differential pathophysiologic mechanisms with sex-specific neuroimaging patterns in cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- J Pinho
- From the Department of Neurology (J.P., A.S.C.), University Hospital RWTH Aachen, Aachen, Germany
| | - F C Almeida
- Life and Health Sciences Research Institute (F.C.A., M.Q.-N., T.G.O.), School of Medicine
- Life and Health Sciences Research Institute/3Bs (F.C.A., M.Q.-N., T.G.O.), Portuguese Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Neuroradiology (F.C.A.), Centro Hospitalar Universitxrário do Porto, Porto, Portugal
| | - J M Araújo
- Departments of Neurology (J.M.A., Á.M., C.F.)
| | - Á Machado
- Departments of Neurology (J.M.A., Á.M., C.F.)
| | - A S Costa
- From the Department of Neurology (J.P., A.S.C.), University Hospital RWTH Aachen, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging (A.S.C.), Forschungszentrum Jülich and RWTH Aachen University, Aachen, Germany
| | - F Silva
- Algoritmi Center (F.S., A.F.), University of Minho, Braga, Portugal
| | - A Francisco
- Algoritmi Center (F.S., A.F.), University of Minho, Braga, Portugal
| | - M Quintas-Neves
- Life and Health Sciences Research Institute (F.C.A., M.Q.-N., T.G.O.), School of Medicine
- Life and Health Sciences Research Institute/3Bs (F.C.A., M.Q.-N., T.G.O.), Portuguese Government Associate Laboratory, Braga/Guimarães, Portugal
- Neuroradiology (M.Q.-N., J.P.S.-F., T.G.O.), Hospital de Braga, Braga, Portugal
| | - C Ferreira
- Departments of Neurology (J.M.A., Á.M., C.F.)
| | | | - T G Oliveira
- Life and Health Sciences Research Institute (F.C.A., M.Q.-N., T.G.O.), School of Medicine
- Life and Health Sciences Research Institute/3Bs (F.C.A., M.Q.-N., T.G.O.), Portuguese Government Associate Laboratory, Braga/Guimarães, Portugal
- Neuroradiology (M.Q.-N., J.P.S.-F., T.G.O.), Hospital de Braga, Braga, Portugal
| |
Collapse
|
23
|
Kelly L, Sharp MM, Thomas I, Brown C, Schrag M, Antunes LV, Solopova E, Martinez-Gonzalez J, Rodríguez C, Carare RO. Targeting lysyl-oxidase (LOX) may facilitate intramural periarterial drainage for the treatment of Alzheimer's disease. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100171. [PMID: 37457664 PMCID: PMC10338210 DOI: 10.1016/j.cccb.2023.100171] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease is the commonest form of dementia. It is likely that a lack of clearance of amyloid beta (Aβ) results in its accumulation in the parenchyma as Aβ oligomers and insoluble plaques, and within the walls of blood vessels as cerebral amyloid angiopathy (CAA). The drainage of Aβ along the basement membranes of blood vessels as intramural periarterial drainage (IPAD), could be improved if the driving force behind IPAD could be augmented, therefore reducing Aβ accumulation. There are alterations in the composition of the vascular basement membrane in Alzheimer's disease. Lysyl oxidase (LOX) is an enzyme involved in the remodelling of the extracellular matrix and its expression and function is altered in various disease states. The expression of LOX is increased in Alzheimer's disease, but it is unclear whether this is a contributory factor in the impairment of IPAD in Alzheimer's disease. The pharmacological inhibition of LOX may be a strategy to improve IPAD and reduce the accumulation of Aβ in the parenchyma and within the walls of blood vessels.
Collapse
Affiliation(s)
- Louise Kelly
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | | | | | - Christopher Brown
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Lissa Ventura Antunes
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Elena Solopova
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - José Martinez-Gonzalez
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | | |
Collapse
|
24
|
Nagaraja N, DeKosky S, Duara R, Kong L, Wang WE, Vaillancourt D, Albayram M. Imaging features of small vessel disease in cerebral amyloid angiopathy among patients with Alzheimer's disease. Neuroimage Clin 2023; 38:103437. [PMID: 37245492 PMCID: PMC10236212 DOI: 10.1016/j.nicl.2023.103437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/07/2023] [Accepted: 05/14/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral small vessel disease biomarkers including white matter hyperintensities (WMH), lacunes, and enlarged perivascular spaces (ePVS) are under investigation to identify those specific to cerebral amyloid angiopathy (CAA). In subjects with Alzheimer's disease (AD), we assessed characteristic features and amounts of WMH, lacunes, and ePVS in four CAA categories (no, mild, moderate and severe CAA) and correlated these with Clinical Dementia Rating sum of boxes (CDRsb) score, ApoE genotype, and neuropathological changes at autopsy. METHODS The study included patients with a clinical diagnosis of dementia due to AD and neuropathological confirmation of AD and CAA in the National Alzheimer's Coordinating Center (NACC) database. The WMH, lacunes, and ePVS were evaluated using semi-quantitative scales. Statistical analyses compared the WMH, lacunes, and ePVS values in the four CAA groups with vascular risk factors and AD severity treated as covariates, and to correlate the imaging features with CDRsb score, ApoE genotype, and neuropathological findings. RESULTS The study consisted of 232 patients, of which 222 patients had FLAIR data available and 105 patients had T2-MRI. Occipital predominant WMH were significantly associated with the presence of CAA (p = 0.007). Among the CAA groups, occipital predominant WMH was associated with severe CAA (β = 1.22, p = 0.0001) compared with no CAA. Occipital predominant WMH were not associated with the CDRsb score performed at baseline (p = 0.68) or at follow-up 2-4 years after the MRI (p = 0.92). There was no significant difference in high grade ePVS in the basal ganglia (p = 0.63) and centrum semiovale (p = 0.95) among the four CAA groups. The WMH and ePVS on imaging did not correlate with the number of ApoE ε4 alleles but the WMH (periventricular and deep) correlated with the presence of infarcts, lacunes and microinfarcts on neuropathology. CONCLUSION Among patients with AD, occipital predominant WMH is more likely to be found in patients with severe CAA than in those without CAA. The high-grade ePVS in centrum semiovale were common in all AD patients regardless of CAA severity.
Collapse
Affiliation(s)
- Nandakumar Nagaraja
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Steven DeKosky
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Ranjan Duara
- Department of Neurology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Lan Kong
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, PA, USA
| | - Wei-En Wang
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - David Vaillancourt
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Mehmet Albayram
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
25
|
Castellani RJ, Shanes E, McCord M, Reish NJ, Flanagan ME, Mesulam MM, Jamshidi P. Neuropathology of Anti-Amyloid-β Immunotherapy: A Case Report. J Alzheimers Dis 2023; 93:803-813. [PMID: 37125554 DOI: 10.3233/jad-221305] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Host responses to anti-amyloid-β (Aβ) antibody therapy are evident in neuroimaging changes and clinical symptoms in a subset of clinical trial subjects receiving such therapy. The pathological basis for the imaging changes and clinical symptoms is not known, nor is the precise mechanism of Aβ clearing. We report the autopsy findings in a 65-year-old woman who received three open label infusions of the experimental anti-Aβ drug lecanemab over about one month. Four days after the last infusion, she was treated with tissue plasminogen activator for acute stroke symptoms and died several days later with multifocal hemorrhage. Neuropathological examination demonstrated histiocytic vasculitis involving blood vessels with cerebral amyloid angiopathy. Fragmentation and phagocytosis of vascular Aβ were present throughout the cerebral cortex. Phagocytosis of parenchymal Aβ plaques was noted. Changes suggestive of Aβ and phosphorylated tau "clearing" were also noted. The findings overall suggest that anti-Aβ treatment stimulated a host response to Aβ, i.e., target engagement. The findings also provide evidence that amyloid-related imaging abnormalities might be indicative of an Aβ phagocytic syndrome within cerebral vasculature and parenchymal brain tissue in some cases.
Collapse
Affiliation(s)
- Rudolph J Castellani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elisheva Shanes
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew McCord
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicholas J Reish
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Margaret E Flanagan
- Department of Pathology, University of Texas, San Antonio, San Antonio, TX, USA
| | - M-Marsel Mesulam
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pouya Jamshidi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
26
|
Badimon A, Torrente D, Norris EH. Vascular Dysfunction in Alzheimer's Disease: Alterations in the Plasma Contact and Fibrinolytic Systems. Int J Mol Sci 2023; 24:7046. [PMID: 37108211 PMCID: PMC10138543 DOI: 10.3390/ijms24087046] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, affecting millions of people worldwide. The classical hallmarks of AD include extracellular beta-amyloid (Aβ) plaques and neurofibrillary tau tangles, although they are often accompanied by various vascular defects. These changes include damage to the vasculature, a decrease in cerebral blood flow, and accumulation of Aβ along vessels, among others. Vascular dysfunction begins early in disease pathogenesis and may contribute to disease progression and cognitive dysfunction. In addition, patients with AD exhibit alterations in the plasma contact system and the fibrinolytic system, two pathways in the blood that regulate clotting and inflammation. Here, we explain the clinical manifestations of vascular deficits in AD. Further, we describe how changes in plasma contact activation and the fibrinolytic system may contribute to vascular dysfunction, inflammation, coagulation, and cognitive impairment in AD. Given this evidence, we propose novel therapies that may, alone or in combination, ameliorate AD progression in patients.
Collapse
Affiliation(s)
| | | | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
27
|
Pal I, Dey SG. The Role of Heme and Copper in Alzheimer's Disease and Type 2 Diabetes Mellitus. JACS AU 2023; 3:657-681. [PMID: 37006768 PMCID: PMC10052274 DOI: 10.1021/jacsau.2c00572] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 06/19/2023]
Abstract
Beyond the well-explored proposition of protein aggregation or amyloidosis as the central event in amyloidogenic diseases like Alzheimer's Disease (AD), and Type 2 Diabetes Mellitus (T2Dm); there are alternative hypotheses, now becoming increasingly evident, which suggest that the small biomolecules like redox noninnocent metals (Fe, Cu, Zn, etc.) and cofactors (Heme) have a definite influence in the onset and extent of such degenerative maladies. Dyshomeostasis of these components remains as one of the common features in both AD and T2Dm etiology. Recent advances in this course reveal that the metal/cofactor-peptide interactions and covalent binding can alarmingly enhance and modify the toxic reactivities, oxidize vital biomolecules, significantly contribute to the oxidative stress leading to cell apoptosis, and may precede the amyloid fibrils formation by altering their native folds. This perspective highlights this aspect of amyloidogenic pathology which revolves around the impact of the metals and cofactors in the pathogenic courses of AD and T2Dm including the active site environments, altered reactivities, and the probable mechanisms involving some highly reactive intermediates as well. It also discusses some in vitro metal chelation or heme sequestration strategies which might serve as a possible remedy. These findings might open up a new paradigm in our conventional understanding of amyloidogenic diseases. Moreover, the interaction of the active sites with small molecules elucidates potential biochemical reactivities that can inspire designing of drug candidates for such pathologies.
Collapse
Affiliation(s)
- Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick
Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick
Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
28
|
Guo MH, Vaishnavi SN. Clinical Management in Alzheimer’s Disease in the Era of Disease-Modifying Therapies. Curr Treat Options Neurol 2023. [DOI: 10.1007/s11940-023-00750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
29
|
van Dijk SE, Lak J, Drenth N, Hafkemeijer A, Rombouts SARB, van der Grond J, van Rooden S. Aging Effect, Reproducibility, and Test-Retest Reliability of a New Cerebral Amyloid Angiopathy MRI Severity Marker-Cerebrovascular Reactivity to Visual Stimulation. J Magn Reson Imaging 2023; 57:909-915. [PMID: 35876045 DOI: 10.1002/jmri.28362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Decreased cerebrovascular reactivity, measured as changes in blood-oxygen-level-dependent (BOLD) signal, is a potential new cerebral amyloid angiopathy (CAA) severity marker. Before clinical application, the effect of aging on BOLD parameters, and reproducibility and test-retest reliability of these parameters should be assessed. PURPOSE Assess the effect of healthy aging on cerebrovascular reactivity (BOLD amplitude, time to peak, and time to baseline). And determine reproducibility and test-retest reliability of these parameters. STUDY TYPE Prospective-observational. POPULATION Eighty-six healthy adults (mean age 56 years, 55% female), 10 presymptomatic D-CAA mutation carriers (mean age 34 years, 70% female), and 10 symptomatic D-CAA mutation carriers (mean age 54 years, 70% female). FIELD STRENGTH/SEQUENCE 3-T, three-dimensional (3D) T1-weighted MRI and gradient echo BOLD fMRI. ASSESSMENT To assess test-retest reliability of BOLD parameters, i.e. BOLD amplitude, time to peak, and time to baseline, BOLD fMRI scans were repeated three times immediately after each other, in both controls and mutation carriers. To assess reproducibility, BOLD fMRI scans were repeated with a 3-week interval for each subject. STATISTICAL TESTS Linear regression analyses and two-way mixed absolute agreement intra-class correlation approach. RESULTS Healthy aging was associated with decreased BOLD amplitude (β = -0.711) and prolonged time to baseline (β = 0.236) in the visual cortex after visual stimulation Reproducibility of BOLD amplitude was excellent (ICC 0.940) in the subgroup of healthy adults. Test-retest reliability for BOLD amplitude was excellent in healthy adults (ICC 0.856-0.910) and presymptomatic D-CAA mutation carriers (ICC 0.959-0.981). In symptomatic D-CAA mutation carriers, test-retest reliability was poor for all parameters (ICCs < 0.5). DATA CONCLUSION Healthy aging is associated with decreased cerebrovascular reactivity, measured by changes in BOLD response to visual stimulation. The BOLD amplitude appears to be a robust measurement in healthy adults and presymptomatic D-CAA mutation carriers, but not in symptomatic D-CAA mutation carriers.
Collapse
Affiliation(s)
- Suzanne E van Dijk
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessie Lak
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadieh Drenth
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Hafkemeijer
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Serge A R B Rombouts
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Institute of Psychology, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sanneke van Rooden
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Weiner MW, Veitch DP, Miller MJ, Aisen PS, Albala B, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nosheny R, Okonkwo OC, Perrin RJ, Petersen RC, Rivera‐Mindt M, Saykin AJ, Shaw LM, Toga AW, Tosun D, Trojanowski JQ. Increasing participant diversity in AD research: Plans for digital screening, blood testing, and a community-engaged approach in the Alzheimer's Disease Neuroimaging Initiative 4. Alzheimers Dement 2023; 19:307-317. [PMID: 36209495 PMCID: PMC10042173 DOI: 10.1002/alz.12797] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to validate biomarkers for Alzheimer's disease (AD) clinical trials. To improve generalizability, ADNI4 aims to enroll 50-60% of its new participants from underrepresented populations (URPs) using new biofluid and digital technologies. ADNI4 has received funding from the National Institute on Aging beginning September 2022. METHODS ADNI4 will recruit URPs using community-engaged approaches. An online portal will screen 20,000 participants, 4000 of whom (50-60% URPs) will be tested for plasma biomarkers and APOE. From this, 500 new participants will undergo in-clinic assessment joining 500 ADNI3 rollover participants. Remaining participants (∼3500) will undergo longitudinal plasma and digital cognitive testing. ADNI4 will add MRI sequences and new PET tracers. Project 1 will optimize biomarkers in AD clinical trials. RESULTS AND DISCUSSION ADNI4 will improve generalizability of results, use remote digital and blood screening, and continue providing longitudinal clinical, biomarker, and autopsy data to investigators.
Collapse
Affiliation(s)
- Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Dallas P. Veitch
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Northern California Institute for Research and Education (NCIRE)Department of Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
| | - Melanie J. Miller
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Northern California Institute for Research and Education (NCIRE)Department of Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Bruce Albala
- Department of NeurologyUniversity of California Irvine School of MedicineIrvineCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's Hospital, Broad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera‐Mindt
- Department of PsychologyLatin American and Latino Studies Institute, & African and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisINUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | |
Collapse
|
31
|
Cerebral Superficial Siderosis. Clin Neuroradiol 2022; 33:293-306. [DOI: 10.1007/s00062-022-01231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
AbstractSuperficial siderosis (SS) of the central nervous system constitutes linear hemosiderin deposits in the leptomeninges and the superficial layers of the cerebrum and the spinal cord. Infratentorial (i) SS is likely due to recurrent or continuous slight bleeding into the subarachnoid space. It is assumed that spinal dural pathologies often resulting in cerebrospinal fluid (CSF) leakage is the most important etiological group which causes iSS and detailed neuroradiological assessment of the spinal compartment is necessary. Further etiologies are neurosurgical interventions, trauma and arteriovenous malformations. Typical neurological manifestations of this classical type of iSS are slowly progressive sensorineural hearing impairment and cerebellar symptoms, such as ataxia, kinetic tremor, nystagmus and dysarthria. Beside iSS, a different type of SS restricted to the supratentorial compartment can be differentiated, i.e. cortical (c) SS, especially in older people often due to cerebral amyloid angiopathy (CAA). Clinical presentation of cSS includes transient focal neurological episodes or “amyloid spells”. In addition, spontaneous and amyloid beta immunotherapy-associated CAA-related inflammation may cause cSS, which is included in the hemorrhagic subgroup of amyloid-related imaging abnormalities (ARIA). Because a definitive diagnosis requires a brain biopsy, knowledge of neuroimaging features and clinical findings in CAA-related inflammation is essential. This review provides neuroradiological hallmarks of the two groups of SS and give an overview of neurological symptoms and differential diagnostic considerations.
Collapse
|
32
|
Sawyer RP, Demel SL, Comeau ME, Marion M, Rosand J, Langefeld CD, Woo D. Alzheimer's disease related single nucleotide polymorphisms and correlation with intracerebral hemorrhage incidence. Medicine (Baltimore) 2022; 101:e30782. [PMID: 36181103 PMCID: PMC9524946 DOI: 10.1097/md.0000000000030782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Apolipoprotein E alleles have been associated with both Alzheimer's disease (AD) and intracerebral hemorrhage (ICH). In addition, ICH is associated with a markedly high risk of subsequent dementia compared to other subtypes of stroke. We sought to evaluate if other genetic markers for AD were also associated with ICH. We examined whether published AD risk single nucleotide polymorphisms (SNPs) and haplotypes were associated with ICH utilizing genome-wide association study data from 2 independent studies (genetic and environmental risk factors for hemorrhagic stroke [GERFHS] study and genetics of cerebral hemorrhage with anticoagulation [GOCHA]). Analyses included evaluation by location of ICH. GERFHS and GOCHA cohorts contained 745 ICH cases and 536 controls for analysis. The strongest association was on 1q32 near Complement receptor type 1 (CR1), where rs6701713 was associated with all ICH (P = .0074, odds ratio [OR] = 2.07) and lobar ICH (P = .0073, OR = 2.80). The 51 most significant 2-SNP haplotypes associated with lobar ICH were identified within the Clusterin (CLU) gene. We identified that variation within CR1 and CLU, previously identified risk factors for AD, and are associated with an increased risk for ICH driven primarily by lobar ICH. Previous work implicated CR1 and CLU in cerebral amyloid clearance, the innate immune system, and cellular stress response.
Collapse
Affiliation(s)
- Russell P. Sawyer
- University of Cincinnati College of Medicine, Department of Neurology and Rehabilitation Medicine, Cincinnati, OH, USA
- *Correspondence: Russell P. Sawyer, Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, 260 Stetson Street ML 0525, Cincinnati, OH 45267-0525, USA (e-mail: )
| | - Stacie L. Demel
- University of Cincinnati College of Medicine, Department of Neurology and Rehabilitation Medicine, Cincinnati, OH, USA
| | - Mary E. Comeau
- Department of Biostatistics and Data Science, Wake Forest University, Winston-Salem, NC, USA
| | - Miranda Marion
- Department of Biostatistics and Data Science, Wake Forest University, Winston-Salem, NC, USA
| | - Jonathan Rosand
- Henry and Allison McCance Center for Brain Health, Division of Neurocritical Care and Emergency Neurology, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Wake Forest University, Winston-Salem, NC, USA
| | - Daniel Woo
- University of Cincinnati College of Medicine, Department of Neurology and Rehabilitation Medicine, Cincinnati, OH, USA
| |
Collapse
|
33
|
Quintin S, Sorrentino ZA, Mehkri Y, Sriram S, Weisman S, Davidson CG, Lloyd GM, Sung E, Figg JW, Lucke-Wold B. Proteinopathies and Neurotrauma: Update on Degenerative Cascades. JSM NEUROSURGERY AND SPINE 2022; 9:1106. [PMID: 36466377 PMCID: PMC9717712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neurotrauma, especially repetitive neurotrauma, is associated with the development of progressive neurodegeneration leading to chronic traumatic encephalopathy (CTE). Exposure to neurotrauma regularly occurs during sports and military service, often not requiring medical care. However, exposure to severe and/or repeated sub-clinical neurotrauma has been shown cause physical and psychological disability, leading to reduce life expectancy. Misfolding of proteins, or proteinopathy, is a pathological hallmark of CTE, in which chronic injury leads to local and diffuse protein aggregates. These aggregates are an overlapping feature of many neurodegenerative diseases such as CTE, Alzheimer's Disease, Parkinsons disease. Neurotrauma is also a significant risk factor for the development of these diseases, however the mechanism's underlying this association are not well understood. While phosphorylated tau aggregates are the primary feature of CTE, amyloid-beta, Transactive response DNA-binding protein 43 (TDP-43), and alpha-synuclein (αSyn) are also well documented. Aberrant misfolding of these proteins has been shown to disrupt brain homeostasis leading to neurodegeneration in a disease dependent manor. In CTE, the interaction between proteinopathies and their associated neurodegeneration is a current area of study. Here we provide an update on current literature surrounding the prevalence, characteristics, and pathogenesis of proteinopathies in CTE.
Collapse
Affiliation(s)
| | | | | | - Sai Sriram
- College of Medicine, University of Florida, USA
| | | | | | - Grace M Lloyd
- Department of Neuroscience, University of Florida, USA
| | - Eric Sung
- College of Medicine, University of Florida, USA
| | - John W Figg
- Department of Neurosurgery, University of Florida, USA
| | | |
Collapse
|
34
|
Valiukas Z, Ephraim R, Tangalakis K, Davidson M, Apostolopoulos V, Feehan J. Immunotherapies for Alzheimer’s Disease—A Review. Vaccines (Basel) 2022; 10:vaccines10091527. [PMID: 36146605 PMCID: PMC9503401 DOI: 10.3390/vaccines10091527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disorder that falls under the umbrella of dementia and is characterised by the presence of highly neurotoxic amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs) of tau protein within the brain. Historically, treatments for AD have consisted of medications that can slow the progression of symptoms but not halt or reverse them. The shortcomings of conventional drugs have led to a growing need for novel, effective approaches to the treatment of AD. In recent years, immunotherapies have been at the forefront of these efforts. Briefly, immunotherapies utilise the immune system of the patient to treat a condition, with common immunotherapies for AD consisting of the use of monoclonal antibodies or vaccines. Most of these treatments target the production and deposition of Aβ due to its neurotoxicity, but treatments specifically targeting tau protein are being researched as well. These treatments have had great variance in their efficacy and safety, leading to a constant need for the research and development of new safe and effective treatments.
Collapse
Affiliation(s)
- Zachary Valiukas
- College of Health and Biomedicine, Victoria University, Melbourne, VIC 3011, Australia
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
| | - Kathy Tangalakis
- First Year College, Victoria University, Melbourne, VIC 3011, Australia
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC 3011, Australia
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
- Correspondence:
| |
Collapse
|
35
|
Malik R, Kalra S, Bhatia S, Harrasi AA, Singh G, Mohan S, Makeen HA, Albratty M, Meraya A, Bahar B, Tambuwala MM. Overview of therapeutic targets in management of dementia. Biomed Pharmacother 2022; 152:113168. [PMID: 35701303 DOI: 10.1016/j.biopha.2022.113168] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Dementia is defined as a gradual cognitive impairment that interferes with everyday tasks, and is a leading cause of dependency, disability, and mortality. According to the current scenario, millions of individuals worldwide have dementia. This review provides with an overview of dementia before moving on to its subtypes (neurodegenerative and non-neurodegenerative) and pathophysiology. It also discusses the incidence and severity of dementia, focusing on Alzheimer's disease with its different hypotheses such as Aβ cascade hypothesis, Tau hypothesis, inflammatory hypothesis, cholinergic and oxidative stress hypothesis. Alzheimer's disease is the most common type and a progressive neurodegenerative illness distinct by neuronal loss and resulting cognitive impairment, leading to dementia. Alzheimer's disease (AD) is considered the most familiar neurodegenerative dementias that affect mostly older population. There are still no disease-modifying therapies available for any dementias at this time, but there are various methods for lowering the risk to dementia patients by using suitable diagnostic and evaluation methods. Thereafter, the management and treatment of primary risk elements of dementia are reviewed. Finally, the future perspectives of dementia (AD) focusing on the impact of the new treatment are discussed.
Collapse
Affiliation(s)
- Rohit Malik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Sunishtha Kalra
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Saurabh Bhatia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Oman
| | - Ahmed Al Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Oman
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India.
| | - Syam Mohan
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim Meraya
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Bojlul Bahar
- Nutrition Sciences and Applied Food Safety Studies, Research Centre for Global Development, School of Sport & Health Sciences, University of Central Lancashire, Preston, UK
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, UK.
| |
Collapse
|
36
|
Nelson PT, Brayne C, Flanagan ME, Abner EL, Agrawal S, Attems J, Castellani RJ, Corrada MM, Cykowski MD, Di J, Dickson DW, Dugger BN, Ervin JF, Fleming J, Graff-Radford J, Grinberg LT, Hokkanen SRK, Hunter S, Kapasi A, Kawas CH, Keage HAD, Keene CD, Kero M, Knopman DS, Kouri N, Kovacs GG, Labuzan SA, Larson EB, Latimer CS, Leite REP, Matchett BJ, Matthews FE, Merrick R, Montine TJ, Murray ME, Myllykangas L, Nag S, Nelson RS, Neltner JH, Nguyen AT, Petersen RC, Polvikoski T, Reichard RR, Rodriguez RD, Suemoto CK, Wang SHJ, Wharton SB, White L, Schneider JA. Frequency of LATE neuropathologic change across the spectrum of Alzheimer's disease neuropathology: combined data from 13 community-based or population-based autopsy cohorts. Acta Neuropathol 2022; 144:27-44. [PMID: 35697880 PMCID: PMC9552938 DOI: 10.1007/s00401-022-02444-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/04/2022] [Accepted: 05/22/2022] [Indexed: 02/02/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) and Alzheimer's disease neuropathologic change (ADNC) are each associated with substantial cognitive impairment in aging populations. However, the prevalence of LATE-NC across the full range of ADNC remains uncertain. To address this knowledge gap, neuropathologic, genetic, and clinical data were compiled from 13 high-quality community- and population-based longitudinal studies. Participants were recruited from United States (8 cohorts, including one focusing on Japanese-American men), United Kingdom (2 cohorts), Brazil, Austria, and Finland. The total number of participants included was 6196, and the average age of death was 88.1 years. Not all data were available on each individual and there were differences between the cohorts in study designs and the amount of missing data. Among those with known cognitive status before death (n = 5665), 43.0% were cognitively normal, 14.9% had MCI, and 42.4% had dementia-broadly consistent with epidemiologic data in this age group. Approximately 99% of participants (n = 6125) had available CERAD neuritic amyloid plaque score data. In this subsample, 39.4% had autopsy-confirmed LATE-NC of any stage. Among brains with "frequent" neuritic amyloid plaques, 54.9% had comorbid LATE-NC, whereas in brains with no detected neuritic amyloid plaques, 27.0% had LATE-NC. Data on LATE-NC stages were available for 3803 participants, of which 25% had LATE-NC stage > 1 (associated with cognitive impairment). In the subset of individuals with Thal Aβ phase = 0 (lacking detectable Aβ plaques), the brains with LATE-NC had relatively more severe primary age-related tauopathy (PART). A total of 3267 participants had available clinical data relevant to frontotemporal dementia (FTD), and none were given the clinical diagnosis of definite FTD nor the pathological diagnosis of frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP). In the 10 cohorts with detailed neurocognitive assessments proximal to death, cognition tended to be worse with LATE-NC across the full spectrum of ADNC severity. This study provided a credible estimate of the current prevalence of LATE-NC in advanced age. LATE-NC was seen in almost 40% of participants and often, but not always, coexisted with Alzheimer's disease neuropathology.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA.
| | | | | | - Erin L Abner
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | - Jing Di
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | - Lea T Grinberg
- University of California, San Francisco, CA, USA
- University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | | | | | | | | | | | - Mia Kero
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | | | | | | | | | | | | | - Liisa Myllykangas
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sukriti Nag
- Rush University Medical Center, Chicago, IL, USA
| | | | - Janna H Neltner
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | | | | | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Lon White
- Pacific Health Research and Education Institute, Honolulu, HI, USA
| | | |
Collapse
|
37
|
Shared pathophysiology: Understanding stroke and Alzheimer’s disease. Clin Neurol Neurosurg 2022; 218:107306. [PMID: 35636382 DOI: 10.1016/j.clineuro.2022.107306] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/19/2022] [Indexed: 12/17/2022]
|
38
|
Rajpoot J, Crooks EJ, Irizarry BA, Amundson A, Van Nostrand WE, Smith SO. Insights into Cerebral Amyloid Angiopathy Type 1 and Type 2 from Comparisons of the Fibrillar Assembly and Stability of the Aβ40-Iowa and Aβ40-Dutch Peptides. Biochemistry 2022; 61:1181-1198. [PMID: 35666749 PMCID: PMC9219409 DOI: 10.1021/acs.biochem.1c00781] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two distinct diseases are associated with the deposition of fibrillar amyloid-β (Aβ) peptides in the human brain in an age-dependent fashion. Alzheimer's disease is primarily associated with parenchymal plaque deposition of Aβ42, while cerebral amyloid angiopathy (CAA) is associated with amyloid formation of predominantly Aβ40 in the cerebral vasculature. In addition, familial mutations at positions 22 and 23 of the Aβ sequence can enhance vascular deposition in the two major subtypes of CAA. The E22Q (Dutch) mutation is associated with CAA type 2, while the D23N (Iowa) mutation is associated with CAA type 1. Here we investigate differences in the formation and structure of fibrils of these mutant Aβ peptides in vitro to gain insights into their biochemical and physiological differences in the brain. Using Fourier transform infrared and nuclear magnetic resonance spectroscopy, we measure the relative propensities of Aβ40-Dutch and Aβ40-Iowa to form antiparallel structure and compare these propensities to those of the wild-type Aβ40 and Aβ42 isoforms. We find that both Aβ40-Dutch and Aβ40-Iowa have strong propensities to form antiparallel β-hairpins in the first step of the fibrillization process. However, there is a marked difference in the ability of these peptides to form elongated antiparallel structures. Importantly, we find marked differences in the stability of the protofibril or fibril states formed by the four Aβ peptides. We discuss these differences with respect to the mechanisms of Aβ fibril formation in CAA.
Collapse
Affiliation(s)
- Jitika Rajpoot
- Department of Biochemistry and Cell Biology, Center for Structural Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Elliot J Crooks
- Department of Biochemistry and Cell Biology, Center for Structural Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Brandon A Irizarry
- Department of Biochemistry and Cell Biology, Center for Structural Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Ashley Amundson
- Department of Biochemistry and Cell Biology, Center for Structural Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | | | - Steven O Smith
- Department of Biochemistry and Cell Biology, Center for Structural Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| |
Collapse
|
39
|
Huynh TT, Wang Y, Terpstra K, Cho HJ, Mirica LM, Rogers BE. 68Ga-Labeled Benzothiazole Derivatives for Imaging Aβ Plaques in Cerebral Amyloid Angiopathy. ACS OMEGA 2022; 7:20339-20346. [PMID: 35721913 PMCID: PMC9202065 DOI: 10.1021/acsomega.2c02369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/20/2022] [Indexed: 05/17/2023]
Abstract
Timely diagnostic imaging plays a crucial role in managing cerebral amyloid angiopathy (CAA)-the condition in which amyloid β is deposited on blood vessels. To selectively map these amyloid plaques, we have designed amyloid-targeting ligands that can effectively complex with 68Ga3+ while maintaining good affinity for amyloid β. In this study, we introduced novel 1,4,7-triazacyclononane-based bifunctional chelators (BFCs) that incorporate a benzothiazole moiety as the Aβ-binding fragment and form charged and neutral species with 68Ga3+. In vitro autoradiography using 5xFAD and WT mouse brain sections (11-month-old) suggested strong and specific binding of the 68Ga complexes to amyloid β. Biodistribution studies in CD-1 mice revealed a low brain uptake of 0.10-0.33% ID/g, thus suggesting 68Ga-labeled novel BFCs as promising candidates for detecting CAA.
Collapse
Affiliation(s)
- Truc T. Huynh
- Department
of Radiation Oncology, Washington University
School of Medicine, 4511
Forest Park Avenue, St. Louis, Missouri 63108, United
States
- Department
of Chemistry, Washington University, St. Louis, Missouri 63130, United States
| | - Yujue Wang
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United
States
| | - Karna Terpstra
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United
States
| | - Hong-Jun Cho
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United
States
| | - Liviu M. Mirica
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United
States
- Hope
Center for Neurological Disorders, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Buck E. Rogers
- Department
of Radiation Oncology, Washington University
School of Medicine, 4511
Forest Park Avenue, St. Louis, Missouri 63108, United
States
| |
Collapse
|
40
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
41
|
The amyloid peptide β disrupts intercellular junctions and increases endothelial permeability in a NADPH oxidase 1-dependent manner. Redox Biol 2022; 52:102287. [PMID: 35358850 PMCID: PMC8966210 DOI: 10.1016/j.redox.2022.102287] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/12/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease is the most common form of dementia and is associated with the accumulation of amyloid peptide β in the brain parenchyma. Vascular damage and microvascular thrombosis contribute to the neuronal degeneration and the loss of brain function typical of this disease. In this study, we utilised a murine model of Alzheimer's disease to evaluate the neurovascular effects of this disease. Upon detection of an increase in the phosphorylation of the endothelial surface receptor VE-cadherin, we focused our attention on endothelial cells and utilised two types of human endothelial cells cultured in vitro: 1) human umbilical vein endothelial cells (HUVECs) and 2) human brain microvascular endothelial cells (hBMECs). Using an electrical current impedance system (ECIS) and FITC-albumin permeability assays, we discovered that the treatment of human endothelial cells with amyloid peptide β causes a loss in their barrier function, which is oxidative stress-dependent and similarly to our observation in mouse brain associates with VE-cadherin phosphorylation. The activation of the superoxide anion-generating enzyme NADPH oxidase 1 is responsible for the oxidative stress that leads to the disruption of barrier function in human endothelial cells in vitro. In summary, we have identified a novel molecular mechanism explaining how the accumulation of amyloid peptide β in the brain parenchyma may induce the loss of neurovascular barrier function, which has been observed in patients. Neurovascular leakiness plays an important role in brain inflammation and neuronal degeneration driving the progression of the Alzheimer's disease. Therefore, this study provides a novel and promising target for the development of a pharmacological treatment to protect neurovascular function and reduce the progression of the neurodegeneration in Alzheimer's patients. Amyloid peptide β induces oxidative changes in mouse hippocampus. The endothelial barrier function is impaired by amyloid peptide β. Oxidative stress is critical for the increase in endothelial monolayer permeability. NADPH oxidase 1 mediates the endothelial barrier damage caused by amyloid peptide β.
Collapse
|
42
|
Zhu WM, Neuhaus A, Beard DJ, Sutherland BA, DeLuca GC. Neurovascular coupling mechanisms in health and neurovascular uncoupling in Alzheimer's disease. Brain 2022; 145:2276-2292. [PMID: 35551356 PMCID: PMC9337814 DOI: 10.1093/brain/awac174] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
To match the metabolic demands of the brain, mechanisms have evolved to couple neuronal activity to vasodilation, thus increasing local cerebral blood flow and delivery of oxygen and glucose to active neurons. Rather than relying on metabolic feedback signals such as the consumption of oxygen or glucose, the main signalling pathways rely on the release of vasoactive molecules by neurons and astrocytes, which act on contractile cells. Vascular smooth muscle cells and pericytes are the contractile cells associated with arterioles and capillaries, respectively, which relax and induce vasodilation. Much progress has been made in understanding the complex signalling pathways of neurovascular coupling, but issues such as the contributions of capillary pericytes and astrocyte calcium signal remain contentious. Study of neurovascular coupling mechanisms is especially important as cerebral blood flow dysregulation is a prominent feature of Alzheimer’s disease. In this article we will discuss developments and controversies in the understanding of neurovascular coupling and finish by discussing current knowledge concerning neurovascular uncoupling in Alzheimer’s disease.
Collapse
Affiliation(s)
- Winston M Zhu
- Oxford Medical School, University of Oxford, Oxford, UK
| | - Ain Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
43
|
Beaman C, Kozii K, Hilal S, Liu M, Spagnolo-Allende AJ, Polanco-Serra G, Chen C, Cheng CY, Zambrano D, Arikan B, Del Brutto VJ, Wright C, Flowers XE, Leskinen SP, Rundek T, Mitchell A, Vonsattel JP, Cortes E, Teich AF, Sacco RL, Elkind MSV, Roh D, Gutierrez J. Cerebral Microbleeds, Cerebral Amyloid Angiopathy, and Their Relationships to Quantitative Markers of Neurodegeneration. Neurology 2022; 98:e1605-e1616. [PMID: 35228332 PMCID: PMC9052569 DOI: 10.1212/wnl.0000000000200142] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/18/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Age-related cognitive impairment is driven by the complex interplay of neurovascular and neurodegenerative disease. There is a strong relationship between cerebral microbleeds (CMBs), cerebral amyloid angiopathy (CAA), and the cognitive decline observed in conditions such as Alzheimer disease. However, in the early, preclinical phase of cognitive impairment, the extent to which CMBs and underlying CAA affect volumetric changes in the brain related to neurodegenerative disease remains unclear. METHODS We performed cross-sectional analyses from 3 large cohorts: The Northern Manhattan Study (NOMAS), Alzheimer's Disease Neuroimaging Initiative (ADNI), and the Epidemiology of Dementia in Singapore study (EDIS). We conducted a confirmatory analysis of 82 autopsied cases from the Brain Arterial Remodeling Study (BARS). We implemented multivariate regression analyses to study the association between 2 related markers of cerebrovascular disease-MRI-based CMBs and autopsy-based CAA-as independent variables and volumetric markers of neurodegeneration as dependent variables. NOMAS included mostly dementia-free participants age 55 years or older from northern Manhattan. ADNI included participants living in the United States age 55-90 years with a range of cognitive status. EDIS included community-based participants living in Singapore age 60 years and older with a range of cognitive status. BARS included postmortem pathologic samples. RESULTS We included 2,657 participants with available MRI data and 82 autopsy cases from BARS. In a meta-analysis of NOMAS, ADNI, and EDIS, superficial CMBs were associated with larger gray matter (β = 4.49 ± 1.13, p = 0.04) and white matter (β = 4.72 ± 2.1, p = 0.03) volumes. The association between superficial CMBs and larger white matter volume was more evident in participants with 1 CMB (β = 5.17 ± 2.47, p = 0.04) than in those with ≥2 CMBs (β = 1.97 ± 3.41, p = 0.56). In BARS, CAA was associated with increased cortical thickness (β = 6.5 ± 2.3, p = 0.016) but not with increased brain weight (β = 1.54 ± 1.29, p = 0.26). DISCUSSION Superficial CMBs are associated with larger morphometric brain measures, specifically white matter volume. This association is strongest in brains with fewer CMBs, suggesting that the CMB/CAA contribution to neurodegeneration may not relate to tissue loss, at least in early stages of disease.
Collapse
Affiliation(s)
- Charles Beaman
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Krystyna Kozii
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Saima Hilal
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Minghua Liu
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Anthony J Spagnolo-Allende
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Guillermo Polanco-Serra
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Christopher Chen
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Ching-Yu Cheng
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Daniela Zambrano
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Burak Arikan
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Victor J Del Brutto
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Clinton Wright
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Xena E Flowers
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Sandra P Leskinen
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Tatjana Rundek
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Amanda Mitchell
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jean Paul Vonsattel
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Etty Cortes
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Andrew F Teich
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Ralph L Sacco
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Mitchell S V Elkind
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - David Roh
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jose Gutierrez
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| |
Collapse
|
44
|
Grangeon L, Paquet C, Guey S, Zarea A, Martinaud O, Rotharmel M, Maltête D, Quillard-Muraine M, Nicolas G, Charbonnier C, Chabriat H, Wallon D. Cerebrospinal Fluid Profile of Tau, Phosphorylated Tau, Aβ42, and Aβ40 in Probable Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 87:791-802. [DOI: 10.3233/jad-215208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: There is no consensus regarding the diagnostic value of cerebrospinal fluid (CSF) Alzheimer’s disease (AD) biomarkers in cerebral amyloid angiopathy (CAA). Objective: To describe the CSF levels of Aβ 42, Aβ 40, total protein Tau, and phosphorylated-Tau (p-Tau) in a large series of probable CAA patients and to compare with AD patients in order to identify a specific pattern in CAA but also to look for correlations with the neuroimaging profile. Methods: We retrospectively included from 2 French centers probable CAA patients according to modified Boston criteria who underwent lumbar puncture (LP) with CSF AD biomarker quantifications. Two neurologists independently analyzed all MRI sequences. A logistic regression and Spearman’s correlation coefficient were used to identify correlation between MRI and CSF biomarkers in CAA. Results: We included 63 probable CAA and 27 AD patients. Among CAA 50.8% presented with decreased Aβ 42 level associated with elevated p-Tau and/or Tau, 34.9% with isolated decreased Aβ 42 level and 14.3% patients with normal Aβ 42 level. Compared to AD, CAA showed lower levels of Tau (p = 0.008), p-Tau (p = 0.004), and Aβ 40 (p = 0.001) but similar Aβ 42 level (p = 0.07). No correlation between Aβ 42 or Aβ 40 levels and neuroimaging was found. Conclusion: CSF biomarkers may improve the accuracy of the modified Boston criteria with altered profile in 85% of the patients fulfilling revised Boston criteria for probable CAA. Aβ 40 appears as an interesting selective biomarker in differential diagnosis.
Collapse
Affiliation(s)
- Lou Grangeon
- Normandie Univ, UNIROUEN, Inserm U1245 and CHURouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Claire Paquet
- CMRR Paris Nord AP-HP, Groupe Hospitalier Lariboisière Fernand-Widal Saint-Louis, INSERM, U942, Université Paris Diderot, Sorbonne Paris Cité, UMRS 942, France
| | - Stéphanie Guey
- Department of Neurology, AP-HP, Groupe Hospitalier Lariboisière Fernand-Widal Saint-Louis, Paris, France
| | - Aline Zarea
- Normandie Univ, UNIROUEN, Inserm U1245 and CHURouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | | | - Maud Rotharmel
- Rouvray Hospital of Rouen, University Department of Psychiatry, France
| | - David Maltête
- Normandie Univ, UNIROUEN, Inserm U1245 and CHURouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | | | - Gael Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Camille Charbonnier
- Normandie Univ, UNIROUEN, Inserm U1245 and CHU Rouen, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Hugues Chabriat
- Department of Neurology, AP-HP, Groupe Hospitalier Lariboisière Fernand-Widal Saint-Louis, Paris, France
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and CHURouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| |
Collapse
|
45
|
Sasaguri H, Hashimoto S, Watamura N, Sato K, Takamura R, Nagata K, Tsubuki S, Ohshima T, Yoshiki A, Sato K, Kumita W, Sasaki E, Kitazume S, Nilsson P, Winblad B, Saito T, Iwata N, Saido TC. Recent Advances in the Modeling of Alzheimer's Disease. Front Neurosci 2022; 16:807473. [PMID: 35431779 PMCID: PMC9009508 DOI: 10.3389/fnins.2022.807473] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
Since 1995, more than 100 transgenic (Tg) mouse models of Alzheimer's disease (AD) have been generated in which mutant amyloid precursor protein (APP) or APP/presenilin 1 (PS1) cDNA is overexpressed ( 1st generation models ). Although many of these models successfully recapitulate major pathological hallmarks of the disease such as amyloid β peptide (Aβ) deposition and neuroinflammation, they have suffered from artificial phenotypes in the form of overproduced or mislocalized APP/PS1 and their functional fragments, as well as calpastatin deficiency-induced early lethality, calpain activation, neuronal cell death without tau pathology, endoplasmic reticulum stresses, and inflammasome involvement. Such artifacts bring two important uncertainties into play, these being (1) why the artifacts arise, and (2) how they affect the interpretation of experimental results. In addition, destruction of endogenous gene loci in some Tg lines by transgenes has been reported. To overcome these concerns, single App knock-in mouse models harboring the Swedish and Beyreuther/Iberian mutations with or without the Arctic mutation (AppNL-G-F and AppNL-F mice) were developed ( 2nd generation models ). While these models are interesting given that they exhibit Aβ pathology, neuroinflammation, and cognitive impairment in an age-dependent manner, the model with the Artic mutation, which exhibits an extensive pathology as early as 6 months of age, is not suitable for investigating Aβ metabolism and clearance because the Aβ in this model is resistant to proteolytic degradation and is therefore prone to aggregation. Moreover, it cannot be used for preclinical immunotherapy studies owing to the discrete affinity it shows for anti-Aβ antibodies. The weakness of the latter model (without the Arctic mutation) is that the pathology may require up to 18 months before it becomes sufficiently apparent for experimental investigation. Nevertheless, this model was successfully applied to modulating Aβ pathology by genome editing, to revealing the differential roles of neprilysin and insulin-degrading enzyme in Aβ metabolism, and to identifying somatostatin receptor subtypes involved in Aβ degradation by neprilysin. In addition to discussing these issues, we also provide here a technical guide for the application of App knock-in mice to AD research. Subsequently, a new double knock-in line carrying the AppNL-F and Psen1 P117L/WT mutations was generated, the pathogenic effect of which was found to be synergistic. A characteristic of this 3rd generation model is that it exhibits more cored plaque pathology and neuroinflammation than the AppNL-G-F line, and thus is more suitable for preclinical studies of disease-modifying medications targeting Aβ. Furthermore, a derivative AppG-F line devoid of Swedish mutations which can be utilized for preclinical studies of β-secretase modifier(s) was recently created. In addition, we introduce a new model of cerebral amyloid angiopathy that may be useful for analyzing amyloid-related imaging abnormalities that can be caused by anti-Aβ immunotherapy. Use of the App knock-in mice also led to identification of the α-endosulfine-K ATP channel pathway as components of the somatostatin-evoked physiological mechanisms that reduce Aβ deposition via the activation of neprilysin. Such advances have provided new insights for the prevention and treatment of preclinical AD. Because tau pathology plays an essential role in AD pathogenesis, knock-in mice with human tau wherein the entire murine Mapt gene has been humanized were generated. Using these mice, the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) was discovered as a mediator linking tau pathology to neurodegeneration and showed that tau humanization promoted pathological tau propagation. Finally, we describe and discuss the current status of mutant human tau knock-in mice and a non-human primate model of AD that we have successfully created.
Collapse
Affiliation(s)
- Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Kaori Sato
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Risa Takamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Kenichi Nagata
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tsubuki
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Toshio Ohshima
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Kenya Sato
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Wakako Kumita
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan
| | - Shinobu Kitazume
- Department of Clinical Laboratory Sciences, School of Health Sciences, Fukushima Medical University, Fukushima, Japan
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Nobuhisa Iwata
- Department of Genome-Based Drug Discovery and Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
46
|
DiFrancesco JC, Stanzani L. Increased Brain Volume: A Novel Biomarker of Neurodegeneration? Neurology 2022; 98:649-650. [PMID: 35228330 DOI: 10.1212/wnl.0000000000200166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Jacopo C DiFrancesco
- Department of Neurology, ASST San Gerardo Hosp., Univ. Milano-Bicocca, Monza, Italy
| | - Lorenzo Stanzani
- Department of Neurology, ASST San Gerardo Hosp., Univ. Milano-Bicocca, Monza, Italy
| |
Collapse
|
47
|
Cipriano L, Saracino D, Oliva M, Campana V, Puoti G, Conforti R, Fulgione L, Signoriello E, Bonavita S, Coppola C. Systematic Review on the Role of Lobar Cerebral Microbleeds in Cognition. J Alzheimers Dis 2022; 86:1025-1035. [PMID: 35180115 DOI: 10.3233/jad-215323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cerebral microbleeds (CMBs) are small round/oval lesions seen in MRI-specific sequences. They are divided in deep and lobar according to their location. Lobar CMBs (L-CMBs) are commonly associated with amyloid angiopathy. Although CMBs have been considered clinically silent for a long time, a growing body of evidence has shown that they could play a crucial role in cognitive functioning. OBJECTIVE The aim of this systematic review was to estimate the role of L-CMBs in cognitive performance. METHODS We selected, from the Cochrane Library, Embase, PubMed, and ScienceDirect databases, clinical studies, published from January 2000 to January 2020 and focused on the association between L-CMBs and cognitive functions. The inclusion criteria were: 1) participants grouped according to presence or absence of CMBs, 2) extensive neuropsychological examination, 3) CMBs differentiation according to topographical distribution, and 4) MRI-based CMB definition (< 10 mm and low signal in T2*/SWI). The impact of L-CMBs was separately assessed for executive functions, visuospatial skills, language, and memory. RESULTS Among 963 potentially eligible studies, six fulfilled the inclusion criteria. Four studies reported a greater reduction in executive performances in participants with L-CMB and two studies showed a statistically significant association between visuospatial dysfunction and L-CMBs. No association was found between hippocampal memory or language abilities and L-CMBs. CONCLUSION Lobar CMBs are associated with a reduction of processing speed and visuospatial performances, thus suggesting the contribution of vascular amyloid deposition to this cognitive profile. This occurrence enables us to suspect an underlying Alzheimer's disease pathology even in absence of typical hippocampal memory impairment.
Collapse
Affiliation(s)
- Lorenzo Cipriano
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau (ICM), INSERM U1127, CNRS UMR 7225 - Aramis Project Team, Inria Research Center of Paris - Reference Center for Rare or Early Dementias, IM2A, Department of Neurology, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Mariano Oliva
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vito Campana
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Gianfranco Puoti
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Renata Conforti
- Department of Medicine of Precision, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ludovica Fulgione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Elisabetta Signoriello
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Cinzia Coppola
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
48
|
Pons V, Rivest S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol Rev 2022; 74:1-17. [PMID: 34987086 DOI: 10.1124/pharmrev.121.000400] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is the first progressive neurodegenerative disease worldwide, and the disease is characterized by an accumulation of amyloid in the brain and neurovasculature that triggers cognitive decline and neuroinflammation. The innate immune system has a preponderant role in AD. The last decade, scientists focused their efforts on therapies aiming to modulate innate immunity. The latter is of great interest, since they participate to the inflammation and phagocytose the amyloid in the brain and blood vessels. We and others have developed pharmacological approaches to stimulate these cells using various ligands. These include toll-like receptor 4, macrophage colony stimulating factor, and more recently nucleotide-binding oligomerization domain-containing 2 receptors. This review will discuss the great potential to take advantage of the innate immune system to fight naturally against amyloid β accumulation and prevent its detrimental consequence on brain functions and its vascular system. SIGNIFICANCE STATEMENT: The focus on amyloid β removal from the perivascular space rather than targeting CNS plaque formation and clearance represents a new direction with a great potential. Small molecules able to act at the level of peripheral immunity would constitute a novel approach for tackling aberrant central nervous system biology, one of which we believe would have the potential of generating a lot of interest.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| |
Collapse
|
49
|
Zhou AL, Sharda N, Sarma VV, Ahlschwede KM, Curran GL, Tang X, Poduslo JF, Kalari KR, Lowe VJ, Kandimalla KK. Age-Dependent Changes in the Plasma and Brain Pharmacokinetics of Amyloid-β Peptides and Insulin. J Alzheimers Dis 2022; 85:1031-1044. [PMID: 34924382 PMCID: PMC10846947 DOI: 10.3233/jad-215128] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Age is the most common risk factor for Alzheimer's disease (AD), a neurodegenerative disorder characterized by the hallmarks of toxic amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. Moreover, sub-physiological brain insulin levels have emerged as a pathological manifestation of AD. OBJECTIVE Identify age-related changes in the plasma disposition and blood-brain barrier (BBB) trafficking of Aβ peptides and insulin in mice. METHODS Upon systemic injection of 125I-Aβ40, 125I-Aβ42, or 125I-insulin, the plasma pharmacokinetics and brain influx were assessed in wild-type (WT) or AD transgenic (APP/PS1) mice at various ages. Additionally, publicly available single-cell RNA-Seq data [GSE129788] was employed to investigate pathways regulating BBB transport in WT mice at different ages. RESULTS The brain influx of 125I-Aβ40, estimated as the permeability-surface area product, decreased with age, accompanied by an increase in plasma AUC. In contrast, the brain influx of 125I-Aβ42 increased with age, accompanied by a decrease in plasma AUC. The age-dependent changes observed in WT mice were accelerated in APP/PS1 mice. As seen with 125I-Aβ40, the brain influx of 125I-insulin decreased with age in WT mice, accompanied by an increase in plasma AUC. This finding was further supported by dynamic single-photon emission computed tomography (SPECT/CT) imaging studies. RAGE and PI3K/AKT signaling pathways at the BBB, which are implicated in Aβ and insulin transcytosis, respectively, were upregulated with age in WT mice, indicating BBB insulin resistance. CONCLUSION Aging differentially affects the plasma pharmacokinetics and brain influx of Aβ isoforms and insulin in a manner that could potentially augment AD risk.
Collapse
Affiliation(s)
- Andrew L. Zhou
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Nidhi Sharda
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Vidur V. Sarma
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Kristen M. Ahlschwede
- Department of Pharmaceutical Sciences, Rosalind Franklin University of Medicine and Science, College of Pharmacy, North Chicago, IL, USA
| | - Geoffry L. Curran
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Xiaojia Tang
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Joseph F. Poduslo
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Krishna R. Kalari
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Karunya K. Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| |
Collapse
|
50
|
Apátiga-Pérez R, Soto-Rojas LO, Campa-Córdoba BB, Luna-Viramontes NI, Cuevas E, Villanueva-Fierro I, Ontiveros-Torres MA, Bravo-Muñoz M, Flores-Rodríguez P, Garcés-Ramirez L, de la Cruz F, Montiel-Sosa JF, Pacheco-Herrero M, Luna-Muñoz J. Neurovascular dysfunction and vascular amyloid accumulation as early events in Alzheimer's disease. Metab Brain Dis 2022; 37:39-50. [PMID: 34406560 DOI: 10.1007/s11011-021-00814-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/23/2021] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is clinically characterized by a progressive loss of cognitive functions and short-term memory. AD patients present two distinctive neuropathological lesions: neuritic plaques and neurofibrillary tangles (NFTs), constituted of beta-amyloid peptide (Aβ) and phosphorylated and truncated tau proteins. Aβ deposits around cerebral blood vessels (cerebral amyloid angiopathy, CAA) is a major contributor to vascular dysfunction in AD. Vascular amyloid deposits could be early events in AD due to dysfunction in the neurovascular unit (NVU) and the blood-brain barrier (BBB), deterioration of the gliovascular unit, and/or decrease of cerebral blood flow (CBF). These pathological events can lead to decreased Aβ clearance, facilitate a neuroinflammatory environment as well as synaptic dysfunction and, finally, lead to neurodegeneration. Here, we review the histopathological AD hallmarks and discuss the two-hit vascular hypothesis of AD, emphasizing the role of neurovascular dysfunction as an early factor that favors vascular Aβ aggregation and neurodegeneration. Addtionally, we emphasize that pericyte degeneration is a key and early element in AD that can trigger amyloid vascular accumulation and NVU/BBB dysfunction. Further research is required to better understand the early pathophysiological mechanisms associated with NVU alteration and CAA to generate early biomarkers and timely treatments for AD.
Collapse
Affiliation(s)
- Ricardo Apátiga-Pérez
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Luis O Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Estado de México, Mexico
| | - B Berenice Campa-Córdoba
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Nabil Itzi Luna-Viramontes
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Elvis Cuevas
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, USA
| | | | | | | | | | - Linda Garcés-Ramirez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Fidel de la Cruz
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - José Francisco Montiel-Sosa
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
| | - Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic.
| | - José Luna-Muñoz
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México.
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo, República Dominicana.
| |
Collapse
|