1
|
Kalaria R, Englund E. Neuropathological features of cerebrovascular diseases. Pathology 2025; 57:207-219. [PMID: 39718486 DOI: 10.1016/j.pathol.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 12/25/2024]
Abstract
Optimal blood flow through a patent cerebral circulation is critical for supply of oxygen and nutrients for brain function. The integrity of vascular elements within arterial vessels of any calibre can be compromised by various disease processes. Pathological changes in the walls of veins and the venous system may also alter the dynamics of cerebral perfusion. The consequences of both systemic vascular and cerebrovascular diseases range from acute focal changes to irreversible chronic restructuring of the brain parenchyma. Cerebral infarcts of different sizes may instigate a cascade of programmed cell death mechanisms including autophagy and mitophagy and processes that range from necroptosis to ferroptosis. Recent advances also emphasise the role of the vascular inflammasome in the pathology of cerebral infarction. Here, we summarise current knowledge on frequencies, epidemiological features and the neuropathology of common cerebrovascular disorders among which cerebral small vessel diseases have become of particular interest. We also highlight the current spectrum of monogenic and polygenic genetic disorders affecting the intracranial vasculature. With the advent of DNA screening technologies, it is now realised that several cerebrovascular disorders exhibit strong genetic traits. Whilst several gene defects and their aberrant products are identified, the precise role or mechanisms of how they influence angiogenesis, vasculogenesis, vessel integrity or the extracellular matrix remain largely unclear. Despite such genetic advances, histopathological examination remains the gold standard for diagnosis and characterisation of most cerebrovascular disorders.
Collapse
Affiliation(s)
- Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| | - Elisabet Englund
- Institutionen för kliniska vetenskaper, Lunds Universitet, Klinisk Patologi & Medicinsk Service, Region Skåne, Lund, Sweden
| |
Collapse
|
2
|
Grangeon L, Charbonnier C, Rousseau S, Richard AC, Quenez O, Zarea A, Boland A, Olaso R, Deleuze J, Tournier‐Lasserve E, Nicolas G, Wallon D. Input of exome sequencing in early-onset cerebral amyloid angiopathy. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70027. [PMID: 39583652 PMCID: PMC11585166 DOI: 10.1002/dad2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Genetics of cerebral amyloid angiopathy (CAA) remains understudied. METHODS We assessed variants in Alzheimer's disease (AD) risk factor genes and differential diagnosis genes by performing exome sequencing among 78 patients with early-onset definite or probable CAA, after negative screening for APP mutation or duplication. RESULTS Among 14 genes involved in non-Aβ CAA, or vascular leukoencephalopathies, we detected pathogenic NOTCH3 variants in two patients, who exhibited lobar hematomas at the ages of 58 and 65, leading to a diagnosis redirection toward CADASIL. Of the remaining 76 patients, 23.1% carried at least one apolipoprotein E (APOE) ε2 allele and 43.6% carried at least one APOE ε4 allele, known as CAA risk factors. A total of 15 out of 76 (19.7%) carried either a loss-of-function or a rare predicted damaging missense or known AD risk variant in SORL1, TREM2, ABCA7, ABCA1, and ATP8B4. DISCUSSION Exome sequencing allowed the redirection toward CADASIL in two patients and suggested shared genetic factors between AD and CAA, beyond the APOE gene. Highlights The genetic component of cerebral amyloid angiopathy (CAA) remains understudied.Rare differential diagnoses such as CADASIL should be considered, even in cases of cerebral hemorrhage.Our study suggests shared genetic factors between AD and CAA, beyond the APOE gene.Rare variants in SORL1, TREM2, ABCA7, ABCA1 and ATP8B4 might be susceptibility factors in early-onset CAA.
Collapse
Affiliation(s)
- Lou Grangeon
- Department of Neurology and CNRMAJUniv Rouen NormandieRouenFrance
| | - Camille Charbonnier
- Department of GeneticsDepartment of Biostatistics and CNRMAJUniv Rouen NormandieRouenFrance
| | | | | | - Olivier Quenez
- Department of Genetics and CNRMAJUniv Rouen NormandieRouenFrance
| | - Aline Zarea
- Department of Neurology and CNRMAJUniv Rouen NormandieRouenFrance
| | - Anne Boland
- Université Paris‐SaclayCEACentre National de Recherche en Génomique Humaine (CNRGH)EvryFrance
| | - Robert Olaso
- Université Paris‐SaclayCEACentre National de Recherche en Génomique Humaine (CNRGH)EvryFrance
| | - Jean‐François Deleuze
- Université Paris‐SaclayCEACentre National de Recherche en Génomique Humaine (CNRGH)EvryFrance
| | | | | | - Gael Nicolas
- Department of Genetics and CNRMAJUniv Rouen NormandieRouenFrance
| | - David Wallon
- Department of Neurology and CNRMAJUniv Rouen NormandieRouenFrance
| |
Collapse
|
3
|
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
|
4
|
Zhang Z, Lim MJR. Incident Dementia After Spontaneous Intracerebral Hemorrhage. J Alzheimers Dis 2024; 99:41-51. [PMID: 38640161 DOI: 10.3233/jad-240111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Post-stroke cognitive impairment and dementia (PSCID) is a complication that affects long-term functional outcomes after stroke. Studies on dementia after long-term follow-up in stroke have focused predominantly on ischemic stroke, which may be different from the development of dementia after spontaneous intracerebral hemorrhage (ICH). In this review, we summarize the existing data and hypotheses on the development of dementia after spontaneous ICH, review the management of post-ICH dementia, and suggest areas for future research. Dementia after spontaneous ICH has a cumulative incidence of up to 32.0-37.4% at 5 years post-ICH. Although the pathophysiology of post-ICH dementia has not been fully understood, two main theoretical frameworks can be considered: 1) the triggering role of ICH (both primary and secondary brain injury) in precipitating cognitive decline and dementia; and 2) the contributory role of pre-existing brain pathology (including small vessel disease and neurodegenerative pathology), reduced cognitive reserve, and genetic factors predisposing to cognitive dysfunction. These pathophysiological pathways may have synergistic effects that converge on dysfunction of the neurovascular unit and disruptions in functional connectivity leading to dementia post-ICH. Management of post-ICH dementia may include screening and monitoring, cognitive therapy, and pharmacotherapy. Non-invasive brain stimulation is an emerging therapeutic modality under investigation for safety and efficacy. Our review highlights that there remains a paucity of data and standardized reporting on incident dementia after spontaneous ICH. Further research is imperative for determining the incidence, risk factors, and pathophysiology of post-ICH dementia, in order to identify new therapies for the treatment of this debilitating condition.
Collapse
Affiliation(s)
- Zheting Zhang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | |
Collapse
|
5
|
Wang L, Liu Q, Yue D, Liu J, Fu Y. Cerebral Amyloid Angiopathy: An Undeniable Small Vessel Disease. J Stroke 2024; 26:1-12. [PMID: 38326703 PMCID: PMC10850457 DOI: 10.5853/jos.2023.01942] [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: 06/17/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 02/09/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) has been proven to be the most common pathological change in cerebral small vessel disease except arteriosclerosis. In recent years, with the discovery of imaging technology and new imaging markers, the diagnostic rate of CAA has greatly improved. CAA plays an important role in non-hypertensive cerebral hemorrhage and cognitive decline. This review comprehensively describes the etiology, epidemiology, pathophysiological mechanisms, clinical features, imaging manifestations, imaging markers, diagnostic criteria, and treatment of CAA to facilitate its diagnosis and treatment and reduce mortality.
Collapse
Affiliation(s)
- Litao Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiong Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqi Yue
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Fu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Nahon DM, Ganesh S, van den Hil FE, Freund C, Mummery CL, Orlova VV. Genetic repair of a human induced pluripotent cell line from patient with Dutch-type cerebral amyloid angiopathy. Stem Cell Res 2023; 71:103180. [PMID: 37603921 DOI: 10.1016/j.scr.2023.103180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023] Open
Abstract
Dutch-type cerebral amyloid angiopathy (D-CAA), also known as hereditary cerebral haemorrhage with amyloidosis-Dutch type (HCHWA-D), is an autosomal dominant disorder caused by a G to C transversion in codon 693 of the amyloid precursor protein (APP) that results in a Gln-to-Glu amino acid substitution. CRISPR-Cas9 editing was used for genetic correction of the mutation in a human induced pluripotent stem cell (hiPSC-) line established previously. The isogenic hiPSCs generated showed typical pluripotent stem cell morphology, expressed all markers of undifferentiated state, displayed a normal karyotype and had the capacity to differentiate into the three germ layers.
Collapse
Affiliation(s)
- Dennis M Nahon
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Sravya Ganesh
- Leiden University Medical Center hiPSC Hotel, Leiden 2333ZA, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Christian Freund
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, The Netherlands; Leiden University Medical Center hiPSC Hotel, Leiden 2333ZA, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, The Netherlands.
| |
Collapse
|
7
|
Grangeon L, Charbonnier C, Zarea A, Rousseau S, Rovelet-Lecrux A, Bendetowicz D, Lemaitre M, Malrain C, Quillard-Muraine M, Cassinari K, Maltete D, Pariente J, Moreaud O, Magnin E, Cretin B, Mackowiak MA, Sillaire AR, Vercelletto M, Dionet E, Felician O, Rod-Olivieri P, Thomas-Antérion C, Godeneche G, Sauvée M, Cartz-Piver L, Le Ber I, Chauvire V, Jonveaux T, Balageas AC, Laquerriere A, Duyckaerts C, Vital A, de Paula AM, Meyronet D, Guyant-Marechal L, Hannequin D, Tournier-Lasserve E, Campion D, Nicolas G, Wallon D. Phenotype and imaging features associated with APP duplications. Alzheimers Res Ther 2023; 15:93. [PMID: 37170141 PMCID: PMC10173644 DOI: 10.1186/s13195-023-01172-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/18/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND APP duplication is a rare genetic cause of Alzheimer disease and cerebral amyloid angiopathy (CAA). We aimed to evaluate the phenotypes of APP duplications carriers. METHODS Clinical, radiological, and neuropathological features of 43 APP duplication carriers from 24 French families were retrospectively analyzed, and MRI features and cerebrospinal fluid (CSF) biomarkers were compared to 40 APP-negative CAA controls. RESULTS Major neurocognitive disorders were found in 90.2% symptomatic APP duplication carriers, with prominent behavioral impairment in 9.7%. Symptomatic intracerebral hemorrhages were reported in 29.2% and seizures in 51.2%. CSF Aβ42 levels were abnormal in 18/19 patients and 14/19 patients fulfilled MRI radiological criteria for CAA, while only 5 displayed no hemorrhagic features. We found no correlation between CAA radiological signs and duplication size. Compared to CAA controls, APP duplication carriers showed less disseminated cortical superficial siderosis (0% vs 37.5%, p = 0.004 adjusted for the delay between symptoms onset and MRI). Deep microbleeds were found in two APP duplication carriers. In addition to neurofibrillary tangles and senile plaques, CAA was diffuse and severe with thickening of leptomeningeal vessels in all 9 autopsies. Lewy bodies were found in substantia nigra, locus coeruleus, and cortical structures of 2/9 patients, and one presented vascular amyloid deposits in basal ganglia. DISCUSSION Phenotypes associated with APP duplications were heterogeneous with different clinical presentations including dementia, hemorrhage, and seizure and different radiological presentations, even within families. No apparent correlation with duplication size was found. Amyloid burden was severe and widely extended to cerebral vessels as suggested by hemorrhagic features on MRI and neuropathological data, making APP duplication an interesting model of CAA.
Collapse
Affiliation(s)
- Lou Grangeon
- Department of Neurology and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France.
- Department of Neurology, Rouen University Hospital, Rouen Cedex, 76031, France.
| | - Camille Charbonnier
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Aline Zarea
- Department of Neurology and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Stephane Rousseau
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Anne Rovelet-Lecrux
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - David Bendetowicz
- Neurology Department, Sorbonne Université, Paris Brain Institute - ICM, Inserm, CNRS and APHP, Hôpital de la Pitié-Salpétrière APHP, Paris, France
| | - Marion Lemaitre
- Geriatric department, Seclin-Carvin Hospital, Seclin, France
| | - Cécile Malrain
- Department of Neurology, Rennes Hospital, Rennes, France
| | | | - Kevin Cassinari
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - David Maltete
- Department of Neurology and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Jeremie Pariente
- Neurology Department, Toulouse University Hospital and Toulouse NeuroImaging Center (ToNIC) INSERM-Univeristy of Toulouse Paul Sabatier, Toulouse, France
| | - Olivier Moreaud
- Department of Neurology, Grenoble Hospital, Grenoble, France
| | - Eloi Magnin
- Department of Neurology, Besancon Hospital, Besancon, France
| | - Benjamin Cretin
- Department of Neurology, Hautepierre Hospital, Strasbourg, France
| | | | | | | | - Elsa Dionet
- Department of Neurology, Clermont-Ferrand Hospital, Clermont-Ferrand, France
| | - Olivier Felician
- APHM, Service de Neurologie et Neuropsychologie, CHU Timone, Marseille, France
- Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | | | | | - Gaelle Godeneche
- Department of Neurology, La Rochelle Hospital, La Rochelle, France
| | - Mathilde Sauvée
- Department of Neurology, Grenoble Hospital, Grenoble, France
| | - Leslie Cartz-Piver
- Centre Mémoire Ressources et Recherche (CMRR), Limoges University Hospital, Limoges, France
| | - Isabelle Le Ber
- Neurology Department, Sorbonne Université, Paris Brain Institute - ICM, Inserm, CNRS and APHP, Hôpital de la Pitié-Salpétrière APHP, Paris, France
| | - Valérie Chauvire
- Department of Neurology, Angers University Hospital, Angers, France
| | - Therèse Jonveaux
- Department of Neurology, Nancy University Hospital, Nancy, France
| | | | - Annie Laquerriere
- Department of Neuropathology, F 76000, Normandy Center for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Rouen, France
| | - Charles Duyckaerts
- Sorbonne Unviersité, INSERM, CNRS U1127, ICM and Laboratoire de Neuropathologie R. Escourolle, Hospital Pitie-Salpêtrière, Paris, France
| | - Anne Vital
- Department of Pathology, University Hospital, Bordeaux, France
| | | | - David Meyronet
- Department of Pathology, Hopital Civil University Hospital, Lyon, France
| | - Lucie Guyant-Marechal
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Didier Hannequin
- Department of Neurology and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Elisabeth Tournier-Lasserve
- AP-HP, Groupe Hospitalier Saint-Louis Lariboisière-Fernand-Widal, Service de Génétique Moléculaire Neurovasculaire, INSERM UMR 1141, NeuroDiderot, Université de Paris, Paris, France
| | - Dominique Campion
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - Gaël Nicolas
- Department of Genetics and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| | - David Wallon
- Department of Neurology and CNR-MAJ, Univ Rouen Normandie, U1245 and CHU Rouen, 76000, Rouen, France
| |
Collapse
|
8
|
Daoutsali E, Pepers BA, Stamatakis S, van der Graaf LM, Terwindt GM, Parfitt DA, Buijsen RAM, van Roon-Mom WMC. Amyloid beta accumulations and enhanced neuronal differentiation in cerebral organoids of Dutch-type cerebral amyloid angiopathy patients. Front Aging Neurosci 2023; 14:1048584. [PMID: 36733499 PMCID: PMC9887998 DOI: 10.3389/fnagi.2022.1048584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction ADutch-type cerebral amyloid angiopathy (D-CAA) is a hereditary brain disorder caused by a point mutation in the amyloid precursor protein (APP) gene. The mutation is located within the amyloid beta (Aβ) domain of APP and leads to Aβ peptide accumulation in and around the cerebral vasculature. There lack of disease models to study the cellular and molecular pathological mechanisms of D-CAA together with the absence of a disease phenotype in vitro in overexpression cell models, as well as the limited availability of D-CAA animal models indicates the need for a D-CAA patient-derived model. Methods We generated cerebral organoids from four D-CAA patients and four controls, cultured them up to 110 days and performed immunofluorescent and targeted gene expression analyses at two time points (D52 and D110). Results D-CAA cerebral organoids exhibited Aβ accumulations, showed enhanced neuronal and astrocytic gene expression and TGFβ pathway de-regulation. Conclusions These results illustrate the potential of cerebral organoids as in vitro disease model of D-CAA that can be used to understand disease mechanisms of D-CAA and can serve as therapeutic intervention platform for various Aβ-related disorders.
Collapse
Affiliation(s)
- Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Willeke M. C. van Roon-Mom, ; Elena Daoutsali,
| | - Barry A. Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Stavros Stamatakis
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Gisela M. Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - David A. Parfitt
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Ronald A. M. Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Willeke M. C. van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Willeke M. C. van Roon-Mom, ; Elena Daoutsali,
| |
Collapse
|
9
|
Zellner A, Müller SA, Lindner B, Beaufort N, Rozemuller AJM, Arzberger T, Gassen NC, Lichtenthaler SF, Kuster B, Haffner C, Dichgans M. Proteomic profiling in cerebral amyloid angiopathy reveals an overlap with CADASIL highlighting accumulation of HTRA1 and its substrates. Acta Neuropathol Commun 2022; 10:6. [PMID: 35074002 PMCID: PMC8785498 DOI: 10.1186/s40478-021-01303-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an age-related condition and a major cause of intracerebral hemorrhage and cognitive decline that shows close links with Alzheimer's disease (AD). CAA is characterized by the aggregation of amyloid-β (Aβ) peptides and formation of Aβ deposits in the brain vasculature resulting in a disruption of the angioarchitecture. Capillaries are a critical site of Aβ pathology in CAA type 1 and become dysfunctional during disease progression. Here, applying an advanced protocol for the isolation of parenchymal microvessels from post-mortem brain tissue combined with liquid chromatography tandem mass spectrometry (LC-MS/MS), we determined the proteomes of CAA type 1 cases (n = 12) including a patient with hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D), and of AD cases without microvascular amyloid pathology (n = 13) in comparison to neurologically healthy controls (n = 12). ELISA measurements revealed microvascular Aβ1-40 levels to be exclusively enriched in CAA samples (mean: > 3000-fold compared to controls). The proteomic profile of CAA type 1 was characterized by massive enrichment of multiple predominantly secreted proteins and showed significant overlap with the recently reported brain microvascular proteome of patients with cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a hereditary cerebral small vessel disease (SVD) characterized by the aggregation of the Notch3 extracellular domain. We found this overlap to be largely attributable to the accumulation of high-temperature requirement protein A1 (HTRA1), a serine protease with an established role in the brain vasculature, and several of its substrates. Notably, this signature was not present in AD cases. We further show that HTRA1 co-localizes with Aβ deposits in brain capillaries from CAA type 1 patients indicating a pathologic recruitment process. Together, these findings suggest a central role of HTRA1-dependent protein homeostasis in the CAA microvasculature and a molecular connection between multiple types of brain microvascular disease.
Collapse
Affiliation(s)
- Andreas Zellner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Barbara Lindner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
10
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
11
|
Daoutsali E, Hailu TT, Buijsen RAM, Pepers BA, van der Graaf LM, Verbeek MM, Curtis D, de Vlaam T, van Roon-Mom WMC. Antisense Oligonucleotide-Induced Amyloid Precursor Protein Splicing Modulation as a Therapeutic Approach for Dutch-Type Cerebral Amyloid Angiopathy. Nucleic Acid Ther 2021; 31:351-363. [PMID: 34061681 PMCID: PMC8823675 DOI: 10.1089/nat.2021.0005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dutch-type cerebral amyloid angiopathy (D-CAA) is a monogenic form of cerebral amyloid angiopathy and is inherited in an autosomal dominant manner. The disease is caused by a point mutation in exon 17 of the amyloid precursor protein (APP) gene that leads to an amino acid substitution at codon 693. The mutation is located within the amyloid beta (Aβ) domain of APP, and leads to accumulation of toxic Aβ peptide in and around the cerebral vasculature. We have designed an antisense oligonucleotide (AON) approach that results in skipping of exon 17, generating a shorter APP isoform that lacks part of the Aβ domain and the D-CAA mutation. We demonstrate efficient AON-induced skipping of exon 17 at RNA level and the occurrence of a shorter APP protein isoform in three different cell types. This resulted in a reduction of Aβ40 in neuronally differentiated, patient-derived induced pluripotent stem cells. AON-treated wild-type mice showed successful exon skipping on RNA and protein levels throughout the brain. These results illustrate APP splice modulation as a promising therapeutic approach for D-CAA.
Collapse
Affiliation(s)
- Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Barry A Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Marcel M Verbeek
- Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Daniel Curtis
- Amylon Therapeutics, Leiden, the Netherlands.,Atalanta Therapeutics, Boston, Massachusetts, USA
| | | | | |
Collapse
|
12
|
Singh PK, Badimon A, Chen Z, Strickland S, Norris EH. The contact activation system and vascular factors as alternative targets for Alzheimer's disease therapy. Res Pract Thromb Haemost 2021; 5:e12504. [PMID: 33977208 PMCID: PMC8105157 DOI: 10.1002/rth2.12504] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 03/04/2021] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, affecting millions of people worldwide. Extracellular beta-amyloid (Aβ) plaques and neurofibrillary tau tangles are classical hallmarks of AD pathology and thus are the prime targets for AD therapeutics. However, approaches to slow or stop AD progression and dementia by reducing Aβ production, neutralizing toxic Aβ aggregates, or inhibiting tau aggregation have been largely unsuccessful in clinical trials. The contribution of dysregulated vascular components and inflammation is evident in AD pathology. Vascular changes are detectable early in AD progression, so treatment of vascular defects along with anti-Aβ/tau therapy could be a successful combination therapeutic strategy for this disease. Here, we explain how vascular dysfunction mechanistically contributes to thrombosis as well as inflammation and neurodegeneration in AD pathogenesis. This review provides evidence that addressing vascular dysfunction in people with AD could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNYUSA
| | - Ana Badimon
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNYUSA
| | - Zu‐Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNYUSA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNYUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNYUSA
| |
Collapse
|
13
|
Kumar S, Kapadia A, Theil S, Joshi P, Riffel F, Heneka MT, Walter J. Novel Phosphorylation-State Specific Antibodies Reveal Differential Deposition of Ser26 Phosphorylated Aβ Species in a Mouse Model of Alzheimer's Disease. Front Mol Neurosci 2021; 13:619639. [PMID: 33519377 PMCID: PMC7844098 DOI: 10.3389/fnmol.2020.619639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Aggregation and deposition of amyloid-β (Aβ) peptides in extracellular plaques and in the cerebral vasculature are prominent neuropathological features of Alzheimer's disease (AD) and closely associated with the pathogenesis of AD. Amyloid plaques in the brains of most AD patients and transgenic mouse models exhibit heterogeneity in the composition of Aβ deposits, due to the occurrence of elongated, truncated, and post-translationally modified Aβ peptides. Importantly, changes in the deposition of these different Aβ variants are associated with the clinical disease progression and considered to mark sequential phases of plaque and cerebral amyloid angiopathy (CAA) maturation at distinct stages of AD. We recently showed that Aβ phosphorylated at serine residue 26 (pSer26Aβ) has peculiar characteristics in aggregation, deposition, and neurotoxicity. In the current study, we developed and thoroughly validated novel monoclonal and polyclonal antibodies that recognize Aβ depending on the phosphorylation-state of Ser26. Our results demonstrate that selected phosphorylation state-specific antibodies were able to recognize Ser26 phosphorylated and non-phosphorylated Aβ with high specificity in enzyme-linked immunosorbent assay (ELISA) and Western Blotting (WB) assays. Furthermore, immunofluorescence analyses with these antibodies demonstrated the occurrence of pSer26Aβ in transgenic mouse brains that show differential deposition as compared to non-phosphorylated Aβ (npAβ) or other modified Aβ species. Notably, pSer26Aβ species were faintly detected in extracellular Aβ plaques but most prominently found intraneuronally and in cerebral blood vessels. In conclusion, we developed new antibodies to specifically differentiate Aβ peptides depending on the phosphorylation state of Ser26, which are applicable in ELISA, WB, and immunofluorescence staining of mouse brain tissues. These site- and phosphorylation state-specific Aβ antibodies represent novel tools to examine phosphorylated Aβ species to further understand and dissect the complexity in the age-related and spatio-temporal deposition of different Aβ variants in transgenic mouse models and human AD brains.
Collapse
Affiliation(s)
- Sathish Kumar
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Akshay Kapadia
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Sandra Theil
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Pranav Joshi
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Florian Riffel
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Michael T. Heneka
- Department of Neurodegenerative Diseases and Geropsychiatry, Neurology, University of Bonn Medical Center, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jochen Walter
- Department of Neurology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
14
|
Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, Kalaria RN, Katsumata Y, Gold BT, Wang DJJ, Ma SJ, Shade LMP, Fardo DW, Hartz AMS, Jicha GA, Nelson KB, Magaki SD, Schmitt FA, Teylan MA, Ighodaro ET, Phe P, Abner EL, Cykowski MD, Van Eldik LJ, Nelson PT. Brain arteriolosclerosis. Acta Neuropathol 2021; 141:1-24. [PMID: 33098484 PMCID: PMC8503820 DOI: 10.1007/s00401-020-02235-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Brain arteriolosclerosis (B-ASC), characterized by pathologic arteriolar wall thickening, is a common finding at autopsy in aged persons and is associated with cognitive impairment. Hypertension and diabetes are widely recognized as risk factors for B-ASC. Recent research indicates other and more complex risk factors and pathogenetic mechanisms. Here, we describe aspects of the unique architecture of brain arterioles, histomorphologic features of B-ASC, relevant neuroimaging findings, epidemiology and association with aging, established genetic risk factors, and the co-occurrence of B-ASC with other neuropathologic conditions such as Alzheimer's disease and limbic-predominant age-related TDP-43 encephalopathy (LATE). There may also be complex physiologic interactions between metabolic syndrome (e.g., hypertension and inflammation) and brain arteriolar pathology. Although there is no universally applied diagnostic methodology, several classification schemes and neuroimaging techniques are used to diagnose and categorize cerebral small vessel disease pathologies that include B-ASC, microinfarcts, microbleeds, lacunar infarcts, and cerebral amyloid angiopathy (CAA). In clinical-pathologic studies that factored in comorbid diseases, B-ASC was independently associated with impairments of global cognition, episodic memory, working memory, and perceptual speed, and has been linked to autonomic dysfunction and motor symptoms including parkinsonism. We conclude by discussing critical knowledge gaps related to B-ASC and suggest that there are probably subcategories of B-ASC that differ in pathogenesis. Observed in over 80% of autopsied individuals beyond 80 years of age, B-ASC is a complex and under-studied contributor to neurologic disability.
Collapse
Affiliation(s)
- Brittney L Blevins
- Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Seth Love
- University of Bristol and Southmead Hospital, Bristol, BS10 5NB, UK
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- LIM-22, Department of Pathology, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Samantha J Ma
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Lincoln M P Shade
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University Kentucky, Lexington, KY, 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | | | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, University Washington, Seattle, WA, 98105, USA
| | | | - Panhavuth Phe
- Sanders-Brown Center on Aging, University Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, Department of Epidemiology, University Kentucky, Lexington, KY, 40536, USA
| | - Matthew D Cykowski
- Departments of Pathology and Genomic Medicine and Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA.
- Rm 311 Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
15
|
Kumar S, Lemere CA, Walter J. Phosphorylated Aβ peptides in human Down syndrome brain and different Alzheimer's-like mouse models. Acta Neuropathol Commun 2020; 8:118. [PMID: 32727580 PMCID: PMC7388542 DOI: 10.1186/s40478-020-00959-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
The deposition of neurotoxic amyloid-β (Aβ) peptides in extracellular plaques in the brain parenchyma is one of the most prominent neuropathological features of Alzheimer's disease (AD), and considered to be closely related to the pathogenesis of this disease. A number of recent studies demonstrate the heterogeneity in the composition of Aβ deposits in AD brains, due to the occurrence of elongated, truncated and post-translationally modified Aβ peptides that have peculiar characteristics in aggregation behavior and biostability. Importantly, the detection of modified Aβ species has been explored to characterize distinct stages of AD, with phosphorylated Aβ being present in the clinical phase of AD. People with Down syndrome (DS) develop AD pathology by 40 years of age likely due to the overproduction of Aβ caused by the additional copy of the gene encoding the amyloid precursor protein on chromosome 21. In the current study, we analysed the deposition of phosphorylated and non-phosphorylated Aβ species in human DS, AD, and control brains. In addition, deposition of these Aβ species was analysed in brains of a series of established transgenic AD mouse models using phosphorylation-state specific Aβ antibodies. Significant amounts of Aβ phosphorylated at serine residue 8 (pSer8Aβ) and unmodified Aβ were detected in the brains of DS and AD cases. The brains of different transgenic mouse models with either only human mutant amyloid precursor protein (APP), or combinations of human mutant APP, Presenilin (PS), and tau transgenes showed distinct age-dependent and spatiotemporal deposition of pSer8Aβ in extracellular plaques and within the vasculature. Together, these results demonstrate the deposition of phosphorylated Aβ species in DS brains, further supporting the similarity of Aβ deposition in AD and DS. Thus, the detection of phosphorylated and other modified Aβ species could contribute to the understanding and dissection of the complexity in the age-related and spatiotemporal deposition of Aβ variants in AD and DS as well as in distinct mouse models.
Collapse
|
16
|
Gatti L, Tinelli F, Scelzo E, Arioli F, Di Fede G, Obici L, Pantoni L, Giaccone G, Caroppo P, Parati EA, Bersano A. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int J Mol Sci 2020; 21:ijms21103435. [PMID: 32414028 PMCID: PMC7279405 DOI: 10.3390/ijms21103435] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), one of the main types of cerebral small vessel disease, is a major cause of spontaneous intracerebral haemorrhage and an important contributor to cognitive decline in elderly patients. Despite the number of experimental in vitro studies and animal models, the pathophysiology of CAA is still largely unknown. Although several pathogenic mechanisms including an unbalance between production and clearance of amyloid beta (Aβ) protein as well as ‘the prion hypothesis’ have been invoked as possible disease triggers, they do not explain completely the disease pathogenesis. This incomplete disease knowledge limits the implementation of treatments able to prevent or halt the clinical progression. The continuous increase of CAA patients makes imperative the development of suitable experimental in vitro or animal models to identify disease biomarkers and new pharmacological treatments that could be administered in the early disease stages to prevent irreversible changes and disease progression.
Collapse
Affiliation(s)
- Laura Gatti
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Francesca Tinelli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Emma Scelzo
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Francesco Arioli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Giuseppe Di Fede
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Laura Obici
- Amyloidosis Research and Treatment Centre, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Leonardo Pantoni
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Giorgio Giaccone
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Paola Caroppo
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Eugenio Agostino Parati
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
- Correspondence: ; Tel.: +39-0223943310
| |
Collapse
|
17
|
Marini S, Anderson CD, Rosand J. Genetics of Cerebral Small Vessel Disease. Stroke 2020; 51:12-20. [PMID: 31752611 PMCID: PMC7337039 DOI: 10.1161/strokeaha.119.024151] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Sandro Marini
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher D. Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
18
|
Grand Moursel L, van der Graaf LM, Bulk M, van Roon‐Mom WM, van der Weerd L. Osteopontin and phospho-SMAD2/3 are associated with calcification of vessels in D-CAA, an hereditary cerebral amyloid angiopathy. Brain Pathol 2019; 29:793-802. [PMID: 30868685 PMCID: PMC6850614 DOI: 10.1111/bpa.12721] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/06/2019] [Indexed: 01/06/2023] Open
Abstract
In severe forms of cerebral amyloid angiopathy (CAA) pathology, vascular calcification has been observed in the cerebral cortex, both in vivo on MRI and CT, and post-mortem using histopathology. However, the pathomechanisms leading to calcification of CAA-laden arteries are unknown. Therefore, we investigated the correlation between calcification of cortical arterioles and several potential modulators of vascular calcification using immunohistochemistry in a unique collection of brain material of patients with a hereditary form of CAA, namely hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D or D-CAA). We show a topographical association of osteopontin (OPN) and TGFβ signaling factor phospho-SMAD2/3 (pSMAD2/3) in calcified CAA vessel walls. OPN and pSMAD2/3 gradually accumulate in vessels prior to calcification. Moreover, we found that the vascular accumulation of Collagen 1 (Col1), OPN and pSMAD2/3 immunomarkers correlated with the CAA severity. This was independently of the vessel size, including capillaries in the most severe cases. We propose that calcification of CAA vessels in the observed HCHWA-D cases may be induced by extracellular OPN trapped in the fibrotic Col1 vessel wall, independently of the presence of vascular amyloid.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Linda M. van der Graaf
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Marjolein Bulk
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | | | - Louise van der Weerd
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
19
|
Schelle J, Wegenast-Braun BM, Fritschi SK, Kaeser SA, Jährling N, Eicke D, Skodras A, Beschorner N, Obermueller U, Häsler LM, Wolfer DP, Mueggler T, Shimshek DR, Neumann U, Dodt HU, Staufenbiel M, Jucker M. Early Aβ reduction prevents progression of cerebral amyloid angiopathy. Ann Neurol 2019; 86:561-571. [PMID: 31359452 DOI: 10.1002/ana.25562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Clinical trials targeting β-amyloid peptides (Aβ) for Alzheimer disease (AD) failed for arguable reasons that include selecting the wrong stages of AD pathophysiology or Aβ being the wrong target. Targeting Aβ to prevent cerebral amyloid angiopathy (CAA) has not been rigorously followed, although the causal role of Aβ for CAA and related hemorrhages is undisputed. CAA occurs with normal aging and to various degrees in AD, where its impact and treatment is confounded by the presence of parenchymal Aβ deposition. METHODS APPDutch mice develop CAA in the absence of parenchymal amyloid, mimicking hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). Mice were treated with a β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitor. We used 3-dimensional ultramicroscopy and immunoassays for visualizing CAA and assessing Aβ in cerebrospinal fluid (CSF) and brain. RESULTS CAA onset in mice was at 22 to 24 months, first in frontal leptomeningeal and superficial cortical vessels followed by vessels penetrating the cortical layers. CSF Aβ increased with aging followed by a decrease of both Aβ40 and Aβ42 upon CAA onset, supporting the idea that combined reduction of CSF Aβ40 and Aβ42 is a specific biomarker for vascular amyloid. BACE1 inhibitor treatment starting at CAA onset and continuing for 4 months revealed a 90% Aβ reduction in CSF and largely prevented CAA progression and associated pathologies. INTERPRETATION This is the first study showing that Aβ reduction at early disease time points largely prevents CAA in the absence of parenchymal amyloid. Our observation provides a preclinical basis for Aβ-reducing treatments in patients at risk of CAA and in presymptomatic HCHWA-D. ANN NEUROL 2019;86:561-571.
Collapse
Affiliation(s)
- Juliane Schelle
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Bettina M Wegenast-Braun
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Sarah K Fritschi
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan A Kaeser
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Nina Jährling
- TU Wien, Vienna, Austria.,Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniel Eicke
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Angelos Skodras
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Natalie Beschorner
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ulrike Obermueller
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lisa M Häsler
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - David P Wolfer
- Institute of Anatomy, University of Zürich, Zürich, Switzerland
| | - Thomas Mueggler
- Institute for Biomedical Engineering, University and Swiss Federal Institute for Technology, Zürich, Switzerland
| | | | - Ulf Neumann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Hans-Ulrich Dodt
- TU Wien, Vienna, Austria.,Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Matthias Staufenbiel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Daoutsali E, Buijsen RA, van de Pas S, Jong A', Mikkers H, Brands T, Eussen B, de Klein A, van der Graaf LM, Pepers BA, Freund C, Terwindt GM, Orlova VV, van Roon-Mom WM. Generation of 3 human induced pluripotent stem cell lines LUMCi005-A, B and C from a Hereditary Cerebral Hemorrhage with Amyloidosis-Dutch type patient. Stem Cell Res 2019; 34:101359. [DOI: 10.1016/j.scr.2018.101359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022] Open
|
21
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1315] [Impact Index Per Article: 219.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
22
|
Bulk M, Moursel LG, van der Graaf LM, van Veluw SJ, Greenberg SM, van Duinen SG, van Buchem MA, van Rooden S, van der Weerd L. Cerebral Amyloid Angiopathy With Vascular Iron Accumulation and Calcification. Stroke 2018; 49:2081-2087. [DOI: 10.1161/strokeaha.118.021872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Marjolein Bulk
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
- Department of Human Genetics (M.B., L.G.M., L.M.v.d.G., L.v.d.W.)
| | - Laure Grand Moursel
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
- Department of Human Genetics (M.B., L.G.M., L.M.v.d.G., L.v.d.W.)
| | - Linda M. van der Graaf
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
- Department of Human Genetics (M.B., L.G.M., L.M.v.d.G., L.v.d.W.)
| | - Susanne J. van Veluw
- Leiden University Medical Center, the Netherlands; and Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (S.J.v.V., S.M.G.)
| | - Steven M. Greenberg
- Leiden University Medical Center, the Netherlands; and Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (S.J.v.V., S.M.G.)
| | | | - Mark A. van Buchem
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
| | - Sanneke van Rooden
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
| | - Louise van der Weerd
- From the Department of Radiology (M.B., L.G.M., L.M.v.d.G., M.A.v.B., S.v.R., L.v.d.W.)
- Department of Human Genetics (M.B., L.G.M., L.M.v.d.G., L.v.d.W.)
| |
Collapse
|
23
|
Grand Moursel L, van Roon-Mom WMC, Kiełbasa SM, Mei H, Buermans HPJ, van der Graaf LM, Hettne KM, de Meijer EJ, van Duinen SG, Laros JFJ, van Buchem MA, 't Hoen PAC, van der Maarel SM, van der Weerd L. Brain Transcriptomic Analysis of Hereditary Cerebral Hemorrhage With Amyloidosis-Dutch Type. Front Aging Neurosci 2018; 10:102. [PMID: 29706885 PMCID: PMC5908973 DOI: 10.3389/fnagi.2018.00102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/26/2018] [Indexed: 11/23/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) caused by a point mutation resulting in an amino acid change (NP_000475.1:p.Glu693Gln) in the amyloid precursor protein (APP). Post-mortem frontal and occipital cortical brain tissue from nine patients and nine age-related controls was used for RNA sequencing to identify biological pathways affected in HCHWA-D. Although previous studies indicated that pathology is more severe in the occipital lobe in HCHWA-D compared to the frontal lobe, the current study showed similar changes in gene expression in frontal and occipital cortex and the two brain regions were pooled for further analysis. Significantly altered pathways were analyzed using gene set enrichment analysis (GSEA) on 2036 significantly differentially expressed genes. Main pathways over-represented by down-regulated genes were related to cellular aerobic respiration (including ATP synthesis and carbon metabolism) indicating a mitochondrial dysfunction. Principal up-regulated pathways were extracellular matrix (ECM)–receptor interaction and ECM proteoglycans in relation with an increase in the transforming growth factor beta (TGFβ) signaling pathway. Comparison with the publicly available dataset from pre-symptomatic APP-E693Q transgenic mice identified overlap for the ECM–receptor interaction pathway, indicating that ECM modification is an early disease specific pathomechanism.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Szymon M Kiełbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, Netherlands
| | - Hailiang Mei
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, Netherlands
| | - Henk P J Buermans
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Kristina M Hettne
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Emile J de Meijer
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen F J Laros
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
24
|
Animal models of cerebral amyloid angiopathy. Clin Sci (Lond) 2017; 131:2469-2488. [PMID: 28963121 DOI: 10.1042/cs20170033] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 02/04/2023]
Abstract
Cerebral amyloid angiopathy (CAA), due to vascular amyloid β (Aβ) deposition, is a risk factor for intracerebral haemorrhage and dementia. CAA can occur in sporadic or rare hereditary forms, and is almost invariably associated with Alzheimer's disease (AD). Experimental (animal) models are of great interest in studying mechanisms and potential treatments for CAA. Naturally occurring animal models of CAA exist, including cats, dogs and non-human primates, which can be used for longitudinal studies. However, due to ethical considerations and low throughput of these models, other animal models are more favourable for research. In the past two decades, a variety of transgenic mouse models expressing the human Aβ precursor protein (APP) has been developed. Many of these mouse models develop CAA in addition to senile plaques, whereas some of these models were generated specifically to study CAA. In addition, other animal models make use of a second stimulus, such as hypoperfusion or hyperhomocysteinemia (HHcy), to accelerate CAA. In this manuscript, we provide a comprehensive review of existing animal models for CAA, which can aid in understanding the pathophysiology of CAA and explore the response to potential therapies.
Collapse
|
25
|
Grand Moursel L, Munting LP, van der Graaf LM, van Duinen SG, Goumans MJTH, Ueberham U, Natté R, van Buchem MA, van Roon-Mom WMC, van der Weerd L. TGFβ pathway deregulation and abnormal phospho-SMAD2/3 staining in hereditary cerebral hemorrhage with amyloidosis-Dutch type. Brain Pathol 2017; 28:495-506. [PMID: 28557134 PMCID: PMC8028662 DOI: 10.1111/bpa.12533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/19/2017] [Indexed: 12/20/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis‐Dutch type (HCHWA‐D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) pathology, caused by the E22Q mutation in the amyloid β (Aβ) peptide. Transforming growth factor β1 (TGFβ1) is a key player in vascular fibrosis and in the formation of angiopathic vessels in transgenic mice. Therefore, we investigated whether the TGFβ pathway is involved in HCHWA‐D pathogenesis in human postmortem brain tissue from frontal and occipital lobes. Components of the TGFβ pathway were analyzed with quantitative RT‐PCR. TGFβ1 and TGFβ Receptor 2 (TGFBR2) gene expression levels were significantly increased in HCHWA‐D in comparison to the controls, in both frontal and occipital lobes. TGFβ‐induced pro‐fibrotic target genes were also upregulated. We further assessed pathway activation by detecting phospho‐SMAD2/3 (pSMAD2/3), a direct TGFβ down‐stream signaling mediator, using immunohistochemistry. We found abnormal pSMAD2/3 granular deposits specifically on HCHWA‐D angiopathic frontal and occipital vessels. We graded pSMAD2/3 accumulation in angiopathic vessels and found a positive correlation with the CAA load independent of the brain area. We also observed pSMAD2/3 granules in a halo surrounding occipital vessels, which was specific for HCHWA‐D. The result of this study indicates an upregulation of TGFβ1 in HCHWA‐D, as was found previously in AD with CAA pathology. We discuss the possible origins and implications of the TGFβ pathway deregulation in the microvasculature in HCHWA‐D. These findings identify the TGFβ pathway as a potential biomarker of disease progression and a possible target of therapeutic intervention in HCHWA‐D.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Leon P Munting
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Uwe Ueberham
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Remco Natté
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
26
|
Drummond E, Wisniewski T. Alzheimer's disease: experimental models and reality. Acta Neuropathol 2017; 133:155-175. [PMID: 28025715 PMCID: PMC5253109 DOI: 10.1007/s00401-016-1662-x] [Citation(s) in RCA: 468] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/15/2022]
Abstract
Experimental models of Alzheimer's disease (AD) are critical to gaining a better understanding of pathogenesis and to assess the potential of novel therapeutic approaches. The most commonly used experimental animal models are transgenic mice that overexpress human genes associated with familial AD (FAD) that result in the formation of amyloid plaques. However, AD is defined by the presence and interplay of both amyloid plaques and neurofibrillary tangle pathology. The track record of success in AD clinical trials thus far has been very poor. In part, this high failure rate has been related to the premature translation of highly successful results in animal models that mirror only limited aspects of AD pathology to humans. A greater understanding of the strengths and weakness of each of the various models and the use of more than one model to evaluate potential therapies would help enhance the success of therapy translation from preclinical studies to patients. In this review, we summarize the pathological features and limitations of the major experimental models of AD, including transgenic mice, transgenic rats, various physiological models of sporadic AD and in vitro human cell culture models.
Collapse
Affiliation(s)
- Eleanor Drummond
- Center for Cognitive Neurology and Department of Neurology, NYU School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY, 10016, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, Pathology and Psychiatry, NYU School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY, 10016, USA.
| |
Collapse
|
27
|
Muenchhoff J, Poljak A, Thalamuthu A, Gupta VB, Chatterjee P, Raftery M, Masters CL, Morris JC, Bateman RJ, Fagan AM, Martins RN, Sachdev PS. Changes in the plasma proteome at asymptomatic and symptomatic stages of autosomal dominant Alzheimer's disease. Sci Rep 2016; 6:29078. [PMID: 27381087 PMCID: PMC4933916 DOI: 10.1038/srep29078] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 06/10/2016] [Indexed: 01/18/2023] Open
Abstract
The autosomal dominant form of Alzheimer's disease (ADAD) is far less prevalent than late onset Alzheimer's disease (LOAD), but enables well-informed prospective studies, since symptom onset is near certain and age of onset is predictable. Our aim was to discover plasma proteins associated with early AD pathology by investigating plasma protein changes at the asymptomatic and symptomatic stages of ADAD. Eighty-one proteins were compared across asymptomatic mutation carriers (aMC, n = 15), symptomatic mutation carriers (sMC, n = 8) and related noncarriers (NC, n = 12). Proteins were also tested for associations with cognitive measures, brain amyloid deposition and glucose metabolism. Fewer changes were observed at the asymptomatic than symptomatic stage with seven and 16 proteins altered significantly in aMC and sMC, respectively. This included complement components C3, C5, C6, apolipoproteins A-I, A-IV, C-I and M, histidine-rich glycoprotein, heparin cofactor II and attractin, which are involved in inflammation, lipid metabolism and vascular health. Proteins involved in lipid metabolism differed only at the symptomatic stage, whereas changes in inflammation and vascular health were evident at asymptomatic and symptomatic stages. Due to increasing evidence supporting the usefulness of ADAD as a model for LOAD, these proteins warrant further investigation into their potential association with early stages of LOAD.
Collapse
Affiliation(s)
- Julia Muenchhoff
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Veer B. Gupta
- Centre of Excellence for Alzheimer’s disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Sir James McCusker Alzheimer’s Disease Research Unit (Hollywood Private Hospital), Perth, Western Australia, Australia
| | - Pratishtha Chatterjee
- Centre of Excellence for Alzheimer’s disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Sir James McCusker Alzheimer’s Disease Research Unit (Hollywood Private Hospital), Perth, Western Australia, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, Western Australia, Australia
| | - Mark Raftery
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, New South Wales, Australia
| | | | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, USA
- Knight Alzheimer’s Disease Research Center at Washington University School of Medicine, St. Louis, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, USA
- Knight Alzheimer’s Disease Research Center at Washington University School of Medicine, St. Louis, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, USA
- Knight Alzheimer’s Disease Research Center at Washington University School of Medicine, St. Louis, USA
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Sir James McCusker Alzheimer’s Disease Research Unit (Hollywood Private Hospital), Perth, Western Australia, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, Western Australia, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|