1
|
Freund BE, Shourav MMI, Feyissa AM, Meschia JF, Yonas A, Barrett KM, Tatum WO, Lin MP. Seizures in Cerebral Amyloid Angiopathy: A Systematic Review and Meta-Analysis. Neurol Clin Pract 2025; 15:e200454. [PMID: 40161255 PMCID: PMC11952699 DOI: 10.1212/cpj.0000000000200454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/16/2025] [Indexed: 04/02/2025]
Abstract
Purpose of Review Cerebral amyloid angiopathy (CAA) is a disease of the cerebral vasculature that can result in microhemorrhages, as well as intraparenchymal and subarachnoid hemorrhage, superficial siderosis (SS), and/or secondary infarct/inflammation. CAA may be encountered as an isolated pathology or with Alzheimer disease and has been demonstrated to be associated with an increased risk of seizures. However, the overall rates of seizures and specific pathologies related to CAA and their subsequent risk of seizures have not been elucidated. Recent findings Prior studies of CAA and seizures are predominantly case reports or small case series, and larger studies have focused primarily on smaller subgroups of patients with CAA. Only 2 prior studies assessed larger heterogeneous populations of patients with CAA. One study focused on long-term outcomes and evaluated the impact of seizures on cognitive and survival outcomes retrospectively, although it did not delineate the effects of acute and chronic seizure disorders (epilepsy) and did not find an association. Long-term prospective or retrospective studies on outcomes regarding seizures/epilepsy and CAA are therefore lacking. Summary A total of 1,376 articles were identified, with 48 (34 case reports/series and 14 cohort studies) included in this review. Acute symptomatic seizures (ASyS) and epilepsy were poorly defined, and the overall prevalence of seizures in cohort studies was 22.8%, with significant heterogeneity (I 2 = 77%; p < 0.01). Epilepsy was diagnosed in 34.4% and ASyS in 10.6% of patients in heterogeneous cohorts. Most of the studies assessed seizures in specific subgroups of CAA with variable prevalence, including CAA with related inflammation (CAA-ri): 56.9%; lobar intracranial hemorrhage (ICH): 17.1%; and cortical SAH (cSAH) or SS: 8.7%. In heterogeneous cohorts, SS (p < 0.001 and p = 0.03, respectively) and CAA-ri (p = 0.005 and p = 0.04, respectively) were significantly associated with epilepsy/seizures. In 1 study, cSAH (p = 0.03) and acute lobar ICH (p = 0.002) were associated with seizures, likely related to inclusion of ASyS. Status epilepticus (14/125) and drug resistance (6/89) were infrequent. Clinical pathologic entities associated with a risk of seizures include cSAH, CAA-ri, SS, and acute ICH.
Collapse
Affiliation(s)
- Brin E Freund
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | | | | | | | - Amen Yonas
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | | | | | | |
Collapse
|
2
|
Fatima G, Ashiquzzaman A, Kim SS, Kim YR, Kwon HS, Chung E. Vascular and glymphatic dysfunction as drivers of cognitive impairment in Alzheimer's disease: Insights from computational approaches. Neurobiol Dis 2025; 208:106877. [PMID: 40107629 DOI: 10.1016/j.nbd.2025.106877] [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: 01/16/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Alzheimer's disease (AD) is driven by complex interactions between vascular dysfunction, glymphatic system impairment, and neuroinflammation. Vascular aging, characterized by arterial stiffness and reduced cerebral blood flow (CBF), disrupts the pulsatile forces necessary for glymphatic clearance, exacerbating amyloid-beta (Aβ) accumulation and cognitive decline. This review synthesizes insights into the mechanistic crosstalk between these systems and explores their contributions to AD pathogenesis. Emerging machine learning (ML) tools, such as DeepLabCut and Motion sequencing (MoSeq), offer innovative solutions for analyzing multimodal data and enhancing diagnostic precision. Integrating ML with imaging and behavioral analyses bridges gaps in understanding vascular-glymphatic dysfunction. Future research must prioritize these interactions to develop early diagnostics and targeted interventions, advancing our understanding of neurovascular health in AD.
Collapse
Affiliation(s)
- Gehan Fatima
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Akm Ashiquzzaman
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Sang Seong Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Young Ro Kim
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Hyuk-Sang Kwon
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea; AI Graduate School, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Rep. of Korea; Research Center for Photon Science Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea.
| | - Euiheon Chung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea; AI Graduate School, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Rep. of Korea; Research Center for Photon Science Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea.
| |
Collapse
|
3
|
Villain N, Planche V, Lilamand M, Cordonnier C, Soto-Martin M, Mollion H, Bombois S, Delrieu J. Lecanemab for early Alzheimer's disease: Appropriate use recommendations from the French federation of memory clinics. J Prev Alzheimers Dis 2025; 12:100094. [PMID: 40011173 DOI: 10.1016/j.tjpad.2025.100094] [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: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
Lecanemab, a monoclonal antibody targeting β-amyloid protofibrils, has shown promising results in a Phase III clinical trial for the treatment of early stages of Alzheimer's disease (AD) and has been approved by the European Medicines Agency. An Early Market Authorization could be submitted to the French regulatory agencies, potentially allowing for the drug's use in clinical practice in France in 2025. To guide French clinicians in administering lecanemab in a standardized way, the French Federation of Memory Clinics has developed appropriate use recommendations for lecanemab that highlight relevant questions established to ensure an optimal risk-benefit ratio. The recommendations emphasize that lecanemab treatment requires a comprehensive individualized evaluation of the risk-benefit ratio, which should occur in multidisciplinary meetings. When approved, the guidelines support the use of blood biomarkers, proposing specific cutoffs for patients eligible for lecanemab under restricted conditions. In addition to the European Medicines Agency restrictions in patients on anticoagulants, and APOE4 homozygotes, the guidelines recommend against lecanemab treatment for patients with high amyloid-related hemorrhagic risk such as probable cerebral amyloid angiopathy (Boston criteria v1.5) until further data become available. Additionally, we recommend that MRI monitoring be started before the third infusion to account for early Amyloid Related Imaging Abnormalities (ARIA) occurring on lecanemab. It is recommended to establish a specific clinical care pathway with protocols for patients with ARIA, with trained physicians and radiologists with expertise in neurological emergency and intensive care. Finally, a discontinuation protocol based on dementia severity assessment after 18 months of lecanemab treatment is suggested. Access to lecanemab requires a personalized biological and genetic diagnosis of AD, which is currently not necessary in most cases. Therefore, the healthcare system must rapidly adjust to new diagnostic procedures and treatment delivery to ensure equal access for all individuals.
Collapse
Affiliation(s)
- Nicolas Villain
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, Paris, France; AP-HP Sorbonne Université, Hôpital Pitié-Salpêtrière, Department of Neurology, Institute of Memory and Alzheimer's Disease, Paris, France.
| | - Vincent Planche
- Univ. Bordeaux, CNRS, UMR 5293, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France; Centre Mémoire Ressources Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Matthieu Lilamand
- Université Paris Cité, INSERM UMR S-1144, Paris, France; AP-HP. Nord Université Paris Cité Department of Geriatrics and Cognitive Neurology Center, Lariboisière-Fernand Widal Hospital, Paris, France
| | - Charlotte Cordonnier
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Maria Soto-Martin
- Maintain Aging Research team, CERPOP, INSERM UMR 1295, Universite Paul Sabatier, Toulouse, France; Centre Mémoire Ressources Recherches de Toulouse, Pôle Gériatrie, Cité de la santé, Toulouse CHU, Toulouse, France
| | - Hélène Mollion
- Centre Mémoire Ressources Recherches de Lyon - Hôpital Neurologique - Hospices Civils de Lyon - F 69677 BRON cedex, France
| | - Stéphanie Bombois
- AP-HP Sorbonne Université, Hôpital Pitié-Salpêtrière, Department of Neurology, Institute of Memory and Alzheimer's Disease, Paris, France
| | - Julien Delrieu
- Maintain Aging Research team, CERPOP, INSERM UMR 1295, Universite Paul Sabatier, Toulouse, France; Centre Mémoire Ressources Recherches de Toulouse, Pôle Gériatrie, Cité de la santé, Toulouse CHU, Toulouse, France
| |
Collapse
|
4
|
Shen Y, Nie Q, Xiang W, Chen S, Cao Q, Hong D. The relationship between Alzheimer's disease and intracerebral hemorrhage based on Mendelian randomization. J Alzheimers Dis 2025; 104:1154-1166. [PMID: 40151895 DOI: 10.1177/13872877251323294] [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: 03/29/2025]
Abstract
BackgroundTraditional epidemiologic studies suggest that Alzheimer's disease (AD) may be associated with intracerebral hemorrhage (ICH).ObjectiveTo explore whether there is a causal relationship between AD and ICH and the underlying mechanisms involved.MethodsMendelian randomization (MR) approach was used to explore causal relationships. The genetic instrumental variables of the candidate genetic instrumental variable AD were obtained from genome-wide association studies. The inverse variance weighted method was the primary method for MR analysis and meta-analysis. The obtained single nucleotide polymorphisms were analyzed for corresponding genes for subsequent pathway enrichment and protein-protein interaction analysis.ResultsFor the single AD dataset, our MR analysis of the AD datasets versus the ICH datasets revealed a genetically predicted causal relationship between AD and ICH (OR 5.947, 95%CI 1.165-30.356, pIVW = 0.032). In addition, the MR-Egger method and MR-PRESSO method revealed no horizontal pleiotropic effect of AD on the risk of ICH. Meta-analysis of each dataset using IVW revealed a final calculated OR of 1.08 (95%CI 1.02-1.15, p = 0.01). Subsequent pathway enrichment analysis revealed that the corresponding genes were involved mainly in the metabolic process of amyloid-β (Aβ) and negatively regulated Aβ formation. In the PPI network analysis, proteins such as ApoE, SROL1, CLU, ABCA7, and AβPP were found to be closely related and located in the key position of the center.ConclusionsWe verified the causal relationship between AD and ICH via MR, and identified the possible pathological mechanisms involved. We also discovered that Aβ plays an important role in this process.
Collapse
Affiliation(s)
- Yu Shen
- Department of Neurology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Quirui Nie
- Department of Gerontology, Nanchang First Hospital (the Third Affiliated Hospital, Jiangxi Medical College, Nanchang University), Nanchang, China
| | - WenWen Xiang
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shenjian Chen
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qian Cao
- Department of Neurology, Saarland University, Homburg, Germany
| | - Daojun Hong
- Department of Neurology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Yin X, Sun J, Wang X, Wu W, Chen Z, Zhang D, Xu Y, Chen Y, Qiu W, Qian X, Ni J, Ma C. Prevalence of cerebral amyloid angiopathy and its correlation with Alzheimer's disease and cognition in an autopsy-confirmed cohort from China. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70100. [PMID: 40201593 PMCID: PMC11973253 DOI: 10.1002/dad2.70100] [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: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND We aimed to investigate the prevalence of cerebral amyloid angiopathy (CAA) and its correlations with Alzheimer's disease (AD) and cognitive impairment in an autopsy-confirmed cohort donated to a human brain bank in Beijing, China. METHODS A total of 483 subjects were neuropathologically evaluated based on standardized protocols. Descriptive statistics and ordinal logistic regression models were used to estimate the correlation between CAA, AD, apolipoprotein E (APOE) genotyping, and cognitive function proximal to death. RESULTS Neuropathological assessment revealed that 53 of 483 subjects (11%) had CAA without AD, 78 of 483 (16%) had AD without CAA, 98 of 483 (20%) had both CAA and AD, and 254 of 483 (53%) had neither condition. A significant correlation was confirmed between CAA severity and AD. Subjects with both CAA and AD exhibited aggravated cognitive impairment. DISCUSSION Our results indicate a substantial prevalence of CAA that is frequently comorbid with AD and may exacerbate cognitive decline in the elderly population in China. Highlights First reporting of cerebral amyloid angiopathy (CAA) based on an autopsy-confirmed cohort from China.The prevalence of CAA was high in the elderly Chinese sample.Age and apolipoprotein E (APOE) ε4 allele were related to the prevalence of CAA.CAA and Alzheimer's disease (AD) were frequently co-occurred and significantly associated.Subjects with both CAA and AD exhibited aggravated cognitive impairment.
Collapse
Affiliation(s)
- Xiang‐Sha Yin
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Jianru Sun
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xue Wang
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Wei Wu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Zhen Chen
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Di Zhang
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Yuanyuan Xu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yongmei Chen
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Wenying Qiu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Xiaojing Qian
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Jun Ni
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Chao Ma
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| |
Collapse
|
6
|
Bonnar O, Eyre B, van Veluw SJ. Perivascular brain clearance as a therapeutic target in cerebral amyloid angiopathy and Alzheimer's disease. Neurotherapeutics 2025; 22:e00535. [PMID: 39890534 DOI: 10.1016/j.neurot.2025.e00535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Although distinct diseases, both cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD) are characterized by the aggregation and accumulation of amyloid-β (Aβ). This is thought to be due, in part, to impaired perivascular Aβ clearance from the brain. This shared failure in both diseases presents a common opportunity for therapeutic intervention. In this review we discuss the idea that promoting perivascular brain clearance could be an effective strategy for safely reducing Aβ levels in CAA and AD thereby improving clinical outcomes, most notably hemorrhagic stroke and cognitive decline. We will explore the evidence for the different forces that are thought to drive perivascular brain clearance, review the literature on potential strategies for potentiating these driving forces, and finally we will discuss the substantial translational challenges and considerations that would accompany such an intervention.
Collapse
Affiliation(s)
- Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Beth Eyre
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Allen J, Ermine CM, Lin R, Cloud GC, Shultz SR, Casillas-Espinosa PM. Proteinopathies and the Neurodegenerative Aftermath of Stroke: Potential Biomarkers and Treatment Targets. Stroke 2025. [PMID: 40145137 DOI: 10.1161/strokeaha.124.049279] [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: 03/28/2025]
Abstract
Stroke remains a predominant cause of death and long-term disability among adults worldwide. Emerging evidence suggests that proteinopathies, characterized by the aggregation and accumulation of misfolded proteins, may play a significant role in the aftermath of stroke and the progression of neurodegenerative disorders. In this review, we explore preclinical and clinical research on key proteinopathies associated with stroke, including tau, Aβ (amyloid-β), TDP-43 (TAR DNA-binding protein 43), α-synuclein, and UCH-L1 (ubiquitin C-terminal hydrolase-L1). We focus on their potential as biomarkers for recovery management and as novel treatment targets that may enhance neuronal repair and mitigate secondary neurodegeneration. The involvement of these proteinopathies in various aspects of stroke, including neuroinflammation, oxidative stress, neuronal damage, and vascular dysfunction, underscores their potential. However, further investigations are essential to validate the clinical utility of these biomarkers, elucidate the mechanisms connecting proteinopathies to poststroke neurodegeneration, and develop targeted interventions. Identifying specific protein signatures associated with stroke outcomes could facilitate the advancement of precision medicine tailored to individual patient needs, significantly enhancing the quality of life for stroke survivors.
Collapse
Affiliation(s)
- Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia (J.A., R.L., G.C.C., S.R.S., P.M.C.-E.)
| | - Charlotte M Ermine
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia. (C.M.E.)
| | - Runxuan Lin
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia (J.A., R.L., G.C.C., S.R.S., P.M.C.-E.)
| | - Geoffrey C Cloud
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia (J.A., R.L., G.C.C., S.R.S., P.M.C.-E.)
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia (G.C.C., S.R.S., P.M.C.-E.)
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia (J.A., R.L., G.C.C., S.R.S., P.M.C.-E.)
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia. (S.R.S., P.M.C.-E.)
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia (G.C.C., S.R.S., P.M.C.-E.)
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia (J.A., R.L., G.C.C., S.R.S., P.M.C.-E.)
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia. (S.R.S., P.M.C.-E.)
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia (G.C.C., S.R.S., P.M.C.-E.)
| |
Collapse
|
8
|
Chu C, Wang Y, Ma L, Mutimer CA, Ji G, Shi H, Yassi N, Masters CL, Goudey B, Jin L, Pan Y. Developing and validating a prediction tool for cerebral amyloid angiopathy neuropathological severity. Alzheimers Dement 2025; 21:e14583. [PMID: 40042448 PMCID: PMC11881621 DOI: 10.1002/alz.14583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/04/2024] [Accepted: 01/12/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) is a cerebrovascular condition, the severity of which can only be determined post mortem. Here, we developed machine learning models, the Florey CAA Score (FCAAS), to predict CAA severity (none/mild/moderate/severe). METHODS Building on an auto-score-ordinal algorithm, the FCAAS models were developed and validated using data collected by three cohort studies of aging and dementia. The developed FCAAS models were digitized as a web-based tool. A pilot trial was conducted using this web-based tool. RESULTS The FCAAS-4 achieved a mean area under the receiver operating characteristic curve (AUC-ROC) of 0.74 (95% confidence interval: 0.71-0.77) and a Harrell generalized c-index of 0.72 (0.70-0.75). Pilot trial results obtained a mean AUC-ROC of 0.82 (0.71-0.85) and Harrell generalized c-index 0.79 (0.73-0.82). DISCUSSION The FCAAS models demonstrate a promising performance in predicting CAA severity. This framework holds the potential for predicting development of amyloid-related imaging abnormalities (ARIAs), given the CAA-ARIAs link. HIGHLIGHTS The severity of cerebral amyloid angiopathy (CAA) can only be determined post mortem. A web tool, the Florey CAA Score (FCAAS), was developed to predict CAA severity. The FCAAS holds the potential to be used for CAA risk stratification in clinics. CAA is linked to increased risk of amyloid-related imaging abnormalities (ARIAs). The framework used by FCAAS can possibly be adapted to predict ARIAs risk.
Collapse
Affiliation(s)
- Chenyin Chu
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Yihan Wang
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Liwei Ma
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Chloe A. Mutimer
- Department of Medicine and NeurologyMelbourne Brain Centre at The Royal Melbourne Hospital, The University of MelbourneParkvilleVictoriaAustralia
| | - Guangyan Ji
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
| | - Huiyu Shi
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
| | - Nawaf Yassi
- Department of Medicine and NeurologyMelbourne Brain Centre at The Royal Melbourne Hospital, The University of MelbourneParkvilleVictoriaAustralia
- Population Health and Immunity DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
| | - Benjamin Goudey
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- The ARC Training Centre in Cognitive Computing for Medical TechnologiesThe University of MelbourneCaltonVictoriaAustralia
| | - Liang Jin
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Yijun Pan
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
9
|
Weidauer S, Hattingen E. Cerebral Amyloid Angiopathy: Clinical Presentation, Sequelae and Neuroimaging Features-An Update. Biomedicines 2025; 13:603. [PMID: 40149580 PMCID: PMC11939913 DOI: 10.3390/biomedicines13030603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
The prevalence of cerebral amyloid angiopathy (CAA) has been shown to increase with age, with rates reported to be around 50-60% in individuals over 80 years old who have cognitive impairment. The disease often presents as spontaneous lobar intracerebral hemorrhage (ICH), which carries a high risk of recurrence, along with transient focal neurologic episodes (TFNE) and progressive cognitive decline, potentially leading to Alzheimer's disease (AD). In addition to ICH, neuroradiologic findings of CAA include cortical and subcortical microbleeds (MB), cortical subarachnoid hemorrhage (cSAH) and cortical superficial siderosis (cSS). Non-hemorrhagic pathologies include dilated perivascular spaces in the centrum semiovale and multiple hyperintense lesions on T2-weighted magnetic resonance imaging (MRI). A definitive diagnosis of CAA still requires histological confirmation. The Boston criteria allow for the diagnosis of a probable or possible CAA by considering specific neurological and MRI findings. The recent version, 2.0, which includes additional non-hemorrhagic MRI findings, increases sensitivity while maintaining the same specificity. The characteristic MRI findings of autoantibody-related CAA-related inflammation (CAA-ri) are similar to the so-called "amyloid related imaging abnormalities" (ARIA) observed with amyloid antibody therapies, presenting in two variants: (a) vasogenic edema and leptomeningeal effusions (ARIA-E) and (b) hemorrhagic lesions (ARIA-H). Clinical and MRI findings enable the diagnosis of a probable or possible CAA-ri, with biopsy remaining the gold standard for confirmation. In contrast to spontaneous CAA-ri, only about 20% of patients treated with monoclonal antibodies who show proven ARIA on MRI also experience clinical symptoms, including headache, confusion, other psychopathological abnormalities, visual disturbances, nausea and vomiting. Recent findings indicate that treatment should be continued in cases of mild ARIA, with ongoing MRI and clinical monitoring. This review offers a concise update on CAA and its associated consequences.
Collapse
Affiliation(s)
- Stefan Weidauer
- Institute of Neuroradiology, Goethe University, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
10
|
Muir RT, Stukas S, Cooper JG, Beaudin AE, McCreary CR, Gee M, Nelles K, Nukala N, Valencia J, Kirmess KM, Black SE, Hill MD, Camicioli R, Wellington CL, Smith EE. Plasma biomarkers distinguish Boston Criteria 2.0 cerebral amyloid angiopathy from healthy controls. Alzheimers Dement 2025; 21:e70010. [PMID: 40156276 PMCID: PMC11953569 DOI: 10.1002/alz.70010] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 01/23/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) is characterized by the deposition of beta-amyloid (Aβ) in small vessels leading to hemorrhagic stroke and dementia. This study examined whether plasma Aβ42/40, phosphorylated-tau (p-tau), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP) differ in CAA and their potential to discriminate Boston Criteria 2.0 probable CAA from healthy controls. METHODS Plasma Aβ42/40, p-tau-181, NfL, and GFAP were quantified using single molecule array (Simoa) and Aβ42/40 was also independently quantified using immunoprecipitation liquid chromatography mass-spectrometry (IPMS). RESULTS Forty-five participants with CAA and 47 healthy controls had available plasma. Aβ42/40 ratios were significantly lower in CAA than healthy controls. While p-tau-181 and NfL were elevated in CAA, GFAP was similar. A combination of Aβ42/40 (Simoa), p-tau-181, and NfL resulted in an area under the curve of 0.90 (95% confidence interval: 0.80, 0.95). DISCUSSION Plasma Aβ42/40, p-tau-181, and NfL differ in those with CAA and together can discriminate CAA from healthy controls. HIGHLIGHTS Participants with CAA had reduced plasma Aβ42/40 ratios compared to controls. Plasma p-tau-181 and NfL concentrations are elevated in CAA compared to controls. Plasma GFAP was similar in CAA and controls. Together, plasma Aβ42/40, p-tau-181, and NfL had excellent discriminability for CAA.
Collapse
Affiliation(s)
- Ryan T. Muir
- Calgary Stroke ProgramUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Community Health Sciences, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
- L.C Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery, and Hurvitz Brain Sciences Program, Sunnybrook Research InstituteUniversity of TorontoNorth YorkOntarioCanada
| | - Sophie Stukas
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Jennifer G. Cooper
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Andrew E. Beaudin
- Hotchkiss Brain Institute, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
| | - Cheryl R. McCreary
- Hotchkiss Brain Institute, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
- Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
| | - Myrlene Gee
- Division of Neurology, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Krista Nelles
- Division of Neurology, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Nikita Nukala
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
| | - Janina Valencia
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | - Sandra E. Black
- L.C Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery, and Hurvitz Brain Sciences Program, Sunnybrook Research InstituteUniversity of TorontoNorth YorkOntarioCanada
| | - Michael D. Hill
- Calgary Stroke ProgramUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Community Health Sciences, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
| | - Richard Camicioli
- Division of Neurology, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada
- Neuroscience and Mental Health InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Eric E. Smith
- Calgary Stroke ProgramUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Community Health Sciences, Departments of RadiologyUniversity of CalgaryCalgaryAlbertaCanada
- Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
11
|
Ma L, Wang Y, Huynh ALH, Amadoru S, Wrigley S, Yates P, Masters CL, Goudey B, Jin L, Pan Y. Exploring epidemiological risk factors for cerebral amyloid angiopathy: Considerations for monoclonal antibody therapy in people with Alzheimer's disease. Alzheimers Dement 2025; 21:e14602. [PMID: 40042470 PMCID: PMC11881626 DOI: 10.1002/alz.14602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) increases the risk of amyloid-related imaging abnormalities in Alzheimer's disease (AD) patients receiving anti-amyloid-beta therapies, emphasizing the need to identify its risk factors. METHODS Data were collected from three cohort studies, and a machine learning model was developed to predict CAA occurrence using the selected risk factors. RESULTS The AD neuropathologic changes (ADNC)-CAA association was significantly positive in the cross-sectional analysis. When stratified by selected risk factors, this association was generally stronger among females, smokers, people with a history of stroke/memory complaints, apolipoprotein E (APOE)-ε4 carriers, and those without diabetes/heart conditions. In the longitudinal analysis of the association between potential risk factors and CAA, a higher risk of CAA was observed among males, older individuals, smokers, people with diabetes/heart conditions, lower Mini-Mental State Examination (MMSE) scores, and APOE-ε4 carriers compared to their respective reference groups. DISCUSSION Our study identified risk factors for cerebral amyloid angiopathy, informing potential prevention strategies. HIGHLIGHTS ADNC were significantly positively associated with the risk of CAA. The ADNC-CAA association was generally stronger among females, smokers, people with a history of stroke/memory complaints, APOE-ε4 carriers, and those without diabetes or heart conditions. Longitudinally, higher CAA risk was observed among males, older individuals, smokers, people with diabetes/heart conditions/lower MMSE scores, and APOE-ε4 carriers compared to their reference groups.
Collapse
Affiliation(s)
- Liwei Ma
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Yihan Wang
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew Liem Hieu Huynh
- Department of Aged CareAustin HealthHeidelbergVictoriaAustralia
- Department of MedicineAustin HealthThe University of MelbourneHeidelbergVictoriaAustralia
| | - Sanka Amadoru
- Department of Aged CareAustin HealthHeidelbergVictoriaAustralia
- Department of MedicineAustin HealthThe University of MelbourneHeidelbergVictoriaAustralia
| | - Scott Wrigley
- Department of Aged CareAustin HealthHeidelbergVictoriaAustralia
| | - Paul Yates
- Department of Aged CareAustin HealthHeidelbergVictoriaAustralia
- Department of MedicineAustin HealthThe University of MelbourneHeidelbergVictoriaAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
| | - Benjamin Goudey
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- The ARC Training Centre in Cognitive Computing for Medical TechnologiesThe University of MelbourneCarltonVictoriaAustralia
| | - Liang Jin
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Yijun Pan
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
12
|
Dohl J, Treadwell Z, Norris C, Head E. Calcineurin inhibition may prevent Alzheimer disease in people with Down syndrome. Alzheimers Dement 2025; 21:e70034. [PMID: 40042516 PMCID: PMC11881635 DOI: 10.1002/alz.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Virtually all people with Down syndrome will develop Alzheimer disease pathology during their lifetime. As Alzheimer disease is the third leading cause of death and a significant factor in end-of-life complications for adults with Down syndrome, identifying interventions is a medical necessity. Calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, has recently been investigated as a possible Alzheimer treatment. This review explores the histories behind Down syndrome and Alzheimer disease, and their intersecting pathologies. This is followed by the role that calcineurin and its U.S. Food and Drug Administration-approved pharmacological inhibitor, tacrolimus, may play in the prevention or treatment of Alzheimer disease. Finally, this review discusses the gap in the literature surrounding the role of calcineurin, its regulators, and calcineurin inhibitors in the context of Down syndrome and comorbid Alzheimer disease. Future studies investigating the role that calcineurin plays in this pathology will be essential in determining the viability of calcineurin inhibitors to treat Alzheimer disease in people with Down syndrome. HIGHLIGHTS: Calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, has become prominent as a possible therapeutic target to treat Alzheimer disease. People with Down syndrome develop Alzheimer pathology as they age, requiring novel therapeutics for treatment. People with Down syndrome may exhibit contraindications to calcineurin inhibition-based therapy, as they overexpress RCAN1 and DYRK1A, regulators of calcineurin. There is a significant gap in the literature involving the expression of calcineurin, RCAN1 and DYRK1A, in people with Down syndrome and Alzheimer disease, which must be addressed to determine the efficacy and safety of newly developed therapeutics.
Collapse
Affiliation(s)
- Jacob Dohl
- Department of Pathology & Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Zoe Treadwell
- Department of Pathology & Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Christopher Norris
- Department of Pharmacology & Nutrition Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Elizabeth Head
- Department of Pathology & Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
13
|
Sanchez N, Boskovic DS, Diamond CW, Lyons TW, Soriano S, Kirsch WM. Downregulation of Parahippocampal Copper Chaperone for Superoxide Dismutase in Alzheimer's Disease. Brain Sci 2025; 15:216. [PMID: 40149738 PMCID: PMC11940324 DOI: 10.3390/brainsci15030216] [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: 02/03/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Proper regulation of copper is essential for maintaining neuronal stability and is facilitated by several chaperone proteins, protecting cells from oxidative damage that would otherwise be caused by improperly regulated copper ions. Oxidative stress, resulting from such dysregulation, is hypothesized to play a significant role in the pathogenesis of Alzheimer's disease (AD). METHODS In this study, we evaluated the concentrations of the copper chaperones CCS, DCTN4, and ATOX1 in control and AD cases via Western blotting and ELISA, and quantified the copper concentrations in fractionated neurons using ICP-MS. RESULTS Our findings reveal a significant reduction in CCS levels in AD cases (p = 0.0085), with a progressive decline observed with advancing age. This decline was more pronounced in women, although the difference did not reach statistical significance (p = 0.0768). No significant differences were observed in copper concentrations within synaptosomal (p = 0.3869) or cytosolic fractions (p = 0.4461) between the AD and control cases. Additionally, comprehensive analyses of the effects of sex and age showed no significant impact on the levels of copper chaperones or copper distribution across cellular compartments. CONCLUSIONS These results suggest a strong association between reduced CCS levels and AD pathology, highlighting a potential role for CCS in the redistribution of copper ions within neurons. This redistribution may contribute to oxidative stress and neuronal dysfunction, offering new insights into the mechanisms underlying AD pathogenesis.
Collapse
Affiliation(s)
- Nicholas Sanchez
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (N.S.); (D.S.B.); (W.M.K.)
- Neurosurgery Center for Research, Training and Education, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Danilo S. Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (N.S.); (D.S.B.); (W.M.K.)
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Charles W. Diamond
- Department of Earth and Planetary Sciences, University of California, Riverside, CA 92521, USA; (C.W.D.); (T.W.L.)
| | - Timothy W. Lyons
- Department of Earth and Planetary Sciences, University of California, Riverside, CA 92521, USA; (C.W.D.); (T.W.L.)
| | - Salvador Soriano
- Laboratory of Neurodegenerative Diseases, Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Wolff M. Kirsch
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (N.S.); (D.S.B.); (W.M.K.)
- Neurosurgery Center for Research, Training and Education, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
14
|
Mojahedi A, Singh A, Chen O. A review of the use of oral anticoagulants in individuals with atrial fibrillation who had experienced intracranial hemorrhage in the past. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2025; 17:1-18. [PMID: 40151394 PMCID: PMC11936751 DOI: 10.62347/rzkc2209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/21/2024] [Indexed: 03/29/2025]
Abstract
Atrial fibrillation (AF) is the most prevalent arrhythmia, significantly increasing the risk of stroke and thromboembolism. Oral anticoagulants (OACs), including direct oral anticoagulants (DOACs) and vitamin K antagonists (VKAs), have been shown to reduce these risks effectively. However, the administration of OACs carries a notable risk of spontaneous intracranial hemorrhage (ICH), a severe complication associated with high morbidity and mortality. Patients with a history of ICH face a complex decision regarding the resumption of anticoagulation therapy, as the likelihood of recurrence is heightened in this population. Current literature reveals inconsistencies in research findings regarding the safety and efficacy of restarting OACs after ICH. A lack of definitive guidelines addressing this issue leaves clinicians uncertain about optimal management strategies. This systematic review aims to analyze existing observational studies and randomized controlled trials (RCTs) to evaluate the safety and effectiveness of resuming OACs in patients with AF who have experienced ICH. The review underscores the urgent need for high-quality research to inform clinical practices and develop comprehensive guidelines for managing anticoagulation therapy in this vulnerable group.
Collapse
Affiliation(s)
- Azad Mojahedi
- Department of Internal Medicine, Stony Brook University HospitalStony Brook, New York, The United States
| | - Abhijeet Singh
- Division of Cardiology, Stony Brook University HospitalStony Brook, New York, The United States
| | - On Chen
- Division of Cardiology, Stony Brook University HospitalStony Brook, New York, The United States
| |
Collapse
|
15
|
Kaloss AM, Browning JL, Li J, Pan Y, Watsen S, Sontheimer H, Theus MH, Olsen ML. Vascular amyloidβ load in the meningeal arterial network correlates with loss of cerebral blood flow and pial collateral vessel enlargement in the J20 murine model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.635937. [PMID: 40161825 PMCID: PMC11952299 DOI: 10.1101/2025.02.05.635937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Global reduction in cerebral blood flow (CBF) is an early pathology in Alzheimer's disease, preceding significant plaque accumulation and neurological decline. Chronic reduced CBF and subsequent reduction in tissue oxygenation and glucose may drive neurodegeneration, yet the underlying cause of globally reduced CBF remains unclear. METHODS Using premortem delivery of Methoxy-XO4 to label Aβ, and arterial vascular labeling, we assessed Aβ burden on the pial artery/arteriole network and cerebral blood flow in aged male and female WT and J20 AD mice. RESULTS The pial artery/arteriole vascular network selectively displayed extensive vascular Aβ burden. Pial collateral arteriole vessels, the by-pass system that reroutes blood flow during occlusion, displayed significant enlargement in J20 mice. Despite this, CBF was decreased by approximately 15% in 12-month J20 mice when compared to WT littermates. DISCUSSION Significant Aβ burden on the meningeal arterial network may contribute to the restriction of CBF. Redistribution of CBF through enlarged pial collateral vessels may serve as a compensatory mechanism to alter CBF during disease progression in cases of CAA.
Collapse
|
16
|
Liao YC, Hsu SL, Hsiao CT, Lee YC. Cerebral microbleeds in Taiwanese patients with neuronal intranuclear inclusion disease. Brain 2025; 148:e8-e11. [PMID: 39395184 DOI: 10.1093/brain/awae326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/14/2024] Open
Affiliation(s)
- Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei 11221, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University School of Medicine, Taipei 11221, Taiwan
| | - Shao-Lun Hsu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Department of Neurology, Fu Jen Catholic University Hospital, New Taipei City 24352, Taiwan
| | - Cheng-Tsung Hsiao
- Department of Neurology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei 11221, Taiwan
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei 11221, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University School of Medicine, Taipei 11221, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| |
Collapse
|
17
|
Koemans EA, van Etten ES. Cerebral amyloid angiopathy: one single entity? Curr Opin Neurol 2025; 38:29-34. [PMID: 39760721 DOI: 10.1097/wco.0000000000001330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
PURPOSE OF REVIEW Cerebral amyloid angiopathy (CAA) is a common brain disorder among the elderly and individuals with Alzheimer's disease, where accumulation of amyloid-ß can lead to intracerebral hemorrhage and dementia. This review discusses recent developments in understanding the pathophysiology and phenotypes of CAA. RECENT FINDINGS CAA has a long preclinical phase starting decades before symptoms emerge. Its pathophysiology follows consecutive stages of amyloid-ß deposition, decreased vascular reactivity, nonhemorrhagic changes, and ultimately hemorrhages. Although impaired perivascular clearance is the leading hypothesis underlying CAA, several lines of evidence suggest that glymphatic dysfunction also plays a significant role in the disease process. Despite its common pathway, the disease course is variable. Some patients develop more microbleeds, while others develop larger hemorrhages, suggesting a differentiation in vascular remodeling. Some patients with CAA develop a symptomatic immune response, and inflammation could be an important contributor to vascular damage in CAA in general. Furthermore, the prion-like transmission of amyloid-β has been identified as a cause of iatrogenic CAA occurring decades after neurosurgical procedures involving cadaveric dura mater. SUMMARY Emerging evidence of sporadic, hereditary, inflammatory, and iatrogenic CAA suggests a complex interplay between brain clearance, inflammation and vascular remodeling leading to a diverse clinical phenotype.
Collapse
Affiliation(s)
- Emma A Koemans
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
18
|
Morotti A, Pilotto A, Zanola D, Galli A, Caratozzolo S, Gasparotti R, Padovani A. Cerebral Amyloid Angiopathy in Alzheimer Disease: A Comparison Between Different Versions of the Boston Criteria. Neurology 2025; 104:e210248. [PMID: 39836667 DOI: 10.1212/wnl.0000000000210248] [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: 05/01/2024] [Accepted: 11/07/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVES Cerebral amyloid angiopathy (CAA) is the main driver of amyloid-related imaging abnormalities (ARIAs) in Alzheimer disease (AD). We compared different versions of the Boston criteria for CAA diagnosis in AD. METHODS This article presents a single-center analysis (outpatient neurodegenerative clinic) of patients with AD with mild cognitive impairment (MCI) or early dementia, meeting NIA-AA criteria and having biological amyloid confirmation (CSF or imaging). Two raters analyzed hemorrhagic (cerebral microbleeds, CMBs; cortical superficial siderosis, cSS) and nonhemorrhagic (severe centrum semiovale perivascular spaces, CSO-PVSs; multispot pattern white matter hyperintensities (WMHs)) markers following the original (V1.0), modified (V1.5), and latest (V2.0) Boston criteria. RESULTS We included 75 patients (mean age 71.6 ± 8.1 years, 53% female, mean disease duration 2.6 ± 2.0 years, 91% MCI). White matter CAA markers were more common than hemorrhagic markers: 41 (55%) had severe CSO-PVSs, 28 (37%) had multispot WHMs, 12 (16%) had 1 lobar CMB, 9 had ≥2 lobar CMBs (12%), 1 (1.3%) had focal cSS, and 5 (6.7) had disseminated cSS. The prevalence of possible and probable CAA was lowest with V1.0 (14.7% and 9.3%) than with V1.5 (13.3% and 13.3%) and V2.0 (42.7% and 26.7%) criteria. DISCUSSION More than 1 in 4 patients with AD had probable CAA according to the V2.0 Boston criteria. These findings might inform future trials.
Collapse
Affiliation(s)
- Andrea Morotti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy
- Neuroradiology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Italy; and
- Brain Health Center, University of Brescia, Italy
| | - Daniele Zanola
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Alice Galli
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | | | - Roberto Gasparotti
- Neuroradiology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy
- Neuroradiology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Italy; and
- Brain Health Center, University of Brescia, Italy
| |
Collapse
|
19
|
Huang X, Zeng X, Tang L, Liu X, Huang X, Liu X, Wang Z, Li N, Fan D, Yang Q. Subarachnoid hemorrhage and finger-like projection predict recurrence in patients with lobar intracerebral hemorrhage. J Neurol 2025; 272:166. [PMID: 39849200 PMCID: PMC11757861 DOI: 10.1007/s00415-025-12900-z] [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: 12/07/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND AND PURPOSE Lobar intracerebral hemorrhage (ICH) is associated with a high risk of recurrence, particularly in elderly patients, where cerebral amyloid angiopathy (CAA) is often the primary cause. Diagnostic markers of CAA-related ICH, including subarachnoid hemorrhage (SAH) and finger-like projection (FLP), have recently been developed. Here, we aimed to explore the associations between SAH, FLP and the risk of ICH recurrence in lobar ICH patients. METHODS We analyzed data from consecutive lobar ICH patients using the method of cohort study. We divided them into 4 groups on the basis of the presence or absence of SAH and FLP on CT imaging. The Cox regression model and competing risk model were used to analyze the associations of SAH and FLP with the risk of ICH recurrence at 1 year. RESULTS In total, 353 patients with lobar ICH (median age 74 [62, 81] years, 57.2% male) were included in our study. During follow-up, recurrence occurred in 34 patients (10.6%), and 90 patients (28.1%) died. The competing risk model revealed that patients in the SAH + FLP- (HR 2.88, 95% CI 1.12-7.44, p = 0.03) and SAH + FLP + (HR 8.38, 95% CI 3.40-20.66, p < 0.001) groups had higher risks of ICH recurrence within 1 year than did those in the SAH-FLP- group. CONCLUSION SAH is an important predictor of ICH recurrence, and this predictive ability is further enhanced when FLP is present. These findings suggest that SAH, especially with FLP, can be a valuable tool for assessing prognosis in lobar ICH patients.
Collapse
Affiliation(s)
- Xin Huang
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiangzhu Zeng
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Lu Tang
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiaolu Liu
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiao Huang
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Zhuoya Wang
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Nan Li
- The Clinical Epidemiology Research Center, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China.
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China.
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.
| | - Qiong Yang
- Department of Neurology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China.
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China.
| |
Collapse
|
20
|
Rech MA, Carpenter CR, Aggarwal NT, Hwang U. Anti-Amyloid Therapies for Alzheimer's Disease and Amyloid-Related Imaging Abnormalities: Implications for the Emergency Medicine Clinician. Ann Emerg Med 2025:S0196-0644(24)01254-X. [PMID: 39818674 DOI: 10.1016/j.annemergmed.2024.12.002] [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: 08/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease is the neurodegenerative disorder responsible for approximately 60% to 70% of all cases of dementia and is expected to affect 152 million by 2050. Recently, anti-amyloid therapies have been developed and approved by the Food and Drug Administration as disease-modifying treatments given as infusions every 2 to 5 weeks for Alzheimer's disease. Although this is an important milestone in mitigating Alzheimer's disease progression, it is critical for emergency medicine clinicians to understand what anti-amyloid therapies are and how they work to recognize, treat, and mitigate their adverse effects. Anti-amyloid therapies may be underrecognized contributors to emergency department visits because they carry the risk of adverse effects, namely amyloid-related imaging abnormalities. Amyloid-related imaging abnormalities are observed as abnormalities on magnetic resonance imaging as computed tomography is not sensitive enough to detect the microvasculature abnormalities causing vasogenic edema (amyloid-related imaging abnormalities-E) microhemorrhages and hemosiderin deposits (amyloid-related imaging abnormalities-H). Patients presenting with amyloid-related imaging abnormalities may have nonspecific neurologic symptoms, including headache, lethargy, confusion, and seizures. Anti-amyloid therapies may increase risk of hemorrhagic conversion of ischemic stroke patients receiving thrombolytics and complicate the initiation of anticoagulation. Given the novelty of anti-amyloid therapies and limited real-world data pertaining to amyloid-related imaging abnormalities, it is important for emergency medicine clinicians to be aware of these agents.
Collapse
Affiliation(s)
- Megan A Rech
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare Edward Hines, Jr. VA Hospital, IL.
| | | | - Neelum T Aggarwal
- Departments of Neurological Sciences, Rush Alzheimer's Disease Center, Chicago, IL
| | - Ula Hwang
- Departments of Emergency Medicine & Population Health, New York University Grossman School of Medicine, New York, NY; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY
| |
Collapse
|
21
|
Vestergaard MB, Bakhtiari A, Osler M, Mortensen EL, Lindberg U, Law I, Lauritzen M, Benedek K, Larsson HBW. The cerebral blood flow response to neuroactivation is reduced in cognitively normal men with β-amyloid accumulation. Alzheimers Res Ther 2025; 17:4. [PMID: 39754275 PMCID: PMC11699738 DOI: 10.1186/s13195-024-01652-z] [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: 03/24/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Accumulation of β-amyloid (Aβ) in the brain is a hallmark of Alzheimer's Disease (AD). Cerebral deposition of Aβ initiates deteriorating pathways which eventually can lead to AD. However, the exact mechanisms are not known. A possible pathway could be that Aβ affects the cerebral vessels, causing inadequate cerebrovascular function. In the present study, we examined if Aβ accumulation is associated with a reduced cerebral blood flow response (CBF) to neuronal activation by visual stimulation (ΔCBFVis.Act.) in cognitively normal subjects from the Metropolit Danish Male Birth Cohort. METHODS 64 subjects participated in the present study. ΔCBFVis.Act. was measured using arterial spin labelling (ASL) combined with blood-oxygen-level-dependent (BOLD) MRI. Neuronal activation was obtained by visual stimulation by a flickering checkerboard presented on a screen in the MRI-scanner. Brain Aβ accumulation and cerebral glucose metabolism were assessed by PET imaging using the radiotracers [11C]Pittsburgh Compound-B (PiB) and [18F]Fluorodeoxyglucose (FDG), respectively. Cortical thickness was measured from structural MRI. RESULTS ΔCBFVis.Act. correlated negatively ( β = -32.1 [95% confidence interval (CI): -60.2; -4.1], r = -0.30, p = 0.025) with PiB standardized uptake value ratio (SUVr) in the brain regions activated by visual stimulation. ΔCBFVis.Act. did not correlate with FDG SUVr ( β = 1.9 [CI: -23.8; 27.6], r = 0.02, p = 0.88) or cortical thickness ( β = 10.3 [CI: -8.4; 29.0], r = 0.15, p = 0.27) in the activated brain regions. Resting CBF did not correlate with PiB SUVr neither in the regions activated by visual stimulation ( β = -17.8 [CI:-71.9; 36.2], r =- 0.09, p = 0.51) nor in the remaining cortex ( β = 5.2 [CI:-3.9; 14.2], r = 0.15, p = 0.26). CONCLUSION We found a correlation between high PiB SUVr and reduced CBF response to neuronal activation, indicating a link between Aβ accumulation and impaired cerebrovascular function. The impairment was not associated with cortical thinning or hypometabolism, suggesting that Aβ accumulation affecting brain vessel function could be a very early pathology leading to neurodegenerative disease.
Collapse
Affiliation(s)
- Mark Bitsch Vestergaard
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark.
| | - Aftab Bakhtiari
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Merete Osler
- Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | - Ulrich Lindberg
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Martin Lauritzen
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krisztina Benedek
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Henrik Bo Wiberg Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Reynolds B, McLaine R. An older man with progressive short-term memory loss and confusion. JAAPA 2025; 38:e5-e8. [PMID: 39699323 DOI: 10.1097/01.jaa.0000000000000151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
ABSTRACT A 72-year-old man with progressive memory loss and confusion presented to a neurology clinic for evaluation. He initially had difficulty remembering names and misplaced objects; however, his memory deficits had progressed, and more recently he had numerous car accidents and difficulty managing his own medications and finances. Cognitive testing revealed significant memory deficits reflecting moderate-stage dementia, and his brain MRI showed several cortical microbleeds and an area of siderosis consistent with the diagnosis of cerebral amyloid angiopathy (CAA). This case report provides an overview of a classic case of CAA and its potential treatment options.
Collapse
Affiliation(s)
- Beverly Reynolds
- Beverly Reynolds practices at the VA Boston Healthcare System in West Roxbury, Mass. Rosalind McLaine is director of clinical operations at Alzheon, Inc., in Framingham, Mass. The authors have disclosed no potential conflicts of interest, financial or otherwise
| | | |
Collapse
|
23
|
Lin S, Zhu G, Xie J, Wen X, Deng L, Li S, Liu G, Wang F, Chen S, Xiao Z. Study on the Therapeutic Effects of Bisdemethoxycurcumin on a Cerebral Amyloid Angiopathy Mouse Model Established via Chronic Treatment With Five Vascular Risk Factors. Brain Behav 2025; 15:e70245. [PMID: 39829173 PMCID: PMC11743980 DOI: 10.1002/brb3.70245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/12/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Cerebral amyloid angiopathy (CAA) is recognized as a major contributor to progressive cognitive decline and cerebral hemorrhages in the elderly population. Currently, there is a global shortage of safe and effective treatments for this condition. Bisdemethoxycurcumin (BDMC) has been demonstrated to exhibit pharmacological effects with anti-Aβ toxicity properties. Thus, the present study mainly focused on the potential therapeutic effects of BDMC on CAA. METHOD The 30 male C57BL/6 mice were subjected to chronic treatment with five vascular risk factors (lipopolysaccharide, social stress, streptozotocin, high-cholesterol diet, and copper-containing drinking water) for 35 weeks to establish a CAA mouse model. Of these, 15 CAA mice received oral administration of BDMC (50 mg/kg) for two consecutive weeks as an intervention, while the remaining 15 CAA mice received an equal volume of physiological saline by gavage. The study observed the levels of Aβ40 and proinflammatory factors in brain tissue and plasma, Aβ deposition in cerebral blood vessels, microbleeds in brain tissue, expression of proteins related to the cGAS/STING signaling pathway in brain tissue, as well as the contents of p-RIPK-1, p-RIPK-3, p-MLKL, neuronal morphology, and learning and memory abilities in mice. RESULT The therapeutic administration of BDMC demonstrates a pronounced efficacy in alleviating Aβ burden and cerebral microbleeding in CAA mice, concurrently enhancing learning and memory capabilities. Interestingly, BDMC may inhibits neuroinflammatory responses by reducing the expression of cGAS/STING signaling pathway proteins and suppresses necroptosis. CONCLUSION Our research findings demonstrate that BDMC exerts therapeutic effects in a mouse model of CAA established through chronic treatment involving five vascular risk factors.
Collapse
Affiliation(s)
- Shudong Lin
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Guanghua Zhu
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Juan Xie
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanPeople's Republic of China
| | - Xuanwei Wen
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Limin Deng
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Sijing Li
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Guozhi Liu
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Feiyan Wang
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Shuangxi Chen
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| | - Zijian Xiao
- Department of Neurology, The First Affiliated Hospital, Multi‐Omics Research Center for Brain Disorders, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
- Clinical Research Center for Immune‐Related Encephalopathy In Hunan Province, Hengyang Medical SchoolUniversity Of South ChinaHengyangHunanChina
| |
Collapse
|
24
|
Tanaka F, Maeda M, Kishi S, Kogue R, Umino M, Ishikawa H, Ii Y, Shindo A, Sakuma H. Updated imaging markers in cerebral amyloid angiopathy: What radiologists need to know. Jpn J Radiol 2024:10.1007/s11604-024-01720-2. [PMID: 39730931 DOI: 10.1007/s11604-024-01720-2] [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: 09/06/2024] [Accepted: 12/06/2024] [Indexed: 12/29/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is an age-related small vessel disease pathologically characterized by the progressive accumulation of amyloid-beta (Aβ) peptide in cerebrovascular walls, affecting both cortical and leptomeningeal vessels. Amyloid deposition results in fragile vessels, which may lead to lobar intracerebral hemorrhage (ICH) and cognitive impairment. To evaluate the probability and severity of CAA, the imaging markers depicted on CT and MRI techniques are crucial, as brain pathological examination is highly invasive. Although the Boston criteria have established diagnostic value and have been updated to version 2.0, due to an aging population, the patients with CAA should also be assessed for their risk of future ICH or cognitive impairment. Furthermore, an increased awareness of CAA is essential when introducing anticoagulants for infarct in elderly patients or anti-amyloid antibodies for Alzheimer's disease, as these may worsen CAA-related hemorrhagic lesions. However, the radiological literature on CAA has not been comprehensively updated. Here, we review the imaging markers of CAA and clinical significance. We also discuss the clinical and imaging characteristics of CAA-related inflammation, amyloid-related imaging abnormalities, and iatrogenic-CAA.
Collapse
Affiliation(s)
- Fumine Tanaka
- Department of Radiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Masayuki Maeda
- Department of Neuroradiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Seiya Kishi
- Department of Radiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Ryota Kogue
- Department of Radiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Maki Umino
- Department of Radiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yuichiro Ii
- Department of Neuroimaging and Pathophysiology, Mie University School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hajime Sakuma
- Department of Radiology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
25
|
Meng Y, Xiao Y, Sun R, Li L, Dong Y. Cerebral amyloid angiopathy-related inflammation (CAA-ri): A case report. Medicine (Baltimore) 2024; 103:e41010. [PMID: 39705440 DOI: 10.1097/md.0000000000041010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
RATIONALE Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a treatable condition characterized by an acute or subacute onset, with its primary pathological hallmark being the deposition of amyloid, predominantly β-amyloid (Aβ), within intracranial microvessels. Despite its potential for treatment, CAA-ri is a rare disorder that is frequently underrecognized by clinicians in practice. This article provides a comprehensive overview of the clinical manifestations and therapeutic approaches associated with CAA-ri, aiming to enhance awareness among healthcare professionals. PATIENT CONCERNS A 67-year-old male patient who suffered from a sudden decline in cognitive functioning, intermittent headache, and dysphoria underwent brain magnetic resonance imaging, susceptibility weighted imaging, and cerebrospinal fluid analysis and was considered probable CAA-ri. DIAGNOSIS INTERVENTIONS During the course of disease development, the patient suffered from a sudden decline in cognitive functioning, mainly in the form of unresponsiveness, decreased comprehension, and increased repetitive language, accompanied by intermittent headaches and dysphoria. Brain magnetic resonance imaging showed numerous white matter in both hemispheres. Susceptibility weighted imaging showed multiple spots of hypointensity in the bilateral cerebral and cerebellar hemispheres, a hypointensity signal in the left occipital lobe, and extensive zones of hypointensity in bilateral sulci. Cerebrospinal fluid analysis was abnormal with elevated levels of protein and low levels of P-tau, Aβ42, and Aβ1-42/Aβ1-40. The use of glucocorticoids greatly reduced his symptoms. This lends credence to the probable CAA-ri diagnosis. The symptoms can be successfully alleviated by administering methylprednisolone sodium succinate. OUTCOMES During the patient's hospitalization, immunosuppressive therapy, primarily consisting of methylprednisolone sodium succinate and methylprednisolone, was administered, resulting in a significant improvement in symptoms. Post-discharge, the patient was monitored regularly, revealing a gradual enhancement in cognitive function without recurrence. Consequently, immunosuppressive therapy was discontinued 1 year following the patient's discharge. LESSONS CAA-ri is a rare clinical condition, and timely diagnosis and early treatment are very critical for patient prognosis.
Collapse
Affiliation(s)
- Yao Meng
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Department of Neurology, Graduate School of Hebei North University, Zhangjiakou, China
| | - Yining Xiao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| | - Ruohan Sun
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Graduate School of Hebei Medical University, Shijiazhuang, China
| | - Lingyu Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Graduate School of North China University of Science and Technology, Tangshan, China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- Department of Neurology, Graduate School of Hebei North University, Zhangjiakou, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| |
Collapse
|
26
|
Koundal S, Chen X, Gursky Z, Lee H, Xu K, Liang F, Xie Z, Xu F, Lin HM, Van Nostrand WE, Gu X, Elkin R, Tannenbaum A, Benveniste H. Divergent brain solute clearance in rat models of cerebral amyloid angiopathy and Alzheimer's disease. iScience 2024; 27:111463. [PMID: 39720539 PMCID: PMC11667077 DOI: 10.1016/j.isci.2024.111463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/05/2024] [Accepted: 11/20/2024] [Indexed: 12/26/2024] Open
Abstract
Brain waste clearance from the interstitial fluid environment is challenging to measure, which has contributed to controversy regarding the significance of glymphatic transport impairment for neurodegenerative processes. Dynamic contrast enhanced MRI (DCE-MRI) with cerebrospinal fluid administration of Gd-tagged tracers is often used to assess glymphatic system function. We previously quantified glymphatic transport from DCE-MRI data utilizing regularized optimal mass transport (rOMT) analysis, however, information specific to glymphatic clearance was not directly derived. To fill this knowledge gap, we here implemented unbalanced rOMT analysis which allows for assessment of both influx and clearance. Dynamic influx/clearance brain maps were derived from rTg-DI rats with cerebral amyloid angiopathy (CAA) and TgSD-AD rats with Alzheimer's disease (AD). The rTg-DI rats with severe CAA disease exhibited abnormal influx/clearance kinetics, while TgSD-AD rats with a moderate Aβ plaque load exhibited normal transport suggesting that different Aβ lesions and their overall burden differentially impact glymphatic system function.
Collapse
Affiliation(s)
- Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xinan Chen
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, NY 10065, USA
| | - Zachary Gursky
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kaiming Xu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Feng Xu
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02906, USA
| | - Hung-Mo Lin
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - William E. Van Nostrand
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02906, USA
| | - Xianfeng Gu
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY 11794, USA
- Departments of Computer Science, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rena Elkin
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, NY 10065, USA
| | - Allen Tannenbaum
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY 11794, USA
- Departments of Computer Science, Stony Brook University, Stony Brook, NY 11794, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale School of Medicine New Haven, New Haven, CT 06510, USA
| |
Collapse
|
27
|
Thiankhaw K, Best J, Srivastava S, Prachee I, Agarwal S, Tan S, Calvert PA, Chughtai A, Ang R, Segal OR, Werring DJ. Left atrial appendage occlusion in patients with atrial fibrillation and intracerebral haemorrhage associated with cerebral amyloid angiopathy: a multicentre observational study and pooled analysis of published studies. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-334718. [PMID: 39694822 DOI: 10.1136/jnnp-2024-334718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a common cause of intracerebral haemorrhage (ICH) with a high recurrence risk. Left atrial appendage occlusion (LAAO) is a method for ischaemic stroke prevention in patients with atrial fibrillation (AF), potentially reducing the risk of intracranial bleeding in CAA-associated ICH. We aimed to determine the outcomes of patients with AF with CAA-associated ICH undergoing LAAO. METHODS We conducted a multicentre study of patients with CAA-associated ICH who underwent LAAO for stroke prevention. We pooled our findings with data from a systematic review of relevant published studies of LAAO for AF in ICH survivors reporting CAA diagnosis. RESULTS We included data from two published studies (n=65) with CAA-specific data and our cohort study (n=37), providing a total of 102 participants (mean age 76.2±8.0 years, 74.6% male) with CAA-related symptomatic ICH and AF treated with LAAO. The median follow-up period was 9.4 months (IQR 4.2-20.6). Postprocedural antithrombotic regimens varied between single (73.0%) or dual antiplatelet therapy (16.2%), or direct oral anticoagulant (DOAC) (10.8%), with a median duration of 42 days (IQR 35-74). Postprocedural complications were uncommon, but included transient arrhythmias (2.1%) and non-life-threatening tamponade (2.1%). Pooled incidence rates of ischaemic stroke and ICH during follow-up were 5.16 (95% CI 1.36 to 17.48) and 2.73 (95% CI 0.41 to 13.94) per 100 patient years, respectively. CONCLUSIONS LAAO followed by short-term antithrombotic therapy might be a safe and effective ischaemic stroke preventive strategy in people with CAA-associated ICH and AF. However, randomised controlled trials are needed to determine how LAAO compares with long-term DOAC in this population. PROSPERO REGISTRATION NUMBER CRD42023415354.
Collapse
Affiliation(s)
- Kitti Thiankhaw
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
- Department of Internal Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jonathan Best
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Sonal Srivastava
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Ishika Prachee
- Department of Cardiac Electrophysiology, Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Smriti Agarwal
- Department of Stroke Medicine, Clinical Neurosciences, Addenbrooke's Hospital, Cambridge, UK
| | - Serena Tan
- Department of Stroke Medicine, Clinical Neurosciences, Addenbrooke's Hospital, Cambridge, UK
| | - Patrick A Calvert
- Department of Cardiology, Royal Papworth Hospital, Cambridge, UK
- University of Cambridge, Cambridge, UK
| | - Asim Chughtai
- Department of Cardiology, Royal Papworth Hospital, Cambridge, UK
| | - Richard Ang
- Department of Cardiac Electrophysiology, Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Oliver R Segal
- Department of Cardiac Electrophysiology, Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - David J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
28
|
Seiffge DJ, Fandler-Höfler S, Du Y, Goeldlin MB, Jolink WMT, Klijn CJM, Werring DJ. Intracerebral haemorrhage - mechanisms, diagnosis and prospects for treatment and prevention. Nat Rev Neurol 2024; 20:708-723. [PMID: 39548285 DOI: 10.1038/s41582-024-01035-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/17/2024]
Abstract
Intracerebral haemorrhage (ICH) is a devastating condition associated with high mortality and substantial residual disability among survivors. Effective treatments for the acute stages of ICH are limited. However, promising findings from randomized trials of therapeutic strategies, including acute care bundles that target anticoagulation therapies, blood pressure control and other physiological parameters, and trials of minimally invasive neurosurgical procedures have led to renewed optimism that patient outcomes can be improved. Currently ongoing areas of research for acute treatment include anti-inflammatory and haemostatic treatments. The implementation of effective secondary prevention strategies requires an understanding of the aetiology of ICH, which involves vascular and brain parenchymal imaging; the use of neuroimaging markers of cerebral small vessel disease improves classification with prognostic relevance. Other data underline the importance of preventing not only recurrent ICH but also ischaemic stroke and cardiovascular events in survivors of ICH. Ongoing and planned randomized controlled trials will assess the efficacy of prevention strategies, including antiplatelet agents, oral anticoagulants or left atrial appendage occlusion (in patients with concomitant atrial fibrillation), and optimal management of long-term blood pressure and statin use. Together, these advances herald a new era of improved understanding and effective interventions to reduce the burden of ICH.
Collapse
Affiliation(s)
- David J Seiffge
- Department of Neurology, Inselspital University Hospital Bern and University of Bern, Bern, Switzerland
| | - Simon Fandler-Höfler
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Yang Du
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Martina B Goeldlin
- Department of Neurology, Inselspital University Hospital Bern and University of Bern, Bern, Switzerland
| | | | - Catharina J M Klijn
- Department of Neurology, Donders Institute of Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - David J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
29
|
Silbert LC. Vascular Cognitive Impairment. Continuum (Minneap Minn) 2024; 30:1699-1725. [PMID: 39620840 DOI: 10.1212/con.0000000000001508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Vascular cognitive impairment is a major contributor to age-associated cognitive decline, both independently and as a contributor to mixed dementia syndromes. This article reviews the current understanding of how vascular dysfunction contributes to cognitive impairment and dementia risk in older individuals and includes updated diagnostic criteria and treatment recommendations. LATEST DEVELOPMENTS Clinical and research criteria have been evolving to more accurately determine the full prevalence of vascular cognitive impairment. The Boston Criteria version 2.0 for cerebral amyloid angiopathy now includes multiple punctate MRI T2 white matter hyperintensities and MR-visible perivascular spaces in addition to previously described T2* hemorrhagic signatures. MR-visible perivascular spaces are associated with both vascular cognitive impairment and Alzheimer disease, potentially linking cerebrovascular dysfunction to neurodegenerative disorders through its role in brain waste clearance. The American Heart Association's goal for cardiovascular health promotion, "Life's Essential 8," has been updated to include sleep health and acknowledges psychological well-being and social determinants of health as fundamental components necessary to achieve optimal cardiovascular health for all adults. ESSENTIAL POINTS Vascular cognitive impairment is a common and often underrecognized contributor to cognitive impairment in older individuals, with heterogeneous etiologies requiring individualized treatment strategies. Effective cerebrovascular disease risk factor modification starting in midlife is critical to reducing the risk of Alzheimer disease and related dementias, with the goal of preventing vascular brain injury and maintaining cognitive reserve in the presence of nonvascular age-related brain pathologies.
Collapse
|
30
|
van den Brink H, Voigt S, Kozberg M, van Etten ES. The role of neuroinflammation in cerebral amyloid angiopathy. EBioMedicine 2024; 110:105466. [PMID: 39608058 DOI: 10.1016/j.ebiom.2024.105466] [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: 05/31/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a cerebrovascular disease characterized by vascular amyloid-β (Aβ) deposition. CAA is often seen in the brains of elderly individuals and in a majority of patients with Alzheimer's disease. The molecular pathways triggered by vascular Aβ, causing vessel wall breakdown and ultimately leading to intracerebral haemorrhage and cognitive decline, remain poorly understood. The occurrence of CAA-related inflammation (CAA-ri) and Amyloid-Related Imaging Abnormalities (ARIA) have sparked interest for a role of neuroinflammation in CAA pathogenesis. This review discusses prior studies of neuroinflammation in CAA and outlines potential future research directions targeting candidates such as matrix metalloproteinases, complement activation, microglial activation, reactive astrocytes and parenchymal border macrophages. Understanding the role of neuroinflammation in CAA pathogenesis could help identify new therapeutic strategies.
Collapse
Affiliation(s)
- Hilde van den Brink
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sabine Voigt
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mariel Kozberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellis S van Etten
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
31
|
Widaja E, Pawitan JA. Integrating epigenetic modification and stem cell therapy strategies: A novel approach for advancing Alzheimer's disease treatment - A literature review. NARRA J 2024; 4:e935. [PMID: 39816083 PMCID: PMC11731673 DOI: 10.52225/narra.v4i3.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia and represents an increasing global burden, particularly in countries like Indonesia, where the population has begun to age significantly. Current medications, including cholinesterase inhibitors and NMDA receptor antagonists, have modest effects on clinical symptoms in the early to middle stages, but there is no curative treatment available so far despite progress. Activating or repressing epigenetic modifications, including DNA methylation, histone modification and microRNA regulation, appears to play an important role in AD development. These alterations further enact transcriptional changes relevant to the signature AD pathologies of amyloid-β deposition, tau protein malfunctioning, neuroinflammation, and neuronal death. Here, we discuss the feasibility of targeting these epigenetic alterations as a new treatment strategy due to the reversibility of epigenetics and their ability to correct faulty gene expression. We also review the combined promise of stem cell therapies and epigenetic modulation in neurodegeneration, inflammation and cognitive decline. This combined approach may provide a multifaceted strategy to slow disease progression, replace lost neurons, and restore neural function. Despite challenges, including ethical, financial, and methodological barriers, ongoing research in epigenetic modulation and stem cell therapy holds promise for pioneering therapies in AD.
Collapse
Affiliation(s)
- Edhijanto Widaja
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Regenerative Medicine and Research Institute of Mandaya Hospital Group, Tangerang, Indonesia
| | - Jeanne A. Pawitan
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Dr. Cipto Mangunkusumo Central Hospital, Jakarta, Indonesia
| |
Collapse
|
32
|
Jäkel L, Claassen KKWJ, De Kort AM, Jolink WMT, Vermeiren Y, Schreuder FHBM, Küsters B, Klijn CJM, Kuiperij HB, Verbeek MM. Decreased microvascular claudin-5 levels in cerebral amyloid angiopathy associated with intracerebral haemorrhage. Brain Pathol 2024; 34:e13270. [PMID: 38763889 PMCID: PMC11483184 DOI: 10.1111/bpa.13270] [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: 01/30/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024] Open
Abstract
Decreased microvascular levels of claudin-5 in the occipital and temporal lobe of patients with cerebral amyloid angiopathy are associated with intracerebral haemorrhage.
Collapse
Affiliation(s)
- Lieke Jäkel
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Kiki K. W. J. Claassen
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Anna M. De Kort
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Yannick Vermeiren
- Division of Human Nutrition and Health, Chair Group Nutritional BiologyWageningen University & Research (WUR)WageningenThe Netherlands
- Faculty of Medicine & Health Sciences, Translational NeurosciencesInstitute Born‐Bunge, University of AntwerpAntwerpBelgium
| | - Floris H. B. M. Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Benno Küsters
- Department of PathologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Catharina J. M. Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - H. Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Marcel M. Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
33
|
Milos T, Vuic B, Balic N, Farkas V, Nedic Erjavec G, Svob Strac D, Nikolac Perkovic M, Pivac N. Cerebrospinal fluid in the differential diagnosis of Alzheimer's disease: an update of the literature. Expert Rev Neurother 2024; 24:1063-1079. [PMID: 39233323 DOI: 10.1080/14737175.2024.2400683] [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: 04/23/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The importance of cerebrospinal fluid (CSF) biomarkers in Alzheimer's disease (AD) diagnosis is rapidly increasing, and there is a growing interest in the use of CSF biomarkers in monitoring the response to therapy, especially in the light of newly available approaches to the therapy of neurodegenerative diseases. AREAS COVERED In this review we discuss the most relevant measures of neurodegeneration that are being used to distinguish patients with AD from healthy controls and individuals with mild cognitive impairment, in order to provide an overview of the latest information available in the scientific literature. We focus on markers related to amyloid processing, markers associated with neurofibrillary tangles, neuroinflammation, neuroaxonal injury and degeneration, synaptic loss and dysfunction, and markers of α-synuclein pathology. EXPERT OPINION In addition to neuropsychological evaluation, core CSF biomarkers (Aβ42, t-tau, and p-tau181) have been recommended for improvement of timely, accurate and differential diagnosis of AD, as well as to assess the risk and rate of disease progression. In addition to the core CSF biomarkers, various other markers related to synaptic dysfunction, neuroinflammation, and glial activation (neurogranin, SNAP-25, Nfl, YKL-40, TREM2) are now investigated and have yet to be validated for future potential clinical use in AD diagnosis.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Nikola Balic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Vladimir Farkas
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | | | | | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
- University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia
| |
Collapse
|
34
|
Charidimou A, Boulouis G. Core CSF Biomarker Profile in Cerebral Amyloid Angiopathy: Updated Meta-Analysis. Neurology 2024; 103:e209795. [PMID: 39270153 DOI: 10.1212/wnl.0000000000209795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES There is a clear need to characterize and validate molecular biomarkers of cerebral amyloid angiopathy (CAA), in an effort to improve diagnostics, especially in the context of patients with Alzheimer disease (AD) receiving immunotherapies (for whom underlying CAA is the driver of amyloid-related imaging abnormalities). We performed an updated meta-analysis of 5 core CSF biomarkers (Aβ42, Aβ40, Aβ438, total tau [T-tau], and phosphorylated tau [P-tau]) to assess which of these are most altered in sporadic CAA. METHODS We systematically searched PubMed for eligible studies reporting data on CSF biomarkers reflecting APP metabolism (Aβ42, Aβ40, Aβ38), neurodegeneration (T-tau), and tangle pathology (P-tau), in symptomatic sporadic CAA cohorts (based on the Boston criteria) vs control groups and/or vs patients with AD. Biomarker performance was assessed in random-effects meta-analysis based on ratio of mean (RoM) biomarker concentrations in (1) patients with CAA to controls and (2) CAA to patients with AD. RoM >1 indicates higher biomarker concentration in CAA vs comparison population, and RoM <1 indicates higher concentration in comparison groups. RESULTS 8 studies met inclusion criteria: a total of 11 CAA cohorts (n = 289), 9 control cohorts (n = 310), and 8 AD cohorts (n = 339). Overall included studies were of medium quality based on our assessment tools. CAA to controls had lower mean level of all amyloid markers with CSF Aβ42, Aβ40, and Aβ38 RoMs of 0.46 (95% CI 0.38-0.55, p < 0.0001), 0.70 (95% CI 0.63-0.78, p < 0.0001), and 0.71 (95% CI 0.56-0.89, p = 0.003), respectively. CSF T-tau and P-tau RoMs of patients with CAA to controls were both greater than 1: 1.56 (95% CI 1.32-1.84, p < 0.0001) and 1.31 (95% CI 1.13-1.51, p < 0.0001), respectively. Differentiation between CAA and AD was strong for CSF Aβ40 (RoM 0.76, 95% CI 0.69-0.83, p < 0.0001) and Aβ38 (RoM 0.55, 95% CI 0.38-0.81, p < 0.0001), but not Aβ42 (RoM 1.00; 95% CI 0.81-1.23, p = 0.970). For T-tau and P-tau, average CSF ratios in patients with CAA vs AD were 0.64 (95% CI 0.58-0.71, p < 0.0001) and 0.64 (95% CI 0.58-0.71, p < 0.0001), respectively. DISCUSSION Specific CSF patterns of Aβ42, Aβ40, Aβ38, T-tau, and P-tau might serve as molecular biomarkers of CAA, in research and clinical settings, offering the potential to improve the clinical diagnostic approach pathway in specific scenarios.
Collapse
Affiliation(s)
- Andreas Charidimou
- From the Department of Neurology (A.C.), Boston University Medical Center, Boston University School of Medicine, MA; and Diagnostic and Interventional Neuroradiology (G.B.), University Hospital, Tours, France
| | - Gregoire Boulouis
- From the Department of Neurology (A.C.), Boston University Medical Center, Boston University School of Medicine, MA; and Diagnostic and Interventional Neuroradiology (G.B.), University Hospital, Tours, France
| |
Collapse
|
35
|
Schrader JM, Majchrzak M, Xu F, Lee H, Agostinucci K, Davis J, Benveniste H, Van Nostrand WE. Cerebral Proteomic Changes in the rTg-D Rat Model of Cerebral Amyloid Angiopathy Type-2 With Cortical Microhemorrhages and Cognitive Impairments. Neurosci Insights 2024; 19:26331055241288172. [PMID: 39386146 PMCID: PMC11462563 DOI: 10.1177/26331055241288172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a common disorder of the elderly, a prominent comorbidity of Alzheimer's disease, and causes vascular cognitive impairment and dementia. Previously, we generated a novel transgenic rat model (rTg-D) that produces human familial CAA Dutch E22Q mutant amyloid β-protein (Aβ) in brain and develops arteriolar CAA type-2. Here, we show that deposition of fibrillar Aβ promotes arteriolar smooth muscle cell loss and cerebral microhemorrhages that can be detected by magnetic resonance imaging and confirmed by histopathology. Aged rTg-D rats also present with cognitive deficits. Cerebral proteomic analyses revealed 241 proteins that were significantly elevated with an increase of >50% in rTg-D rats presenting with CAA compared to wild-type rats. Fewer proteins were significantly decreased in rTg-D rats. Of note, high temperature requirement peptidase A (HTRA1), a proteinase linked to transforming growth factor beta 1 (TGF-β1) signaling, was elevated and found to accumulate in cerebral vessels harboring amyloid deposits. Pathway analysis indicated elevation of the TGF-β1 pathway and increased TGF-β1 levels were detected in rTg-D rats. In conclusion, the present findings provide new molecular insights into the pathogenesis of CAA and suggest a role for interactions between HTRA1 and TGF-β1 in the disease process.
Collapse
Affiliation(s)
- Joseph M Schrader
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Mark Majchrzak
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Kevin Agostinucci
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Judianne Davis
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - William E Van Nostrand
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
36
|
Weidauer S, Tafreshi M, Förch C, Hattingen E, Arendt CT, Friedauer L. Clinical and neuroimaging precursors in cerebral amyloid angiopathy: impact of the Boston criteria version 2.0. Eur J Neurol 2024; 31:e16425. [PMID: 39105407 DOI: 10.1111/ene.16425] [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: 04/02/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND AND PURPOSE Although the Boston criteria version 2.0 facilitates the sensitivity of cerebral amyloid angiopathy (CAA) diagnosis, there are only limited data about precursor symptoms. This study aimed to determine the impact of neurological and imaging features in relation to the time of CAA diagnosis. METHODS Patients diagnosed with probable CAA according to the Boston criteria version 1.5, treated between 2010 and 2020 in our neurocentre, were identified through a keyword search in our medical database. Neuroimaging was assessed using Boston criteria versions 1.5 and 2.0. Medical records with primary focus on the clinical course and the occurrence of transient focal neurological episodes were prospectively evaluated. RESULTS Thirty-eight out of 81 patients (46.9%) exhibited transient focal neurological episodes, most often sensory (13.2%) or aphasic disorders (13.2%), or permanent deficits at a mean time interval of 31.1 months (SD ±26.3; range 1-108 months) before diagnosis of probable CAA (Boston criteria version 1.5). If using Boston criteria version 2.0, all patients receiving magnetic resonance imaging (MRI) met the criteria for probable CAA, and diagnosis could have been made on average 44 months earlier. Four patients were younger than 50 years, three of them with supporting pathology. Cognitive deficits were most common (34.6%) at the time of diagnosis. CONCLUSIONS Non-haemorrhagic MRI markers enhance the sensitivity of diagnosing probable CAA; however, further prospective studies are proposed to establish a minimum age for inclusion. As the neurological overture of CAA may occur several years before clinical diagnosis, early clarification by MRI including haemosensitive sequences are suggested.
Collapse
Affiliation(s)
- Stefan Weidauer
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mona Tafreshi
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christian Förch
- Department of Neurology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christophe T Arendt
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Lucie Friedauer
- Department of Neurology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
37
|
Castellani RJ, Jamshidi P. Cerebral amyloid-β-related angiitis and iatrogenic cerebral amyloid angiopathy-related vasculitis: Implications for amyloid-related imaging abnormalities. J Neuropathol Exp Neurol 2024; 83:890-892. [PMID: 38970388 DOI: 10.1093/jnen/nlae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024] Open
Affiliation(s)
- Rudy J Castellani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Pouya Jamshidi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
38
|
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] [MESH Headings] [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
|
39
|
Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. GeroScience 2024; 46:5103-5132. [PMID: 38639833 PMCID: PMC11336042 DOI: 10.1007/s11357-024-01139-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) are a critical but frequently underestimated aspect of cerebral small vessel disease (CSVD), bearing substantial clinical consequences. Detectable through sensitive neuroimaging techniques, CMHs reveal an extensive pathological landscape. They are prevalent in the aging population, with multiple CMHs often being observed in a given individual. CMHs are closely associated with accelerated cognitive decline and are increasingly recognized as key contributors to the pathogenesis of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). This review paper delves into the hypothesis that atherosclerosis, a prevalent age-related large vessel disease, extends its pathological influence into the cerebral microcirculation, thereby contributing to the development and progression of CSVD, with a specific focus on CMHs. We explore the concept of vascular aging as a continuum, bridging macrovascular pathologies like atherosclerosis with microvascular abnormalities characteristic of CSVD. We posit that the same risk factors precipitating accelerated aging in large vessels (i.e., atherogenesis), primarily through oxidative stress and inflammatory pathways, similarly instigate accelerated microvascular aging. Accelerated microvascular aging leads to increased microvascular fragility, which in turn predisposes to the formation of CMHs. The presence of hypertension and amyloid pathology further intensifies this process. We comprehensively overview the current body of evidence supporting this interconnected vascular hypothesis. Our review includes an examination of epidemiological data, which provides insights into the prevalence and impact of CMHs in the context of atherosclerosis and CSVD. Furthermore, we explore the shared mechanisms between large vessel aging, atherogenesis, microvascular aging, and CSVD, particularly focusing on how these intertwined processes contribute to the genesis of CMHs. By highlighting the role of vascular aging in the pathophysiology of CMHs, this review seeks to enhance the understanding of CSVD and its links to systemic vascular disorders. Our aim is to provide insights that could inform future therapeutic approaches and research directions in the realm of neurovascular health.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Illes Kovacs
- Department of Ophthalmology, Semmelweis University, 1085, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Angelia C Kirkpartrick
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xin A Zhang
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
40
|
Koemans EA, Perosa V, Freeze WM, Lee H, Kozberg MG, Coughlan GT, Buckley RF, Wermer MJ, Greenberg SM, van Veluw SJ. Sex differences in histopathological markers of cerebral amyloid angiopathy and related hemorrhage. Int J Stroke 2024; 19:947-956. [PMID: 38703035 PMCID: PMC11408965 DOI: 10.1177/17474930241255276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024]
Abstract
BACKGROUND Men with cerebral amyloid angiopathy (CAA) may have an earlier onset of intracerebral hemorrhage and a more hemorrhagic disease course compared to women. In this cohort study, we investigated sex differences in histopathological markers associated with amyloid-β burden and hemorrhage in cognitively impaired individuals and patients with CAA, using neuropathological data from two autopsy databases. METHODS First, we investigated presence of parenchymal (Thal score) and vascular amyloid-β (CAA severity score) in cognitively impaired individuals from the National Alzheimer's Coordinating Center (NACC) neuropathology database. Next, we examined sex differences in hemorrhagic ex vivo magnetic resonance imaging (MRI) markers and local cortical iron burden and the interaction of sex on factors associated with cortical iron burden (CAA percentage area and vessel remodeling) in patients with pathologically confirmed clinical CAA from the Massachusetts General Hospital (MGH) CAA neuropathology database. RESULTS In 6120 individuals from the NACC database (45% women, mean age 80 years), the presence of parenchymal amyloid-β (odds ratio (OR) (95% confidence interval (CI)) =0.68 (0.53-0.88)) but not vascular amyloid-β was less in men compared to women. In 19 patients with definite CAA from the MGH CAA database (35% women, mean age 75 years), a lower microbleed count (p < 0.001) but a higher proportion of cortical superficial siderosis and a higher local cortical iron burden was found in men (p < 0.001) compared to women. CAA percentage area was comparable in men and women (p = 0.732). Exploratory analyses demonstrated a possible stronger negative relation between cortical CAA percentage area and cortical iron density in men compared to women (p = 0.03). CONCLUSION Previously observed sex differences in hemorrhage onset and progression in CAA patients are likely not due to differences in global CAA severity between men and women. Other factors, such as vascular remodeling, may contribute, but future studies are necessary to replicate our findings in larger data sets and to further investigate the underlying mechanisms behind these complex sex differences.
Collapse
Affiliation(s)
- Emma A Koemans
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Valentina Perosa
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Whitney M Freeze
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hang Lee
- Department of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mariel G Kozberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gillian T Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marieke Jh Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
41
|
Howe MD, Caruso MR, Manoochehri M, Kunicki ZJ, Emrani S, Rudolph JL, Huey ED, Salloway SP, Oh H. Utility of cerebrovascular imaging biomarkers to detect cerebral amyloidosis. Alzheimers Dement 2024; 20:7220-7231. [PMID: 39219209 PMCID: PMC11485066 DOI: 10.1002/alz.14207] [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: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION The relationship between cerebrovascular disease (CVD) and amyloid beta (Aβ) in Alzheimer's disease (AD) is understudied. We hypothesized that magnetic resonance imaging (MRI)-based CVD biomarkers-including cerebral microbleeds (CMBs), lacunar infarction, and white matter hyperintensities (WMHs)-would correlate with Aβ positivity on positron emission tomography (Aβ-PET). METHODS We cross-sectionally analyzed data from the Alzheimer's Disease Neuroimaging Initiative (ADNI, N = 1352). Logistic regression was used to calculate odds ratios (ORs), with Aβ-PET positivity as the standard-of-truth. RESULTS Following adjustment, WMHs (OR = 1.25) and superficial CMBs (OR = 1.45) remained positively associated with Aβ-PET positivity (p < 0.001). Deep CMBs and lacunes exhibited a varied relationship with Aβ-PET in cognitive subgroups. The combined diagnostic model, which included CVD biomarkers and other accessible measures, significantly predicted Aβ-PET (pseudo-R2 = 0.41). DISCUSSION The study highlights the translational value of CVD biomarkers in diagnosing AD, and underscores the need for more research on their inclusion in diagnostic criteria. CLINICALTRIALS gov: ADNI-2 (NCT01231971), ADNI-3 (NCT02854033). HIGHLIGHTS Cerebrovascular biomarkers linked to amyloid beta (Aβ) in Alzheimer's disease (AD). White matter hyperintensities and cerebral microbleeds reliably predict Aβ-PET positivity. Relationships with Aβ-PET vary by cognitive stage. Novel accessible model predicts Aβ-PET status. Study supports multimodal diagnostic approaches.
Collapse
Affiliation(s)
- Matthew D. Howe
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Megan R. Caruso
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
| | | | - Zachary J. Kunicki
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Sheina Emrani
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - James L. Rudolph
- Center of Innovation in Long‐Term Services and Supports, Providence VA Medical CenterProvidenceRhode IslandUSA
- Department of MedicineThe Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Edward D. Huey
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Stephen P. Salloway
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Hwamee Oh
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | | |
Collapse
|
42
|
Gu H, Liu LL, Wu A, Yu Y, Emir U, Sawiak SJ, Territo PR, Farlow MR, Zheng W, Du Y. Lead Acetate Exposure and Cerebral Amyloid Accumulation: Mechanistic Evaluations in APP/PS1 Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:107004. [PMID: 39412896 PMCID: PMC11482597 DOI: 10.1289/ehp14384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The role of environmental factors in Alzheimer's disease (AD) pathogenesis remains elusive. Mounting evidence suggests that acute and past exposure to the environmental toxicant lead (Pb) is associated with longitudinal decline in cognitive function, brain atrophy, and greater brain β -amyloid (A β ) deposition. However, the nature of Pb-induced amyloid deposition and how it contributes to AD development remain unclear. OBJECTIVES This study investigates the role of Pb in the pathogenesis of cerebral amyloid angiopathy (CAA) and whether plasminogen activator inhibitor-1 (PAI-1) contributes to this process in the APP/PS1 mouse model. METHODS Female APP/PS1 mice at 8 wk of age were administered either 50 mg / kg Pb-acetate (PbAc) (i.e., 27 mg Pb / kg ) or an equivalent molar concentration of sodium acetate (NaAc) via oral gavage once daily for 8 wk. Amyloid deposition and vascular amyloid were determined by immunostaining. In addition, A β perivascular drainage, vascular binding assay, and microglial endocytosis were examined to determine underlying mechanisms. Furthermore, magnetic resonance imaging demyelination imaging was performed in vivo measure the level of demyelination. Finally, Y-maze and Morris water maze tests were assessed to evaluate the cognitive function of mice. RESULTS APP/PS1 mice (an AD mice model) exposed to PbAc demonstrated more vascular amyloid deposition less neocortical myelination, and lower cognitive function, as well as greater vascular binding to A β 40 , higher A β 40 / A β 42 ratios, strikingly lower A β 40 levels in the perivascular drainage, and microglial endocytosis. Importantly, exposure to a specific PAI-1 inhibitor, tiplaxtinin, which previously was reported to lower CAA pathology in mice, resulted in less CAA-related outcomes following PbAc exposure. DISCUSSION Our findings suggest that PbAc induced CAA/AD pathogenesis via the PAI-1 signaling in the APP/PS1 mouse model, and the inhibition of PAI-1 could be a potential therapeutic target for PbAc-mediated CAA/AD disorders. https://doi.org/10.1289/EHP14384.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Luqing L. Liu
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Alanna Wu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yongqi Yu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Uzay Emir
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Stephen J. Sawiak
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Paul R. Territo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
43
|
Rohm Z, Goldman MD, Riley C, Zamvil SS, Pawate S. A 73-Year-Old Woman With Confusion, Visual Field Disturbances, and Edematous White Matter Lesions: From the National Multiple Sclerosis Society Case Conference Proceedings. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200300. [PMID: 39141887 PMCID: PMC11379432 DOI: 10.1212/nxi.0000000000200300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
We describe the case of a 73-year-old woman presenting with headaches, confusion, and vision disturbances. Brain MRI showed a large T2-hyperintense lesion in the right temporo-occipital region with vasogenic edema and leptomeningeal enhancement. A leptomeningeal biopsy was performed, which led to a definitive diagnosis.
Collapse
Affiliation(s)
- Zachery Rohm
- From the Department of Neurology (Z.R., S.P.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurology (M.D.G.), Virginia Commonwealth University, Richmond; Department of Neurology (C.R.), Columbia University; and Department of Neurology (S.S.Z.), University of California San Francisco
| | - Myla D Goldman
- From the Department of Neurology (Z.R., S.P.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurology (M.D.G.), Virginia Commonwealth University, Richmond; Department of Neurology (C.R.), Columbia University; and Department of Neurology (S.S.Z.), University of California San Francisco
| | - Claire Riley
- From the Department of Neurology (Z.R., S.P.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurology (M.D.G.), Virginia Commonwealth University, Richmond; Department of Neurology (C.R.), Columbia University; and Department of Neurology (S.S.Z.), University of California San Francisco
| | - Scott S Zamvil
- From the Department of Neurology (Z.R., S.P.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurology (M.D.G.), Virginia Commonwealth University, Richmond; Department of Neurology (C.R.), Columbia University; and Department of Neurology (S.S.Z.), University of California San Francisco
| | - Siddharama Pawate
- From the Department of Neurology (Z.R., S.P.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurology (M.D.G.), Virginia Commonwealth University, Richmond; Department of Neurology (C.R.), Columbia University; and Department of Neurology (S.S.Z.), University of California San Francisco
| |
Collapse
|
44
|
Pathan N, Kharod MK, Nawab S, Di Scipio M, Paré G, Chong M. Genetic Determinants of Vascular Dementia. Can J Cardiol 2024; 40:1412-1423. [PMID: 38579965 DOI: 10.1016/j.cjca.2024.03.025] [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: 02/05/2024] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
Vascular dementia (VaD) is a prevalent form of cognitive impairment with underlying vascular etiology. In this review, we examine recent genetic advancements in our understanding of VaD, encompassing a range of methodologies including genome-wide association studies, polygenic risk scores, heritability estimates, and family studies for monogenic disorders revealing the complex and heterogeneous nature of the disease. We report well known genetic associations and highlight potential pathways and mechanisms implicated in VaD and its pathological risk factors, including stroke, cerebral small vessel disease, and cerebral amyloid angiopathy. Moreover, we discuss important modifiable risk factors such as hypertension, diabetes, and dyslipidemia, emphasizing the importance of a multifactorial approach in prevention, treatment, and understanding the genetic basis of VaD. Last, we outline several areas of scientific advancements to improve clinical care, highlighting that large-scale collaborative efforts, together with an integromics approach can enhance the robustness of genetic discoveries. Indeed, understanding the genetics of VaD and its pathophysiological risk factors hold the potential to redefine VaD on the basis of molecular mechanisms and to generate novel diagnostic, prognostic, and therapeutic tools.
Collapse
Affiliation(s)
- Nazia Pathan
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada
| | - Muskaan Kaur Kharod
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Sajjha Nawab
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada; Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.
| | - Michael Chong
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada.
| |
Collapse
|
45
|
Bonaterra-Pastra A, Solé M, Lope-Piedrafita S, Lucas-Parra M, Castellote L, Marazuela P, Pancorbo O, Rodríguez-Luna D, Hernández-Guillamon M. The presence of circulating human apolipoprotein J reduces the occurrence of cerebral microbleeds in a transgenic mouse model with cerebral amyloid angiopathy. Alzheimers Res Ther 2024; 16:169. [PMID: 39069622 DOI: 10.1186/s13195-024-01541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β (Aβ) deposition in cerebral vessels, leading to lobar cerebral microbleeds (CMB) and intracerebral hemorrhages (ICH). Apolipoprotein J (ApoJ) is a multifunctional chaperone related to Aβ aggregation and clearance. Our study investigated the vascular impact of chronic recombinant human Apolipoprotein J (rhApoJ) treatment in a transgenic mouse model of β-amyloidosis with prominent CAA. METHODS Twenty-month-old APP23 C57BL/6 mice received 25 doses of rhApoJ (1 mg/kg) (n = 9) or saline (n = 8) intraperitoneally for 13 weeks, while Wild-type (WT) mice received saline (n = 13). Postmortem brains underwent T2*-weighted magnetic resonance imaging (MRI) to detect hemorrhagic lesions. Aβ levels and distribution, cerebral fibrinogen leakage, brain smooth muscle actin (sma), and plasma matrix metalloproteinases and inflammatory markers were analyzed after treatments. Additionally, plasma samples from 22 patients with lobar ICH were examined to determine the clinical relevance of the preclinical findings. RESULTS rhApoJ-treated APP23 presented fewer cortical CMBs (50-300 μm diameter) (p = 0.012) and cortical larger hemorrhages (> 300 μm) (p = 0.002) than saline-treated mice, independently of Aβ brain levels. MRI-detected hemorrhagic lesions correlated with fibrinogen cerebral extravasation (p = 0.011). Additionally, rhApoJ-treated mice presented higher number of sma-positive vessels than saline-treated mice (p = 0.038). In rhApoJ-treated mice, human ApoJ was detected in plasma and in occasional leptomeningeal vessels, but not in the parenchyma, suggesting that its mechanism of action operates through the periphery. The administration of rhApoJ induced an increase in plasma Groα (p = 0.035) and MIP-1α (p = 0.035) levels, while lower MMP-12 (p = 0.046) levels, compared to the saline-treated group. In acute lobar ICH patients, MMP-12 plasma levels correlated with larger hemorrhage volume (p = 0.040) and irregular ICH shape (p = 0.036). CONCLUSIONS Chronic rhApoJ treatment in aged APP23 mice ameliorated CAA-related neurovascular damage by reducing the occurrence of CMB. We propose that rhApoJ may prevent blood-brain barrier (BBB) leakage and CMB appearance partly through circulating MMP-12 modulation.
Collapse
Affiliation(s)
- Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
- Department of Bioquímica i Biologia Molecular i Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Silvia Lope-Piedrafita
- Nuclear Magnetic Resonance Service, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria Lucas-Parra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Laura Castellote
- Department of Clinical Biochemistry, Clinical Laboratories, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Olalla Pancorbo
- Stroke Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain.
| |
Collapse
|
46
|
Kasri A, Camporesi E, Gkanatsiou E, Boluda S, Brinkmalm G, Stimmer L, Ge J, Hanrieder J, Villain N, Duyckaerts C, Vermeiren Y, Pape SE, Nicolas G, Laquerrière A, De Deyn PP, Wallon D, Blennow K, Strydom A, Zetterberg H, Potier MC. Amyloid-β peptide signature associated with cerebral amyloid angiopathy in familial Alzheimer's disease with APPdup and Down syndrome. Acta Neuropathol 2024; 148:8. [PMID: 39026031 PMCID: PMC11258176 DOI: 10.1007/s00401-024-02756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid plaques containing amyloid-β (Aβ) peptides, intraneuronal neurofibrillary tangles, extracellular neuropil threads, and dystrophic neurites surrounding plaques composed of hyperphosphorylated tau protein (pTau). Aβ can also deposit in blood vessel walls leading to cerebral amyloid angiopathy (CAA). While amyloid plaques in AD brains are constant, CAA varies among cases. The study focuses on differences observed between rare and poorly studied patient groups with APP duplications (APPdup) and Down syndrome (DS) reported to have higher frequencies of elevated CAA levels in comparison to sporadic AD (sAD), most of APP mutations, and controls. We compared Aβ and tau pathologies in postmortem brain tissues across cases and Aβ peptides using mass spectrometry (MS). We further characterized the spatial distribution of Aβ peptides with MS-brain imaging. While intraparenchymal Aβ deposits were numerous in sAD, DS with AD (DS-AD) and AD with APP mutations, these were less abundant in APPdup. On the contrary, Aβ deposits in the blood vessels were abundant in APPdup and DS-AD while only APPdup cases displayed high Aβ deposits in capillaries. Investigation of Aβ peptide profiles showed a specific increase in Aβx-37, Aβx-38 and Aβx-40 but not Aβx-42 in APPdup cases and to a lower extent in DS-AD cases. Interestingly, N-truncated Aβ2-x peptides were particularly increased in APPdup compared to all other groups. This result was confirmed by MS-imaging of leptomeningeal and parenchymal vessels from an APPdup case, suggesting that CAA is associated with accumulation of shorter Aβ peptides truncated both at N- and C-termini in blood vessels. Altogether, this study identified striking differences in the localization and composition of Aβ deposits between AD cases, particularly APPdup and DS-AD, both carrying three genomic copies of the APP gene. Detection of specific Aβ peptides in CSF or plasma of these patients could improve the diagnosis of CAA and their inclusion in anti-amyloid immunotherapy treatments.
Collapse
Affiliation(s)
- Amal Kasri
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Elena Camporesi
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eleni Gkanatsiou
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Susana Boluda
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lev Stimmer
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Junyue Ge
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörg Hanrieder
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Nicolas Villain
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Charles Duyckaerts
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
| | - Sarah E Pape
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Gaël Nicolas
- Department of Genetics, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Peter Paul De Deyn
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - David Wallon
- Department of Neurology, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, 76000, Rouen, France
| | - Kaj Blennow
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Andre Strydom
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
- UK Dementia Research Institute at UCL, London, UK.
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China.
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA.
| | - Marie-Claude Potier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France.
| |
Collapse
|
47
|
van den Berg E, Kersten I, Brinkmalm G, Johansson K, de Kort AM, Klijn CJ, Schreuder FH, Gobom J, Stoops E, Portelius E, Gkanatsiou E, Zetterberg H, Blennow K, Kuiperij HB, Verbeek MM. Profiling amyloid-β peptides as biomarkers for cerebral amyloid angiopathy. J Neurochem 2024; 168:1254-1264. [PMID: 38362804 PMCID: PMC11260253 DOI: 10.1111/jnc.16074] [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: 10/25/2023] [Revised: 01/08/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Brain amyloid-β (Aβ) deposits are key pathological hallmarks of both cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD). Microvascular deposits in CAA mainly consist of the Aβ40 peptide, whereas Aβ42 is the predominant variant in parenchymal plaques in AD. The relevance in pathogenesis and diagnostic accuracy of various other Aβ isoforms in CAA remain understudied. We aimed to investigate the biomarker potential of various Aβ isoforms in cerebrospinal fluid (CSF) to differentiate CAA from AD pathology. We included 25 patients with probable CAA, 50 subjects with a CSF profile indicative of AD pathology (AD-like), and 23 age- and sex-matched controls. CSF levels of Aβ1-34, Aβ1-37, Aβ1-38, Aβ1-39, Aβ1-40, and Aβ1-42 were quantified by liquid chromatography mass spectrometry. Lower CSF levels of all six Aβ peptides were observed in CAA patients compared with controls (p = 0.0005-0.03). Except for Aβ1-42 (p = 1.0), all peptides were decreased in CAA compared with AD-like subjects (p = 0.007-0.03). Besides Aβ1-42, none of the Aβ peptides were decreased in AD-like subjects compared with controls. All Aβ peptides combined differentiated CAA from AD-like subjects better (area under the curve [AUC] 0.84) than individual peptide levels (AUC 0.51-0.75). Without Aβ1-42 in the model (since decreased Aβ1-42 served as AD-like selection criterion), the AUC was 0.78 for distinguishing CAA from AD-like subjects. CAA patients and AD-like subjects showed distinct disease-specific CSF Aβ profiles. Peptides shorter than Aβ1-42 were decreased in CAA patients, but not AD-like subjects, which could suggest different pathological mechanisms between vascular and parenchymal Aβ accumulation. This study supports the potential use of this panel of CSF Aβ peptides to indicate presence of CAA pathology with high accuracy.
Collapse
Affiliation(s)
- Emma van den Berg
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris Kersten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kjell Johansson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Anna M. de Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catharina J.M. Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris H.B.M. Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Gobom
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Erik Portelius
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eleni Gkanatsiou
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - H. Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M. Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Narasimhan S, Holtzman DM, Apostolova LG, Cruchaga C, Masters CL, Hardy J, Villemagne VL, Bell J, Cho M, Hampel H. Apolipoprotein E in Alzheimer's disease trajectories and the next-generation clinical care pathway. Nat Neurosci 2024; 27:1236-1252. [PMID: 38898183 DOI: 10.1038/s41593-024-01669-5] [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: 08/14/2023] [Accepted: 04/18/2024] [Indexed: 06/21/2024]
Abstract
Alzheimer's disease (AD) is a complex, progressive primary neurodegenerative disease. Since pivotal genetic studies in 1993, the ε4 allele of the apolipoprotein E gene (APOE ε4) has remained the strongest single genome-wide associated risk variant in AD. Scientific advances in APOE biology, AD pathophysiology and ApoE-targeted therapies have brought APOE to the forefront of research, with potential translation into routine AD clinical care. This contemporary Review will merge APOE research with the emerging AD clinical care pathway and discuss APOE genetic risk as a conduit to genomic-based precision medicine in AD, including ApoE's influence in the ATX(N) biomarker framework of AD. We summarize the evidence for APOE as an important modifier of AD clinical-biological trajectories. We then illustrate the utility of APOE testing and the future of ApoE-targeted therapies in the next-generation AD clinical-diagnostic pathway. With the emergence of new AD therapies, understanding how APOE modulates AD pathophysiology will become critical for personalized AD patient care.
Collapse
Affiliation(s)
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University in St. Louis, St. Louis, MO, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Neurosciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- Florey Institute and the University of Melbourne, Parkville, Victoria, Australia
| | - John Hardy
- Department of Neurodegenerative Disease and Dementia Research Institute, Reta Lila Weston Research Laboratories, UCL Institute of Neurology, Queen Square, London, UK
| | | | | | | | | |
Collapse
|
49
|
Costa AS, Albrecht M, Reich A, Nikoubashman O, Schulz JB, Reetz K, Pinho J. Non-hemorrhagic imaging markers of cerebral amyloid angiopathy in memory clinic patients. Alzheimers Dement 2024; 20:4792-4802. [PMID: 38865440 PMCID: PMC11247708 DOI: 10.1002/alz.13920] [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: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION The Boston criteria v2.0 for cerebral amyloid angiopathy (CAA) incorporated non-hemorrhagic imaging markers. Their prevalence and significance in patients with cognitive impairment remain uncertain. METHODS We studied 622 memory clinic patients with available magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) biomarkers. Two raters assessed non-hemorrhagic markers, and we explored their association with clinical characteristics through multivariate analyses. RESULTS Most patients had mild cognitive impairment; median age was 71 years and 50% were female. Using the v2.0 criteria, possible or probable CAA increased from 75 to 383 patients. Sixty-eight percent of the sample had non-hemorrhagic CAA markers, which were independently associated with age (odds ratio [OR] = 1.04, 95% confidence interval [CI] = 1.01-1.07), female sex (OR = 1.68, 95% CI = 1.11-2.54), and hemorrhagic CAA markers (OR = 2.11, 95% CI = 1.02-4.35). DISCUSSION Two-thirds of patients from a memory clinic cohort had non-hemorrhagic CAA markers, increasing the number of patients meeting the v2.0 CAA criteria. Longitudinal approaches should explore the implications of these markers, particularly the hemorrhagic risk in this population. HIGHLIGHTS The updated Boston criteria for cerebral amyloid angiopathy (CAA) now include non-hemorrhagic markers. The prevalence of non-hemorrhagic CAA markers in memory clinic patients is unknown. Two-thirds of patients in our memory clinic presented non-hemorrhagic CAA markers. The presence of these markers was associated with age, female sex, and hemorrhagic CAA markers. The hemorrhagic risk of patients presenting these type of markers remains unclear.
Collapse
Affiliation(s)
- Ana Sofia Costa
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - Milena Albrecht
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| | - Arno Reich
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| | - Omid Nikoubashman
- Department of Diagnostic and Interventional NeuroradiologyUniversity Hospital RWTH AachenAachenGermany
| | - Jörg B. Schulz
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - Kathrin Reetz
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - João Pinho
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| |
Collapse
|
50
|
Jäkel L, De Kort AM, Stellingwerf A, Hernández Utrilla C, Kersten I, Vervuurt M, Vermeiren Y, Küsters B, Schreuder FHBM, Klijn CJM, Kuiperij HB, Verbeek MM. Altered brain expression and cerebrospinal fluid levels of TIMP4 in cerebral amyloid angiopathy. Acta Neuropathol Commun 2024; 12:103. [PMID: 38915119 PMCID: PMC11194996 DOI: 10.1186/s40478-024-01823-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a highly prevalent and progressive pathology, involving amyloid-β (Aβ) deposition in the cerebral blood vessel walls. CAA is associated with an increased risk for intracerebral hemorrhages (ICH). Insight into the molecular mechanisms associated with CAA pathology is urgently needed, to develop additional diagnostic tools to allow for reliable and early diagnosis of CAA and to obtain novel leads for the development of targeted therapies. Tissue inhibitor of matrix metalloproteinases 4 (TIMP4) is associated with cardiovascular functioning and disease and has been linked to vascular dementia. Using immunohistochemistry, we studied occipital brain tissue samples of 57 patients with CAA (39 without ICH and 18 with ICH) and 42 controls, and semi-quantitatively assessed expression levels of TIMP4. Patients with CAA had increased vascular expression of TIMP4 compared to controls (p < 0.001), and in these patients, TIMP4 expression correlated with CAA severity (τb = 0.38; p = 0.001). Moreover, TIMP4 expression was higher in CAA-ICH compared to CAA-non-ICH cases (p = 0.024). In a prospective cross-sectional study of 38 patients with CAA and 37 age- and sex-matched controls, we measured TIMP4 levels in cerebrospinal fluid (CSF) and serum using ELISA. Mean CSF levels of TIMP4 were decreased in patients with CAA compared to controls (3.36 ± 0.20 vs. 3.96 ± 0.22 ng/ml, p = 0.033), whereas median serum levels were increased in patients with CAA (4.51 ng/ml [IQR 3.75-5.29] vs 3.60 ng/ml [IQR 3.11-4.85], p-9.013). Moreover, mean CSF TIMP4 levels were lower in CAA patients who had experienced a symptomatic hemorrhage compared to CAA patients who did not (2.13 ± 0.24 vs. 3.57 ± 0.24 ng/ml, p = 0.007). CSF TIMP4 levels were associated with CSF levels of Aβ40 (spearman r (rs) = 0.321, p = 0.009). In summary, we show that TIMP4 is highly associated with CAA and CAA-related ICH, which is reflected by higher levels in the cerebral vasculature and lower levels in CSF. With these findings we provide novel insights into the pathophysiology of CAA, and more specifically in CAA-associated ICH.
Collapse
Affiliation(s)
- Lieke Jäkel
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna M De Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arno Stellingwerf
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla Hernández Utrilla
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris Kersten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marc Vervuurt
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yannick Vermeiren
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
- Faculty of Medicine and Health Sciences, Translational Neurosciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Catharina J M Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 830 TML, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
- Radboud Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|