1
|
Sin MK, Cheng Y, Ahmed A, Roseman JM, Dowling NM, Zamrini E. Cerebral Amyloid Angiopathy, Dementia, and Alzheimer Neuropathologic Changes: Findings From the ACT Autopsy Cohort. Neurology 2024; 103:e210009. [PMID: 39481068 PMCID: PMC11527483 DOI: 10.1212/wnl.0000000000210009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/04/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral amyloid angiopathy (CAA) is common in older adults and is associated with dementia. Less is known whether this association is mediated by Alzheimer disease (AD) neuropathologic changes, the examination of which was the objective of this study. METHODS This was a retrospective cross-sectional examination of the Kaiser Permanente Washington database of the Adult Changes in Thought (ACT) autopsy cohort with information on CAA, dementia, the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) (amyloid neuritic plaques), and Braak (tau neurofibrillary tangles). CAA was diagnosed by immunohistochemistry and dementia by ACT Consensus Diagnostic Conference. AD neuropathology was categorized by CERAD scores and Braak stages. Multivariable logistic regression models were used to estimate odds ratios (ORs) and 95% CIs of the associations of CAA with dementia, adjusting for age at death and sex, and with additional adjustments separately for CERAD scores (moderate-severe vs mild-absent), Braak stages (V-VI vs 0-IV), APOE ε4, and stroke. Formal mediation analyses were conducted to estimate age-sex-adjusted OR (95% CI) for natural indirect effects (NIEs) of CERAD scores and Braak stages. RESULTS The 848 participants had a mean age of 86.7 ± 4.6 years at death, and 57.6% were female. CAA was present in 322 participants (38.0%), of whom 152, 145, and 25 had mild, moderate, and severe CAA, respectively. Dementia was present in 384 participants (45.3%), of whom 317 had AD. Dementia was more common in those with CAA than without (53.7% vs 40.1%; age-sex-adjusted OR 1.57, 95% CI 1.18-2.10). This association remained significant after separate adjustment for other covariates but lost significance when adjusted for CERAD scores (OR 1.27, 95% CI 0.93-1.71) and Braak stages (OR 0.96, 95% CI 0.69-1.33). Findings from our formal mediation analyses show that ORs (95% CIs) for NIE of CERAD scores and Braak stages were 1.25 (1.13-1.37) and 1.63 (1.38-1.88), respectively, and CERAD scores and Braak stages mediated 53% and 111% of the total association, respectively. DISCUSSION We observed a significant association between CAA and dementia that disappeared when adjusted for CERAD or Braak stages. Findings from our mediation analyses suggest that the CAA-dementia association may be potentially mediated by AD neuropathologic changes. This hypothesis needs to be tested in future mechanistic studies in AD accounting for unmeasured confounders.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Yan Cheng
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Ali Ahmed
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Jeffrey M Roseman
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - N Maritza Dowling
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Edward Zamrini
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Sousa L, Pinto C, Azevedo A, Igreja L, Marta A, Fernandes J, Oliveira P, Cardoso M, Alves C, Silva AMD, Mendonça Pinto M, Sousa AP, Coelho T, Taipa R. Brain MRI in patients with V30M hereditary transthyretin amyloidosis. Amyloid 2024:1-6. [PMID: 39153196 DOI: 10.1080/13506129.2024.2391842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Central nervous system dysfunction is common in longstanding hereditary transthyretin amyloidosis (ATTRv) caused by the V30M (p.V50M) mutation. Neuropathology studies show leptomeningeal amyloid deposition and cerebral amyloid angiopathy (CAA). Brain MRI is widely used in the assessment of Aβ associated CAA but there are no systematic studies with brain MRI in ATTRv amyloidosis. METHODS we performed 3 T brain MRIs in 16 patients with longstanding (>14 years) ATTRV30M. We additionally retrospectively reviewed 48 brain MRIs from patients followed at our clinic. CNS symptoms and signs were systematically accessed, and MRIs were blindly reviewed for ischaemic and haemorrhagic lesions. RESULTS in the prospective cohort, we found white matter hyperintensities in 8/16 patients (50%, Fazekas score> =1). There were no relevant microbleeds, large ischaemic or haemorrhagic lesions or superficial siderosis. In the retrospective cohort, microbleeds were found in 5/48 patients (10,4%), two of which with > =20 microbleeds. White matter hyperintensities were found in 20/48 cases (41.7%). White matter lesions, microbleeds and cortical atrophy were not associated with disease duration. CONCLUSIONS white matter hyperintensities are common in ATTRV30M, irrespective of disease duration. Haemorrhagic lesions are rare, even in patients with longstanding disease, suggesting the existence of other risk factors.
Collapse
Affiliation(s)
- Luísa Sousa
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
- Neurology Department, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Catarina Pinto
- Neuroradiology Department, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Ana Azevedo
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Neurology Department, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Liliana Igreja
- Neuroradiology Department, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Ana Marta
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Ophthalmology Department, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Joana Fernandes
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Pedro Oliveira
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Márcio Cardoso
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Cristina Alves
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Ana Martins da Silva
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Miguel Mendonça Pinto
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
- Portuguese Brain Bank, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Ana Paula Sousa
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Teresa Coelho
- Unidade Corino de Andrade, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Ricardo Taipa
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Portuguese Brain Bank, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| |
Collapse
|
5
|
Wu J, Liu Z, Yao M, Zhu Y, Peng B, Ni J. Clinical characteristics of cerebral amyloid angiopathy and risk factors of cerebral amyloid angiopathy related intracerebral hemorrhage. J Neurol 2024; 271:5025-5034. [PMID: 38796800 DOI: 10.1007/s00415-024-12451-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVES There is limited understanding of the differences between cerebral amyloid angiopathy (CAA) with and without intracerebral hemorrhage (ICH). This article aimed to describe the characteristics of CAA and identify the risk factors of CAA-ICH in a multicenter cohort. METHODS Patients consecutively enrolled in the national multicenter prospective Cerebral Small Vessel Disease Cohort Study who met the Boston diagnostic criteria for CAA or CAA-related inflammation were included in this study. The demographic characteristics and clinical data were collected. The clinical and radiographic differences between CAA with and without ICH were compared to identify the risk factors for CAA-ICH. RESULTS A total of 219 CAA patients were included, with an average age of 67.12 ± 9.93. Of all patients, 26.0% were CAA with ICH. Univariate analysis showed that CAA-ICH is associated with carrying more APOE ε2 allele, less lobar cerebral microbleeds (CMBs), cortical superficial siderosis (cSS), lower Fazekas scale, a tendency of gait disorder, and acute onset (P < 0.05). The generalized linear mixed model yielded statistically significant associations between CAA with ICH and carrying the APOE ε2 allele, cSS, the lower number of lobar CMBs, and the lower Fazekas scale (P < 0.05). CONCLUSION It is meaningful to classify CAA with and without ICH, as there may be different mechanisms between the two. CAA with ICH has a susceptibility to carrying APOE ε2, cSS, and a relatively small number of CMBs. Fewer CMBs do not mean lower susceptibility to ICH in CAA. Larger prospective cohort studies are necessary to further clarify these conclusions.
Collapse
Affiliation(s)
- Juanjuan Wu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziyue Liu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ming Yao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yicheng Zhu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bin Peng
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jun Ni
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
6
|
Sin MK, Dowling NM, Roseman JM, Ahmed A, Zamrini E. Late-Life Blood Pressure and Cerebral Amyloid Angiopathy: Findings from the U.S. National Alzheimer's Coordinating Center Uniform Dataset. Neurol Int 2024; 16:821-832. [PMID: 39195563 DOI: 10.3390/neurolint16040061] [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: 05/16/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
High blood pressure (BP) and cerebral amyloid angiopathy (CAA) are two common risk factors for intracranial hemorrhage, potentially leading to cognitive impairment. Less is known about the relationship between BP and CAA, the examination of which was the objective of this study. We analyzed data from 2510 participants in the National Alzheimer's Coordinating Center (NACC) who had information on longitudinal BP measurements before death and on CAA from autopsy. Using the average of four systolic BPs (SBPs) prior to death, SBP was categorized into three groups: <120 mmHg (n = 435), 120-139 mmHg (n = 1335), and ≥140 mmHg (n = 740). CAA was diagnosed using immunohistochemistry in 1580 participants and categorized as mild (n = 759), moderate (n = 529), or severe (n = 292). When adjusted for age at death, sex, APOE genotype, Braak, CERAD, antihypertensive medication use, and microinfarcts, the odds ratios (95% CIs) for CAA associated with SBPs of 120-139 and ≥140 mmHg were 0.91 (0.74-1.12) and 1.00 (0.80-1.26), respectively. Findings from predictor effect plots show no variation in the probability of CAA between the three SBP categories. Microbleeds had no association with CAA, but among those with SBP ≥ 130 mmHg, the proportion of those with microbleeds was numerically greater in those with more severe CAA (p for trend, 0.084). In conclusion, we found no evidence of an association between SBP and CAA. Future studies need to develop non-invasive laboratory tests to diagnose CAA and prospectively examine this association and its implication on the pathophysiology and outcome of Alzheimer's disease.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- College of Nursing, Seattle University, Seattle, WA 98122, USA
| | - N Maritza Dowling
- Department of Acute & Chronic Care, School of Nursing, George Washington University, Washington, DC 20147, USA
- Department of Epidemiology & Biostatistics, Milken School of Public Health, George Washington University, Washington, DC 20147, USA
| | - Jeffrey M Roseman
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ali Ahmed
- Center for Data Science and Outcomes Research, Veterans Affairs Medical Center, Washington, DC 20242, USA
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA
- Department of Medicine, School of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Edward Zamrini
- Center for Data Science and Outcomes Research, Veterans Affairs Medical Center, Washington, DC 20242, USA
- Department of Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA
- Biomedical Informatics Center, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
7
|
Im Y, Kang SH, Park G, Yoo H, Chun MY, Kim CH, Park CJ, Kim JP, Jang H, Kim HJ, Oh K, Koh SB, Lee JM, Na DL, Seo SW, Kim H. Ethnic differences in the effects of apolipoprotein E ɛ4 and vascular risk factors on accelerated brain aging. Brain Commun 2024; 6:fcae213. [PMID: 39007039 PMCID: PMC11242459 DOI: 10.1093/braincomms/fcae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/30/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024] Open
Abstract
The frequency of the apolipoprotein E ɛ4 allele and vascular risk factors differs among ethnic groups. We aimed to assess the combined effects of apolipoprotein E ɛ4 and vascular risk factors on brain age in Korean and UK cognitively unimpaired populations. We also aimed to determine the differences in the combined effects between the two populations. We enrolled 2314 cognitively unimpaired individuals aged ≥45 years from Korea and 6942 cognitively unimpaired individuals from the UK, who were matched using propensity scores. Brain age was defined using the brain age index. The apolipoprotein E genotype (ɛ4 carriers, ɛ2 carriers and ɛ3/ɛ3 homozygotes) and vascular risk factors (age, hypertension and diabetes) were considered predictors. Apolipoprotein E ɛ4 carriers in the Korean (β = 0.511, P = 0.012) and UK (β = 0.302, P = 0.006) groups had higher brain age index values. The adverse effects of the apolipoprotein E genotype on brain age index values increased with age in the Korean group alone (ɛ2 carriers × age, β = 0.085, P = 0.009; ɛ4 carriers × age, β = 0.100, P < 0.001). The apolipoprotein E genotype, age and ethnicity showed a three-way interaction with the brain age index (ɛ2 carriers × age × ethnicity, β = 0.091, P = 0.022; ɛ4 carriers × age × ethnicity, β = 0.093, P = 0.003). The effects of apolipoprotein E on the brain age index values were more pronounced in individuals with hypertension in the Korean group alone (ɛ4 carriers × hypertension, β = 0.777, P = 0.038). The apolipoprotein E genotype, age and ethnicity showed a three-way interaction with the brain age index (ɛ4 carriers × hypertension × ethnicity, β=1.091, P = 0.014). We highlight the ethnic differences in the combined effects of the apolipoprotein E ɛ4 genotype and vascular risk factors on accelerated brain age. These findings emphasize the need for ethnicity-specific strategies to mitigate apolipoprotein E ɛ4-related brain aging in cognitively unimpaired individuals.
Collapse
Affiliation(s)
- Yanghee Im
- USC Steven Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Korea
| | - Sung Hoon Kang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Gilsoon Park
- USC Steven Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Heejin Yoo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Min Young Chun
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea
| | - Chi-Hun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| | - Chae Jung Park
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Kyungmi Oh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
| | - Hosung Kim
- USC Steven Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
8
|
Koemans EA, Rasing I, Voigt S, van Harten TW, van der Zwet RG, Kaushik K, Schipper MR, van der Weerd N, van Zwet EW, van Etten ES, van Osch MJ, Kuiperij B, Verbeek MM, Terwindt GM, Greenberg SM, van Walderveen MA, Wermer MJ. Temporal Ordering of Biomarkers in Dutch-Type Hereditary Cerebral Amyloid Angiopathy. Stroke 2024; 55:954-962. [PMID: 38445479 PMCID: PMC10962436 DOI: 10.1161/strokeaha.123.044688] [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: 07/31/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND The temporal ordering of biomarkers for cerebral amyloid angiopathy (CAA) is important for their use in trials and for the understanding of the pathological cascade of CAA. We investigated the presence and abnormality of the most common biomarkers in the largest (pre)symptomatic Dutch-type hereditary CAA (D-CAA) cohort to date. METHODS We included cross-sectional data from participants with (pre)symptomatic D-CAA and controls without CAA. We investigated CAA-related cerebral small vessel disease markers on 3T-MRI, cerebrovascular reactivity with functional 7T-MRI (fMRI) and amyloid-β40 and amyloid-β42 levels in cerebrospinal fluid. We calculated frequencies and plotted biomarker abnormality according to age to form scatterplots. RESULTS We included 68 participants with D-CAA (59% presymptomatic, mean age, 50 [range, 26-75] years; 53% women), 53 controls (mean age, 51 years; 42% women) for cerebrospinal fluid analysis and 36 controls (mean age, 53 years; 100% women) for fMRI analysis. Decreased cerebrospinal fluid amyloid-β40 and amyloid-β42 levels were the earliest biomarkers present: all D-CAA participants had lower levels of amyloid-β40 and amyloid-β42 compared with controls (youngest participant 30 years). Markers of nonhemorrhagic injury (>20 enlarged perivascular spaces in the centrum semiovale and white matter hyperintensities Fazekas score, ≥2, present in 83% [n=54]) and markers of impaired cerebrovascular reactivity (abnormal BOLD amplitude, time to peak and time to baseline, present in 56% [n=38]) were present from the age of 30 years. Finally, markers of hemorrhagic injury were present in 64% (n=41) and only appeared after the age of 41 years (first microbleeds and macrobleeds followed by cortical superficial siderosis). CONCLUSIONS Our results suggest that amyloid biomarkers in cerebrospinal fluid are the first to become abnormal in CAA, followed by MRI biomarkers for cerebrovascular reactivity and nonhemorrhagic injury and lastly hemorrhagic injury. This temporal ordering probably reflects the pathological stages of CAA and should be taken into account when future therapeutic trials targeting specific stages are designed.
Collapse
Affiliation(s)
- Emma A. Koemans
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Ingeborg Rasing
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Sabine Voigt
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
- Radiology (S.V., T.W.v.H., M.R.S., M.J.v.P.O., M.A.A.v.W.), Leiden University Medical Center, the Netherlands
| | - Thijs W. van Harten
- Radiology (S.V., T.W.v.H., M.R.S., M.J.v.P.O., M.A.A.v.W.), Leiden University Medical Center, the Netherlands
| | - Reinier G.J. van der Zwet
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Kanishk Kaushik
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Manon R. Schipper
- Radiology (S.V., T.W.v.H., M.R.S., M.J.v.P.O., M.A.A.v.W.), Leiden University Medical Center, the Netherlands
| | - Nelleke van der Weerd
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Erik W. van Zwet
- Biostatistics (E.W.v.Z.), Leiden University Medical Center, the Netherlands
| | - Ellis S. van Etten
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Matthias J.P. van Osch
- Radiology (S.V., T.W.v.H., M.R.S., M.J.v.P.O., M.A.A.v.W.), Leiden University Medical Center, the Netherlands
| | - Bea Kuiperij
- Department Neurology and Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen (B.K., M.M.V.)
| | - Marcel M. Verbeek
- Department Neurology and Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen (B.K., M.M.V.)
| | - Gisela M. Terwindt
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
| | - Steven M. Greenberg
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (S.M.G.)
| | | | - Marieke J.H. Wermer
- Departments of Neurology (E.A.K., I.R., S.V., R.G.J.v.d.Z., K.K., N.v.d.W., E.S.v.E., G.M.T., M.J.H.W.), Leiden University Medical Center, the Netherlands
- Department of Neurology, University Medical Center Groningen, the Netherlands (M.J.H.W.)
| |
Collapse
|
9
|
Raposo N, Périole C, Planton M. In-vivo diagnosis of cerebral amyloid angiopathy: an updated review. Curr Opin Neurol 2024; 37:19-25. [PMID: 38038409 DOI: 10.1097/wco.0000000000001236] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
PURPOSE OF REVIEW Sporadic cerebral amyloid angiopathy (CAA) is a highly prevalent small vessel disease in ageing population with potential severe complications including lobar intracerebral hemorrhage (ICH), cognitive impairment, and dementia. Although diagnosis of CAA was made only with postmortem neuropathological examination a few decades ago, diagnosing CAA without pathological proof is now allowed in living patients. This review focuses on recently identified biomarkers of CAA and current diagnostic criteria. RECENT FINDINGS Over the past few years, clinicians and researchers have shown increased interest for CAA, and important advances have been made. Thanks to recent insights into mechanisms involved in CAA and advances in structural and functional neuroimaging, PET amyloid tracers, cerebrospinal fluid and plasma biomarkers analysis, a growing number of biomarkers of CAA have been identified. Imaging-based diagnostic criteria including emerging biomarkers have been recently developed or updated, enabling accurate and earlier diagnosis of CAA in living patients. SUMMARY Recent advances in neuroimaging allow diagnosing CAA in the absence of pathological examination. Current imaging-based criteria have high diagnostic performance in patients presenting with ICH, but is more limited in other clinical context such as cognitively impaired patients or asymptomatic individuals. Further research is still needed to improve diagnostic accuracy.
Collapse
Affiliation(s)
- Nicolas Raposo
- Department of neurology, Toulouse University Hospital
- Clinical Investigation Center, CIC1436, Toulouse University Hospital, F-CRIN/Strokelink Network, Toulouse
- Toulouse NeuroImaging Center, University of Toulouse, Inserm, UPS, France
| | - Charlotte Périole
- Department of neurology, Toulouse University Hospital
- Clinical Investigation Center, CIC1436, Toulouse University Hospital, F-CRIN/Strokelink Network, Toulouse
| | - Mélanie Planton
- Department of neurology, Toulouse University Hospital
- Toulouse NeuroImaging Center, University of Toulouse, Inserm, UPS, France
| |
Collapse
|
10
|
Helven C, Burel J, Vannier M, Maltête D, Ozkul-Wermester O, Hermary C, Wallon D, Grangeon L. Impact of previous statin use on first intracerebral hemorrhage in cerebral amyloid angiopathy. Rev Neurol (Paris) 2023; 179:1074-1080. [PMID: 37598087 DOI: 10.1016/j.neurol.2023.02.071] [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: 10/23/2022] [Revised: 01/29/2023] [Accepted: 02/25/2023] [Indexed: 08/21/2023]
Abstract
OBJECTIVES Statins have been associated with an increased risk of spontaneous intracerebral hemorrhage (ICH), but without dedicated study in cerebral amyloid angiopathy (CAA). We aimed to evaluate the association between previous statin treatment and radiological hemorrhagic lesions in a CAA population during a first lobar ICH event. MATERIALS AND METHODS We retrospectively included all patients meeting the modified Boston criteria for probable CAA and admitted for a first lobar ICH between 2010 and 2021 at Rouen University Hospital. Patients were classified as having previous statin treatment or not. We compared the ICH volume, the number of associated cerebral microbleeds (CMBs), and cortical superficial siderosis (CSS) according to previous statin treatment or not. We also compared functional outcomes and ICH recurrence during the follow-up period between the two groups. RESULTS We included 99 patients, 27 of whom had statin treatment prior to their ICH. The ICH volume and the number of CMBs did not differ between groups. Disseminated CSS was initially more frequent in the statin group (88% versus 57%; P=0.019), but this was no longer significant after adjustment for antiplatelet treatment (P=0.13). The long-term outcome was similar between the two groups with no increased risk of ICH recurrence in the statin-treated group (29.63% versus 23.61%, P=0.54). CONCLUSIONS Previous statin treatment was not associated with more severe hemorrhagic lesions in CAA in terms of ICH volume or number of microbleeds, but a trend for increased disseminated CSS was highlighted, which will require further larger studies.
Collapse
Affiliation(s)
- C Helven
- Department of Neurology, Rouen University Hospital, 76000 Rouen, France.
| | - J Burel
- Department of Radiology, Rouen University Hospital, 76000 Rouen, France
| | - M Vannier
- Department of Biostatistics, University of Rouen, Rouen University Hospital, 76000 Rouen, France
| | - D Maltête
- Department of Neurology, Rouen University Hospital, 76000 Rouen, France
| | - O Ozkul-Wermester
- Department of Neurology, Rouen University Hospital, 76000 Rouen, France
| | - C Hermary
- Department of Radiology, Rouen University Hospital, 76000 Rouen, France
| | - D Wallon
- Department of Neurology, Rouen University Hospital, 76000 Rouen, France; Inserm U1245, CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Department of Neurology, CHU of Rouen, University of Rouen Normandie, 76000 Rouen, France
| | - L Grangeon
- Department of Neurology, Rouen University Hospital, 76000 Rouen, France; Inserm U1245, CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Department of Neurology, CHU of Rouen, University of Rouen Normandie, 76000 Rouen, France
| |
Collapse
|
11
|
Chan E, Bonifacio GB, Harrison C, Banerjee G, Best JG, Sacks B, Harding N, Del Rocio Hidalgo Mas M, Jäger HR, Cipolotti L, Werring DJ. Domain-specific neuropsychological investigation of CAA with and without intracerebral haemorrhage. J Neurol 2023; 270:6124-6132. [PMID: 37672105 PMCID: PMC10632296 DOI: 10.1007/s00415-023-11977-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is associated with cognitive impairment, but the contributions of lobar intracerebral haemorrhage (ICH), underlying diffuse vasculopathy, and neurodegeneration, remain uncertain. We investigated the domain-specific neuropsychological profile of CAA with and without ICH, and their associations with structural neuroimaging features. METHODS Data were collected from patients with possible or probable CAA attending a specialist outpatient clinic. Patients completed standardised neuropsychological assessment covering seven domains. MRI scans were scored for markers of cerebral small vessel disease and neurodegeneration. Patients were grouped into those with and without a macro-haemorrhage (CAA-ICH and CAA-non-ICH). RESULTS We included 77 participants (mean age 72, 65% male). 26/32 (81%) CAA-non-ICH patients and 41/45 (91%) CAA-ICH patients were impaired in at least one cognitive domain. Verbal IQ and non-verbal IQ were the most frequently impaired, followed by executive functions and processing speed. We found no significant differences in the frequency of impairment across domains between the two groups. Medial temporal atrophy was the imaging feature most consistently associated with cognitive impairment (both overall and in individual domains) in both univariable and multivariable analyses. DISCUSSION Cognitive impairment is common in CAA, even in the absence of ICH, suggesting a key role for diffuse processes related to small vessel disease and/or neurodegeneration. Our findings indicate that neurodegeneration, possibly due to co-existing Alzheimer's disease pathology, may be the most important contributor. The observation that general intelligence is the most frequently affected domain suggests that CAA has a generalised rather than focal cognitive impact.
Collapse
Affiliation(s)
- Edgar Chan
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK.
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK.
| | - Guendalina B Bonifacio
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Corin Harrison
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Gargi Banerjee
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan G Best
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Benjamin Sacks
- Comprehensive Stroke Service, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Nicola Harding
- Comprehensive Stroke Service, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Maria Del Rocio Hidalgo Mas
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - H Rolf Jäger
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Lisa Cipolotti
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - David J Werring
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
12
|
Raber J, Silbert LC. Role of white matter hyperintensity in effects of apolipoprotein E on cognitive injury. Front Hum Neurosci 2023; 17:1176690. [PMID: 37275347 PMCID: PMC10237322 DOI: 10.3389/fnhum.2023.1176690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Magnetic Resonance Imaging (MRI) T2-weighted white matter hyperintensity (WMH) is a marker of small vessel cerebrovascular pathology and is of ischemic origin. The prevalence and severity of WMH is associated with cardiovascular risk factors, aging, and cognitive injury in mild cognitive impairment (MCI), vascular dementia, and Alzheimer's disease (AD). WMH especially affects executive function, with additional effects on memory and global cognition. Apolipoprotein E (apoE) plays a role in cholesterol metabolism and neuronal repair after injury. Human and animal studies support a role for apoE in maintaining white matter integrity. In humans, there are three major human apoE isoforms, E2, E3, and E4. Human apoE isoforms differ in risk to develop AD and in association with WMH. In this Mini Review, we propose an increased focus on the role of WMH in cognitive health and cognitive injury and the likely role of apoE and apoE isoform in modulating these effects. We hypothesize that apoE and apoE isoforms play a role in modulating WMH via apoE isoform-dependent effects on oxylipins and 7-ketocholesterol, as well as amyloid related vascular injury, as seen in cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Lisa C. Silbert
- Department of Neurology, Oregon Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Veterans Affairs Portland Health Care System, Portland, OR, United States
| |
Collapse
|
13
|
Feng X, Li X, Feng J, Xia J. Intracranial hemorrhage management in the multi-omics era. Heliyon 2023; 9:e14749. [PMID: 37101482 PMCID: PMC10123201 DOI: 10.1016/j.heliyon.2023.e14749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Intracranial hemorrhage (ICH) is a devastating disorder. Neuroprotective strategies that prevent tissue injury and improve functional outcomes have been identified in multiple animal models of ICH. However, these potential interventions in clinical trials produced generally disappointing results. With progress in omics, studies of omics data, including genomics, transcriptomics, epigenetics, proteomics, metabolomics, and the gut microbiome, may help promote precision medicine. In this review, we focused on introducing the applications of all omics in ICH and shed light on all of the considerable advantages to systematically analyze the necessity and importance of multiple omics technology in ICH.
Collapse
Affiliation(s)
- Xianjing Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Corresponding author. Department of Neurology, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, China
| |
Collapse
|
14
|
Jang H, Chun MY, Kim HJ, Na DL, Seo SW. The effects of imaging markers on clinical trajectory in cerebral amyloid angiopathy: a longitudinal study in a memory clinic. Alzheimers Res Ther 2023; 15:14. [PMID: 36635759 PMCID: PMC9835259 DOI: 10.1186/s13195-023-01161-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND We investigated the relevance of various imaging markers for the clinical trajectory of cerebral amyloid angiopathy (CAA) patients in a memory clinic. METHODS A total of 226 patients with probable CAA were included in this study with a mean follow-up period of 3.5 ± 2.7 years. Although all had more than one follow-up visit, 173 underwent follow-up Mini-Mental Status Examination (MMSE) and Clinical Dementia Rating Sum of Boxes (CDR-SB) ranging from 2 to 15 time points. Among 226, 122 patients underwent amyloid-β (Aβ) PET imaging. The prevalence of intracerebral hemorrhage (ICH) and its imaging predictors was investigated. The effects of CAA imaging markers and Aβ PET positivity on longitudinal cognition based on the MMSE and CDR-SB were evaluated using mixed effects models. RESULTS During the follow-up, 10 (4.4%) patients developed ICH: cortical superficial siderosis (cSS; hazard ratio [HR], 6.45) and previous lobar ICH (HR, 4.9), but lobar cerebral microbleeds (CMBs) were not predictors of ICH development. The presence of CMIs (p = 0.045) and Aβ positivity (p = 0.002) were associated with worse MMSE trajectory in CAA patients. Regarding CDR-SB trajectory, only Aβ positivity was marginally associated with worse longitudinal change (p = 0.050). CONCLUSION The results of the present study indicated that various imaging markers in CAA patients have different clinical relevance and predictive values for further clinical courses.
Collapse
Affiliation(s)
- Hyemin Jang
- grid.414964.a0000 0001 0640 5613Samsung Alzheimer’s Convergence Research Center, Samsung Medical Center, Seoul, South Korea ,grid.264381.a0000 0001 2181 989XDepartments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351 South Korea ,grid.414964.a0000 0001 0640 5613Neuroscience Center, Samsung Medical Center, Seoul, South Korea ,grid.264381.a0000 0001 2181 989XDepartment of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Min Young Chun
- grid.264381.a0000 0001 2181 989XDepartments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351 South Korea ,grid.414964.a0000 0001 0640 5613Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Hee Jin Kim
- grid.264381.a0000 0001 2181 989XDepartments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351 South Korea ,grid.414964.a0000 0001 0640 5613Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Duk L. Na
- grid.264381.a0000 0001 2181 989XDepartments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351 South Korea ,Happymind Clinic, Seoul, South Korea
| | - Sang Won Seo
- grid.414964.a0000 0001 0640 5613Samsung Alzheimer’s Convergence Research Center, Samsung Medical Center, Seoul, South Korea ,grid.264381.a0000 0001 2181 989XDepartments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351 South Korea ,grid.414964.a0000 0001 0640 5613Neuroscience Center, Samsung Medical Center, Seoul, South Korea ,grid.264381.a0000 0001 2181 989XDepartment of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
15
|
Nagaraja N, Wang WE, Duara R, DeKosky ST, Vaillancourt D. Mediation of Reduced Hippocampal Volume by Cerebral Amyloid Angiopathy in Pathologically Confirmed Patients with Alzheimer's Disease. J Alzheimers Dis 2023; 93:495-507. [PMID: 37038809 DOI: 10.3233/jad-220624] [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: 04/07/2023]
Abstract
BACKGROUND Hippocampal atrophy in cerebral amyloid angiopathy (CAA) has been reported to be similar to that in Alzheimer's disease (AD). OBJECTIVE To evaluate if CAA pathology partly mediates reduced hippocampal volume in patients with AD. METHODS Patients with a clinical diagnosis of AD and neuropathological confirmation of AD+/-CAA in the National Alzheimer's Coordinating Center database were included in the study. The volumes of temporal lobe structures were calculated on T1-weighted imaging (T1-MRI) using automated FreeSurfer software, from images acquired on average 5 years prior to death. Multivariate regression analysis was performed to compare brain volumes in four CAA groups. The hippocampal volume on T1-MRI was correlated with Clinical Dementia Rating sum of boxes (CDRsb) score, apolipoprotein E (APOE) genotype, and hippocampal atrophy at autopsy. RESULTS The study included 231 patients with no (n = 45), mild (n = 70), moderate (n = 67), and severe (n = 49) CAA. Among the four CAA groups, patients with severe CAA had a smaller mean left hippocampal volume (p = 0.023) but this was not significant when adjusted for APOE ɛ4 (p = 0.07). The left hippocampal volume on MRI correlated significantly with the hippocampal atrophy grading on neuropathology (p = 0.0003). Among patients with severe CAA, the left hippocampal volume on T1-MRI: (a) decreased with an increase in the number of APOE ɛ4 alleles (p = 0.04); but (b) had no evidence of correlation with CDRsb score (p = 0.57). CONCLUSION Severe CAA was associated with smaller left hippocampal volume on T1-MRI up to five years prior to death among patients with neuropathologically confirmed AD. This relationship was dependent on APOE ɛ4 genotype.
Collapse
Affiliation(s)
- Nandakumar Nagaraja
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Wei-En Wang
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Ranjan Duara
- Department of Neurology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Steven T DeKosky
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David Vaillancourt
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
16
|
Bonaterra-Pastra A, Benítez S, Pancorbo O, Rodríguez-Luna D, Vert C, Rovira A, Freijo MM, Tur S, Martínez-Zabaleta M, Cardona Portela P, Vera R, Lebrato-Hernández L, Arenillas JF, Pérez-Sánchez S, Domínguez-Mayoral A, Fàbregas JM, Mauri G, Montaner J, Sánchez-Quesada JL, Hernández-Guillamon M. Association of candidate genetic variants and circulating levels of ApoE/ApoJ with common neuroimaging features of cerebral amyloid angiopathy. Front Aging Neurosci 2023; 15:1134399. [PMID: 37113571 PMCID: PMC10126235 DOI: 10.3389/fnagi.2023.1134399] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of amyloid-β (Aβ) in brain vessels and is a main cause of lobar intracerebral hemorrhage (ICH) in the elderly. CAA is associated with magnetic resonance imaging (MRI) markers of small vessel disease (SVD). Since Aβ is also accumulated in Alzheimer's disease (AD) in the brain parenchyma, we aimed to study if several single nucleotide polymorphisms (SNPs) previously associated with AD were also associated with CAA pathology. Furthermore, we also studied the influence of APOE and CLU genetic variants in apolipoprotein E (ApoE) and clusterin/apolipoprotein J (ApoJ) circulating levels and their distribution among lipoproteins. Methods The study was carried out in a multicentric cohort of 126 patients with lobar ICH and clinical suspicion of CAA. Results We observed several SNPs associated with CAA neuroimaging MRI markers [cortical superficial siderosis (cSS), enlarged perivascular spaces in the centrum semiovale (CSO-EPVS), lobar cerebral microbleeds (CMB), white matter hyperintensities (WMH), corticosubcortical atrophy and CAA-SVD burden score]. Concretely, ABCA7 (rs3764650), CLU (rs9331896 and rs933188), EPHA1 (rs11767557), and TREML2 (rs3747742) were significantly associated with a CAA-SVD burden score. Regarding circulating levels of apolipoproteins, protective AD SNPs of CLU [rs11136000 (T) and rs9331896 (C)] were significantly associated with higher HDL ApoJ content in the lobar ICH cohort. APOEε2 carriers presented higher plasma and LDL-associated ApoE levels whereas APOEε4 carriers presented lower plasma ApoE levels. Additionally, we observed that lower circulating ApoJ and ApoE levels were significantly associated with CAA-related MRI markers. More specifically, lower LDL-associated ApoJ and plasma and HDL-associated ApoE levels were significantly associated with CSO-EPVS, lower ApoJ content in HDL with brain atrophy and lower ApoE content in LDL with the extent of cSS. Discussion This study reinforces the relevance of lipid metabolism in CAA and cerebrovascular functionality. We propose that ApoJ and ApoE distribution among lipoproteins may be associated with pathological features related to CAA with higher ApoE and ApoJ levels in HDL possibly enhancing atheroprotective, antioxidative, and anti-inflammatory responses in cerebral β-amyloidosis.
Collapse
Affiliation(s)
- Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sònia Benítez
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
- Center for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Olalla Pancorbo
- Stroke Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | | | - Carla Vert
- Section of Neuroradiology, Department of Radiology, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Rovira
- Section of Neuroradiology, Department of Radiology, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M. Mar Freijo
- Neurovascular Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Silvia Tur
- Department of Neurology, Son Espases University Hospital, Balearic Islands, Spain
| | | | - Pere Cardona Portela
- Department of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Spain
| | - Rocío Vera
- Stroke Unit, Department of Neurology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Lucia Lebrato-Hernández
- Stroke Unit, Department of Neurology and Neurophysiology, Virgen del Rocío University Hospital, Seville, Spain
| | - Juan F. Arenillas
- Stroke Program, Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain
- Clinical Neurosciences Research Group, Department of Medicine, University of Valladolid, Valladolid, Spain
| | | | | | - Joan Martí Fàbregas
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Gerard Mauri
- Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova de Lleida, Lleida, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Stroke Research Program, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, Seville, Spain
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
- Center for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Jose Luis Sánchez-Quesada,
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- *Correspondence: Mar Hernández-Guillamon,
| |
Collapse
|
17
|
Zanon Zotin MC, Schoemaker D, Raposo N, Perosa V, Bretzner M, Sveikata L, Li Q, van Veluw SJ, Horn MJ, Etherton MR, Charidimou A, Gurol ME, Greenberg SM, Duering M, dos Santos AC, Pontes-Neto OM, Viswanathan A. Peak width of skeletonized mean diffusivity in cerebral amyloid angiopathy: Spatial signature, cognitive, and neuroimaging associations. Front Neurosci 2022; 16:1051038. [PMID: 36440281 PMCID: PMC9693722 DOI: 10.3389/fnins.2022.1051038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Peak width of skeletonized mean diffusivity (PSMD) is a promising diffusion tensor imaging (DTI) marker that shows consistent and strong cognitive associations in the context of different cerebral small vessel diseases (cSVD). Purpose Investigate whether PSMD (1) is higher in patients with Cerebral Amyloid Angiopathy (CAA) than those with arteriolosclerosis; (2) can capture the anteroposterior distribution of CAA-related abnormalities; (3) shows similar neuroimaging and cognitive associations in comparison to other classical DTI markers, such as average mean diffusivity (MD) and fractional anisotropy (FA). Materials and methods We analyzed cross-sectional neuroimaging and neuropsychological data from 90 non-demented memory-clinic subjects from a single center. Based on MRI findings, we classified them into probable-CAA (those that fulfilled the modified Boston criteria), subjects with MRI markers of cSVD not attributable to CAA (presumed arteriolosclerosis; cSVD), and subjects without evidence of cSVD on MRI (non-cSVD). We compared total and lobe-specific (frontal and occipital) DTI metrics values across the groups. We used linear regression models to investigate how PSMD, MD, and FA correlate with conventional neuroimaging markers of cSVD and cognitive scores in CAA. Results PSMD was comparable in probable-CAA (median 4.06 × 10–4 mm2/s) and cSVD (4.07 × 10–4 mm2/s) patients, but higher than in non-cSVD (3.30 × 10–4 mm2/s; p < 0.001) subjects. Occipital-frontal PSMD gradients were higher in probable-CAA patients, and we observed a significant interaction between diagnosis and region on PSMD values [F(2, 87) = 3.887, p = 0.024]. PSMD was mainly associated with white matter hyperintensity volume, whereas MD and FA were also associated with other markers, especially with the burden of perivascular spaces. PSMD correlated with worse executive function (β = −0.581, p < 0.001) and processing speed (β = −0.463, p = 0.003), explaining more variance than other MRI markers. MD and FA were not associated with performance in any cognitive domain. Conclusion PSMD is a promising biomarker of cognitive impairment in CAA that outperforms other conventional and DTI-based neuroimaging markers. Although global PSMD is similarly increased in different forms of cSVD, PSMD’s spatial variations could potentially provide insights into the predominant type of underlying microvascular pathology.
Collapse
Affiliation(s)
- Maria Clara Zanon Zotin
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Imaging Sciences and Medical Physics, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Maria Clara Zanon Zotin, ,
| | - Dorothee Schoemaker
- Department of Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Nicolas Raposo
- Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | | | - Martin Bretzner
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog (JPARC) - Lille Neurosciences & Cognition, Lille, France
| | - Lukas Sveikata
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Qi Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Susanne J. van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Mitchell J. Horn
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Mark R. Etherton
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Andreas Charidimou
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston University Medical Center, Boston, MA, United States
| | - M. Edip Gurol
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marco Duering
- Department of Biomedical Engineering, Medical Imaging Analysis Center (MIAC), University of Basel, Basel, Switzerland
| | - Antonio Carlos dos Santos
- Center for Imaging Sciences and Medical Physics, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Octavio M. Pontes-Neto
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Malhotra K, Theodorou A, Katsanos AH, Zompola C, Shoamanesh A, Boviatsis E, Paraskevas GP, Spilioti M, Cordonnier C, Werring DJ, Alexandrov AV, Tsivgoulis G. Prevalence of Clinical and Neuroimaging Markers in Cerebral Amyloid Angiopathy: A Systematic Review and Meta-Analysis. Stroke 2022; 53:1944-1953. [PMID: 35264008 DOI: 10.1161/strokeaha.121.035836] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Limited data exist regarding the prevalence of clinical and neuroimaging manifestations among patients diagnosed with cerebral amyloid angiopathy (CAA). We sought to determine the prevalence of clinical phenotypes and radiological markers in patients with CAA. METHODS Systematic review and meta-analysis of studies including patients with CAA was conducted to primarily assess the prevalence of clinical phenotypes and neuroimaging markers as available in the included studies. Sensitivity analyses were performed based on the (1) retrospective or prospective study design and (2) probable or unspecified CAA status. We pooled the prevalence rates using random-effects models and assessed the heterogeneity using the Cochran Q and I2 statistics. RESULTS We identified 12 prospective and 34 retrospective studies including 7159 patients with CAA. The pooled prevalence rates were cerebral microbleeds (52% [95% CI, 43%-60%]; I2=93%), cortical superficial siderosis (49% [95% CI, 38%-59%]; I2=95%), dementia or mild cognitive impairment (50% [95% CI, 35%-65%]; I2=97%), intracerebral hemorrhage (ICH; 44% [95% CI, 27%-61%]; I2=98%), transient focal neurological episodes (48%; 10 studies [95% CI, 29%-67%]; I2=97%), lacunar infarcts (30% [95% CI, 25%-36%]; I2=78%), high grades of perivascular spaces located in centrum semiovale (56% [95% CI, 44%-67%]; I2=88%) and basal ganglia (21% [95% CI, 2%-51%]; I2=98%), and white matter hyperintensities with moderate or severe Fazekas score (53% [95% CI, 40%-65%]; I2=91%). The only neuroimaging marker that was associated with higher odds of recurrent ICH was cortical superficial siderosis (odds ratio, 1.57 [95% CI, 1.01-2.46]; I2=47%). Sensitivity analyses demonstrated a higher prevalence of ICH (53% versus 16%; P=0.03) and transient focal neurological episodes (57% versus 17%; P=0.03) among retrospective studies compared with prospective studies. No difference was documented between the prevalence rates based on the CAA status. CONCLUSIONS Approximately one-half of hospital-based cohort of CAA patients was observed to have cerebral microbleeds, cortical superficial siderosis, mild cognitive impairment, dementia, ICH, or transient focal neurological episodes. Cortical superficial siderosis was the only neuroimaging marker that was associated with higher odds of ICH recurrence. Future population-based studies among well-defined CAA cohorts are warranted to corroborate our findings.
Collapse
Affiliation(s)
- Konark Malhotra
- Department of Neurology, Allegheny Health Network, Pittsburgh, PA (K.M.)
| | - Aikaterini Theodorou
- Second Department of Neurology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (A.T., A.H.K., C.Z., G.P.P., G.T.)
| | - Aristeidis H Katsanos
- Second Department of Neurology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (A.T., A.H.K., C.Z., G.P.P., G.T.).,Department of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada (A.H.K., A.S.)
| | - Christina Zompola
- Second Department of Neurology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (A.T., A.H.K., C.Z., G.P.P., G.T.)
| | - Ashkan Shoamanesh
- Department of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada (A.H.K., A.S.)
| | - Efstathios Boviatsis
- Department of Neurosurgery, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (E.B.)
| | - George P Paraskevas
- Second Department of Neurology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (A.T., A.H.K., C.Z., G.P.P., G.T.)
| | - Martha Spilioti
- First Department of Neurology, AHEPA General Hospital, Aristotle University of Thessaloniki, Greece (M.S.)
| | - Charlotte Cordonnier
- University Lille, Inserm, CHU Lille, U1172, LilNCog, Lille Neuroscience and Cognition, France (C.C.)
| | - David J Werring
- Stroke Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom (D.J.W.)
| | - Andrei V Alexandrov
- Department of Neurology, University of Tennessee Health Science Center, Memphis (A.V.A., G.T.)
| | - Georgios Tsivgoulis
- Second Department of Neurology, National and Kapodistrian University of Athens, "Attikon" University Hospital, Greece. (A.T., A.H.K., C.Z., G.P.P., G.T.).,Department of Neurology, University of Tennessee Health Science Center, Memphis (A.V.A., G.T.)
| |
Collapse
|
19
|
Jo S, Cheong EN, Kim N, Oh JS, Shim WH, Kim HJ, Lee SJ, Lee Y, Oh M, Kim JS, Kim BJ, Roh JH, Kim SJ, Lee JH. Role of White Matter Abnormalities in the Relationship Between Microbleed Burden and Cognitive Impairment in Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 86:667-678. [DOI: 10.3233/jad-215094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Cerebral amyloid angiopathy (CAA) often presents as cognitive impairment, but the mechanism of cognitive decline is unclear. Recent studies showed that number of microbleeds were associated with cognitive decline. Objective: We aimed to investigate how microbleeds contribute to cognitive impairment in association with white matter tract abnormalities or cortical thickness in CAA. Methods: This retrospective comparative study involved patients with probable CAA according to the Boston criteria (Aβ + CAA) and patients with Alzheimer’s disease (Aβ + AD), all of whom showed severe amyloid deposition on amyloid PET. Using mediation analysis, we investigated how FA or cortical thickness mediates the correlation between the number of lobar microbleeds and cognition. Results: We analyzed 30 patients with Aβ + CAA (age 72.2±7.6, female 53.3%) and 30 patients with Aβ + AD (age 71.5±7.6, female 53.3%). The two groups showed similar degrees of cortical amyloid deposition in AD-related regions. The Aβ + CAA group had significantly lower FA values in the clusters of the posterior area than did the Aβ + AD group (family-wise error-corrected p < 0.05). The correlation between the number of lobar microbleeds and visuospatial function was indirectly mediated by white matter tract abnormality of right posterior thalamic radiation (PTR) and tapetum, while lobar microbleeds and language function was indirectly mediated by the abnormality of left PTR and sagittal stratum. Cortical thickness did not mediate the association between lobar microbleeds and cognition. Conclusion: This result supports the hypothesis that microbleeds burden leads to white matter tract damage and subsequent cognitive decline in CAA.
Collapse
Affiliation(s)
- Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - E-Nae Cheong
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Nayoung Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jungsu S. Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Hyun Shim
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyung-Ji Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sun Ju Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoojin Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bum Joon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jee Hoon Roh
- Department of Physiology, Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sang Joon Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Moore BD, Levites Y, Xu G, Hampton H, Adamo MF, Croft CL, Futch HS, Moran C, Fromholt S, Janus C, Prokop S, Dickson D, Lewis J, Giasson BI, Golde TE, Borchelt DR. Soluble brain homogenates from diverse human and mouse sources preferentially seed diffuse Aβ plaque pathology when injected into newborn mouse hosts. FREE NEUROPATHOLOGY 2022; 3. [PMID: 35494163 DOI: 10.17879/freeneuropathology-2022-3766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Seeding of pathology related to Alzheimer's disease (AD) and Lewy body disease (LBD) by tissue homogenates or purified protein aggregates in various model systems has revealed prion-like properties of these disorders. Typically, these homogenates are injected into adult mice stereotaxically. Injection of brain lysates into newborn mice represents an alternative approach of delivering seeds that could direct the evolution of amyloid-β (Aβ) pathology co-mixed with either tau or α-synuclein (αSyn) pathology in susceptible mouse models. Methods Homogenates of human pre-frontal cortex were injected into the lateral ventricles of newborn (P0) mice expressing a mutant humanized amyloid precursor protein (APP), human P301L tau, human wild type αSyn, or combinations thereof. The homogenates were prepared from AD and AD/LBD cases displaying variable degrees of Aβ pathology and co-existing tau and αSyn deposits. Behavioral assessments of APP transgenic mice injected with AD brain lysates were conducted. For comparison, homogenates of aged APP transgenic mice that preferentially exhibit diffuse or cored deposits were similarly injected into the brains of newborn APP mice. Results We observed that lysates from the brains with AD (Aβ+, tau+), AD/LBD (Aβ+, tau+, αSyn+), or Pathological Aging (Aβ+, tau-, αSyn-) efficiently seeded diffuse Aβ deposits. Moderate seeding of cerebral amyloid angiopathy (CAA) was also observed. No animal of any genotype developed discernable tau or αSyn pathology. Performance in fear-conditioning cognitive tasks was not significantly altered in APP transgenic animals injected with AD brain lysates compared to nontransgenic controls. Homogenates prepared from aged APP transgenic mice with diffuse Aβ deposits induced similar deposits in APP host mice; whereas homogenates from APP mice with cored deposits induced similar cored deposits, albeit at a lower level. Conclusions These findings are consistent with the idea that diffuse Aβ pathology, which is a common feature of human AD, AD/LBD, and PA brains, may arise from a distinct strain of misfolded Aβ that is highly transmissible to newborn transgenic APP mice. Seeding of tau or αSyn comorbidities was inefficient in the models we used, indicating that additional methodological refinement will be needed to efficiently seed AD or AD/LBD mixed pathologies by injecting newborn mice.
Collapse
Affiliation(s)
- Brenda D Moore
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Yona Levites
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Guilian Xu
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Hailey Hampton
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Munir F Adamo
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Cara L Croft
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Hunter S Futch
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Corey Moran
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Susan Fromholt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Christopher Janus
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Pathology, University of Florida, Gainesville, FL 32610 USA.,Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Dennis Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jada Lewis
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd E Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Neurology, College of Medicine, University of Florida, Gainesville FL 32610, USA
| | - David R Borchelt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Michiels L, Dobbels L, Demeestere J, Demaerel P, Van Laere K, Lemmens R. Simplified Edinburgh and modified Boston criteria in relation to amyloid PET for lobar intracerebral hemorrhage. NEUROIMAGE: CLINICAL 2022; 35:103107. [PMID: 35853346 PMCID: PMC9421490 DOI: 10.1016/j.nicl.2022.103107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/28/2022] [Accepted: 07/10/2022] [Indexed: 11/26/2022] Open
Abstract
Amyloid PET was positive in 63% of patients with lobar ICH. Simplified Edinburgh criteria and amyloid PET have similar accuracy vs Boston criteria. Simplified Edinburgh and Boston criteria have similar accuracy vs amyloid PET. Amyloid PET could assist in diagnosing CAA.
Background Histopathological evidence of cerebral vascular amyloid β accumulation is the gold standard to diagnose cerebral amyloid angiopathy (CAA). Neuroimaging findings obtained with CT and MRI can suggest the presence of CAA when histopathology is lacking. We explored the role of amyloid PET in patients with lobar intracerebral hemorrhage (ICH) as this may provide molecular evidence for CAA as well. Methods In this retrospective, monocenter analysis, we included consecutive patients with non-traumatic lobar ICH who had undergone amyloid PET. We categorized patients according to amyloid PET status and compared demographics and neuroimaging findings. We calculated sensitivity and specificity of the simplified Edinburgh criteria and amyloid PET with probable modified Boston criteria as reference standard, as well as sensitivity and specificity of the simplified Edinburgh and modified Boston criteria with amyloid PET status as molecular marker for presence or absence of CAA. Results We included 38 patients of whom 24 (63%) were amyloid PET positive. Amyloid PET positive patients were older at presentation (p = 0.004). We observed no difference in prevalence of subarachnoid hemorrhages, fingerlike projections or microbleeds between both groups, but cortical superficial siderosis (p = 0.003) was more frequent in the amyloid PET positive group. In 5 out of 38 patients (13%), the modified Boston criteria were not fulfilled due to young age or concomitant vitamin K antagonist use with INR > 3.0. With the modified Boston criteria as reference standard, there was no difference in sensitivity nor specificity between the simplified Edinburgh criteria and amyloid PET status. With amyloid PET status as reference standard, there was also no difference in sensitivity nor specificity between the simplified Edinburgh and modified Boston criteria. Conclusions Amyloid PET was positive in 63% of lobar ICH patients. Under certain circumstances, patients might not be diagnosed with probable CAA according to the modified Boston criteria and in these cases, amyloid PET may be useful. Accuracy to predict CAA based on amyloid PET status did not differ between the simplified Edinburgh and modified Boston criteria.
Collapse
|
22
|
Relationship between Urinary AD7c-NTP with Cerebral Microbleeds Based on APOE Genotype. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3928060. [PMID: 34660786 PMCID: PMC8519669 DOI: 10.1155/2021/3928060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 06/27/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022]
Abstract
Objective This study was performed to investigate the association between urinary Alzheimer-associated neuronal thread protein (AD7c-NTP) with cerebral microbleeds (CMBs) based on the apolipoprotein E (APOE) genotypes. Methods A total of 471 patients with acute cerebral infarction screened by magnetic sensitive imaging were enrolled in this study. Among them, twenty-seven cases of mixed CMBs were excluded. A total of 444 patients were divided into two groups according to the presence or absence of CMBs: CMBs group (n = 92) and noncerebral microbleeds group (nCMBs) (n = 352). Urine AD7c-NTP levels were measured using a human enzyme immunoassay kit. Results In patients with lobar CMBs, there was an interaction between urine AD7c-NTP levels and APOE genotypes (p = 0.01). In patients with APOE ε3/ε3 allele, the odds ratio of lobar CMBs per standard deviation of urinary AD7c-NTP levels was 0.92 (95% CI: 0.70-1.19). In patients with APOE ε2+ or ε4+ allele, the multivariate-corrected odds ratio of lobar CMBs per standard deviation of urinary AD7c-NTP levels was 2.95 (95% CI: 1.38-6.27). Conclusion A higher level of urinary AD7c-NTP is involved in lobar CMBs, not deep CMBs.
Collapse
|
23
|
Haußmann R, Homeyer P, Donix M, Linn J. [Current findings on the coincidence of cerebral amyloid angiopathy and Alzheimer's disease]. DER NERVENARZT 2021; 93:605-611. [PMID: 34652483 PMCID: PMC9200677 DOI: 10.1007/s00115-021-01213-x] [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] [Accepted: 09/09/2021] [Indexed: 11/27/2022]
Abstract
Die zerebrale Amyloidangiopathie (CAA) tritt trotz verschiedener Pathomechanismen häufig koinzident zur Alzheimer-Demenz auf. Sie moduliert kognitive Defizite im Rahmen der Alzheimer-Erkrankung (AD) annehmbar durch additive Effekte, auch wenn die diesbezüglichen Zusammenhänge komplex sind. Die pathophysiologische Gemeinsamkeit beider Erkrankungen besteht in einem gestörten Amyloidmetabolismus, distinkt ist jedoch die pathologische Prozessierung von Amyloidvorläuferproteinen. Die CAA mit ihren verschiedenen Subtypen ist eine pathomechanistisch heterogene Gefäßerkrankung des Gehirns. Vaskuläre und parenchymatöse Amyloidablagerungen kommen gemeinsam, aber auch isoliert und unabhängig voneinander vor. Um den spezifischen Beitrag der CAA zu kognitiven Defiziten im Rahmen der AD zu untersuchen, bedarf es daher geeigneter diagnostischer Methoden, die der Komplexität der histopathologischen bzw. bildmorphologischen Charakteristika der CAA gerecht werden, sowie differenzierender testpsychometrischer Verfahren, anhand derer der Beitrag der CAA zu kognitiven Defiziten deskriptiv erfasst und damit ätiologisch besser zuordenbar wird.
Collapse
Affiliation(s)
- R Haußmann
- Universitäts DemenzCentrum (UDC), Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland.
| | - P Homeyer
- Universitäts DemenzCentrum (UDC), Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland
| | - M Donix
- Universitäts DemenzCentrum (UDC), Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland.,DZNE, Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Deutschland
| | - J Linn
- Institut und Poliklinik für diagnostische und interventionelle Neuroradiologie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland
| |
Collapse
|
24
|
Shoamanesh A, Akoudad S, Himali JJ, Beiser AS, DeCarli C, Seshadri S, Ikram MA, Romero JR, Vernooij MW. Cortical superficial siderosis in the general population: The Framingham Heart and Rotterdam studies. Int J Stroke 2021; 16:798-808. [PMID: 33478376 PMCID: PMC9822782 DOI: 10.1177/1747493020984559] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE We aimed to characterize cortical superficial siderosis, its determinants and sequel, in community-dwelling older adults. METHODS The sample consisted of Framingham (n = 1724; 2000-2009) and Rotterdam (n = 4325; 2005-2013) study participants who underwent brain MRI. In pooled individual-level analysis, we compared baseline characteristics in patients with cortical superficial siderosis to two reference groups: (i) persons without hemorrhagic MRI markers of cerebral amyloid angiopathy (no cortical superficial siderosis and no microbleeds) and (ii) those with presumed cerebral amyloid angiopathy based on the presence of strictly lobar microbleeds but without cortical superficial siderosis. RESULTS Among a total of 6049 participants, 4846 did not have any microbleeds or cortical superficial siderosis (80%), 401 had deep/mixed microbleeds (6.6%), 776 had strictly lobar microbleeds without cortical superficial siderosis (12.8%) and 26 had cortical superficial siderosis with/without microbleeds (0.43%). In comparison to participants without microbleeds or cortical superficial siderosis and to those with strictly lobar microbleeds but without cortical superficial siderosis, participants with cortical superficial siderosis were older (OR 1.09 per year, 95% CI 1.05, 1.14; p < 0.001 and 1.04, 95% CI 1.00, 1.09; p = 0.058, respectively), had overrepresentation of the APOE ɛ4 allele (5.19, 2.04, 13.25; p = 0.001 and 3.47, 1.35, 8.92; p = 0.01), and greater prevalence of intracerebral hemorrhage (72.57, 9.12, 577.49; p < 0.001 and 81.49, 3.40, >999.99; p = 0.006). During a mean follow-up of 5.6 years, 42.4% participants with cortical superficial siderosis had a stroke (five intracerebral hemorrhage, two ischemic strokes and four undetermined strokes), 19.2% had transient neurological deficits and 3.8% developed incident dementia. CONCLUSION Our study adds supporting evidence to the association between cortical superficial siderosis and cerebral amyloid angiopathy within the general population. Community-dwelling persons with cortical superficial siderosis may be at high risk for intracerebral hemorrhage and future neurological events.
Collapse
Affiliation(s)
- Ashkan Shoamanesh
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada,Boston University School of Medicine, Boston, MA, USA
| | | | - Jayandra J. Himali
- Boston University School of Medicine, Boston, MA, USA,The Framingham Heart Study, Framingham, MA, USA,Boston University School of Public Health, Boston, MA, USA,Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Alexa S. Beiser
- Boston University School of Medicine, Boston, MA, USA,The Framingham Heart Study, Framingham, MA, USA,Boston University School of Public Health, Boston, MA, USA
| | - Charles DeCarli
- Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Sudha Seshadri
- Boston University School of Medicine, Boston, MA, USA,The Framingham Heart Study, Framingham, MA, USA,Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | | | - Jose R Romero
- Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
25
|
Kozberg MG, Perosa V, Gurol ME, van Veluw SJ. A practical approach to the management of cerebral amyloid angiopathy. Int J Stroke 2021; 16:356-369. [PMID: 33252026 PMCID: PMC9097498 DOI: 10.1177/1747493020974464] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cerebral amyloid angiopathy is a common small vessel disease in the elderly involving vascular amyloid-β deposition. Cerebral amyloid angiopathy is one of the leading causes of intracerebral hemorrhage and a significant contributor to age-related cognitive decline. The awareness of a diagnosis of cerebral amyloid angiopathy is important in clinical practice as it impacts decisions to use lifelong anticoagulation or nonpharmacological alternatives to anticoagulation such as left atrial appendage closure in patients who have concurrent atrial fibrillation, another common condition in older adults. This review summarizes the latest literature regarding the management of patients with sporadic cerebral amyloid angiopathy, including diagnostic criteria, imaging biomarkers for cerebral amyloid angiopathy severity, and management strategies to decrease intracerebral hemorrhage risk. In a minority of patients, the presence of cerebral amyloid angiopathy triggers an autoimmune inflammatory reaction, referred to as cerebral amyloid angiopathy-related inflammation, which is often responsive to immunosuppressive treatment in the acute phase. Diagnosis and management of cerebral amyloid angiopathy-related inflammation will be presented separately. While there are currently no effective therapeutics available to cure or halt the progression of cerebral amyloid angiopathy, we discuss emerging avenues for potential future interventions.
Collapse
Affiliation(s)
- Mariel G Kozberg
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Valentina Perosa
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - M Edip Gurol
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| |
Collapse
|
26
|
Inoue Y, Ando Y, Misumi Y, Ueda M. Current Management and Therapeutic Strategies for Cerebral Amyloid Angiopathy. Int J Mol Sci 2021; 22:ijms22083869. [PMID: 33918041 PMCID: PMC8068954 DOI: 10.3390/ijms22083869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by accumulation of amyloid β (Aβ) in walls of leptomeningeal vessels and cortical capillaries in the brain. The loss of integrity of these vessels caused by cerebrovascular Aβ deposits results in fragile vessels and lobar intracerebral hemorrhages. CAA also manifests with progressive cognitive impairment or transient focal neurological symptoms. Although development of therapeutics for CAA is urgently needed, the pathogenesis of CAA remains to be fully elucidated. In this review, we summarize the epidemiology, pathology, clinical and radiological features, and perspectives for future research directions in CAA therapeutics. Recent advances in mass spectrometric methodology combined with vascular isolation techniques have aided understanding of the cerebrovascular proteome. In this paper, we describe several potential key CAA-associated molecules that have been identified by proteomic analyses (apolipoprotein E, clusterin, SRPX1 (sushi repeat-containing protein X-linked 1), TIMP3 (tissue inhibitor of metalloproteinases 3), and HTRA1 (HtrA serine peptidase 1)), and their pivotal roles in Aβ cytotoxicity, Aβ fibril formation, and vessel wall remodeling. Understanding the interactions between cerebrovascular Aβ deposits and molecules that accumulate with Aβ may lead to discovery of effective CAA therapeutics and to the identification of biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (M.U.)
- Correspondence: ; Tel.: +81-96-373-5893; Fax: +81-96-373-5895
| | - Yukio Ando
- Department of Amyloidosis Research, Nagasaki International University, Sasebo 859-3298, Japan;
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (M.U.)
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (M.U.)
| |
Collapse
|
27
|
Sporns PB, Psychogios MN, Boulouis G, Charidimou A, Li Q, Fainardi E, Dowlatshahi D, Goldstein JN, Morotti A. Neuroimaging of Acute Intracerebral Hemorrhage. J Clin Med 2021; 10:1086. [PMID: 33807843 PMCID: PMC7962049 DOI: 10.3390/jcm10051086] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/16/2021] [Accepted: 03/02/2021] [Indexed: 01/25/2023] Open
Abstract
Intracerebral hemorrhage (ICH) accounts for 10% to 20% of all strokes worldwide and is associated with high morbidity and mortality. Neuroimaging is clinically important for the rapid diagnosis of ICH and underlying etiologies, but also for identification of ICH expansion, often as-sociated with an increased risk for poor outcome. In this context, rapid assessment of early hema-toma expansion risk is both an opportunity for therapeutic intervention and a potential hazard for hematoma evacuation surgery. In this review, we provide an overview of the current literature surrounding the use of multimodal neuroimaging of ICH for etiological diagnosis, prediction of early hematoma expansion, and prognostication of neurological outcome. Specifically, we discuss standard imaging using computed tomography, the value of different vascular imaging modalities to identify underlying causes and present recent advances in magnetic resonance imaging and computed tomography perfusion.
Collapse
Affiliation(s)
- Peter B. Sporns
- Department of Neuroradiology, Clinic for Radiology & Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marios-Nikos Psychogios
- Department of Neuroradiology, Clinic for Radiology & Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland;
| | - Grégoire Boulouis
- Neuroradiology Department, University Hospital of Tours, CEDEX 09, 37044 Tours, France;
| | - Andreas Charidimou
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
- Department of Neurology, Boston University School of Medicine, Boston Medical Centre, Boston, MA 02118, USA
| | - Qi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 40016, China;
| | - Enrico Fainardi
- Section of Neuroradiology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy;
| | - Dar Dowlatshahi
- Department of Medicine (Neurology), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Joshua N. Goldstein
- Department of Emergency Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Andrea Morotti
- ASST Valcamonica, UOSD Neurology, Esine (BS), 25040 Brescia, Italy;
| |
Collapse
|
28
|
Raposo N, Zanon Zotin MC, Schoemaker D, Xiong L, Fotiadis P, Charidimou A, Pasi M, Boulouis G, Schwab K, Schirmer MD, Etherton MR, Gurol ME, Greenberg SM, Duering M, Viswanathan A. Peak Width of Skeletonized Mean Diffusivity as Neuroimaging Biomarker in Cerebral Amyloid Angiopathy. AJNR Am J Neuroradiol 2021; 42:875-881. [PMID: 33664113 DOI: 10.3174/ajnr.a7042] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE Whole-brain network connectivity has been shown to be a useful biomarker of cerebral amyloid angiopathy and related cognitive impairment. We evaluated an automated DTI-based method, peak width of skeletonized mean diffusivity, in cerebral amyloid angiopathy, together with its association with conventional MRI markers and cognitive functions. MATERIALS AND METHODS We included 24 subjects (mean age, 74.7 [SD, 6.0] years) with probable cerebral amyloid angiopathy and mild cognitive impairment and 62 patients with MCI not attributable to cerebral amyloid angiopathy (non-cerebral amyloid angiopathy-mild cognitive impairment). We compared peak width of skeletonized mean diffusivity between subjects with cerebral amyloid angiopathy-mild cognitive impairment and non-cerebral amyloid angiopathy-mild cognitive impairment and explored its associations with cognitive functions and conventional markers of cerebral small-vessel disease, using linear regression models. RESULTS Subjects with Cerebral amyloid angiopathy-mild cognitive impairment showed increased peak width of skeletonized mean diffusivity in comparison to those with non-cerebral amyloid angiopathy-mild cognitive impairment (P < .001). Peak width of skeletonized mean diffusivity values were correlated with the volume of white matter hyperintensities in both groups. Higher peak width of skeletonized mean diffusivity was associated with worse performance in processing speed among patients with cerebral amyloid angiopathy, after adjusting for other MRI markers of cerebral small vessel disease. The peak width of skeletonized mean diffusivity did not correlate with cognitive functions among those with non-cerebral amyloid angiopathy-mild cognitive impairment. CONCLUSIONS Peak width of skeletonized mean diffusivity is altered in cerebral amyloid angiopathy and is associated with performance in processing speed. This DTI-based method may reflect the degree of white matter structural disruption in cerebral amyloid angiopathy and could be a useful biomarker for cognition in this population.
Collapse
Affiliation(s)
- N Raposo
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts .,Department of Neurology (N.R.), Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Toulouse NeuroImaging Center (N.R.), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Toulouse, UPS, France
| | - M C Zanon Zotin
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Center for Imaging Sciences and Medical Physics (M.C.Z.Z.). Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil;, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - D Schoemaker
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - L Xiong
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - P Fotiadis
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - A Charidimou
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - M Pasi
- Department of Neurology (M.P.), Centre Hospitalier Universitaire de Lille, Lille, France
| | - G Boulouis
- Department of Neuroradiology (G.B.), Centre Hospitalier Sainte-Anne, Université Paris-Descartes, Paris, France
| | - K Schwab
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - M D Schirmer
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Computer Science and Artificial Intelligence Lab (M.D.S.), Massachusetts Institute of Technology, Boston, Massachusetts.,Department of Population Health Sciences (M.D.S.), German Center for Neurodegenerative Diseases, Bonn, Germany
| | - M R Etherton
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - M E Gurol
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - S M Greenberg
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - M Duering
- Medical Image Analysis Center and Quantitative Biomedical Imaging Group (M.D.), Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - A Viswanathan
- From the Stroke Research Center (N.R., M.C.Z.Z., D.S., L.X., P.F., A.C., K.S., M.D.S., M.R.E., M.E.G., S.M.G., A.V.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Alber J, Arthur E, Goldfarb D, Drake J, Boxerman JL, Silver B, Ott BR, Johnson LN, Snyder PJ. The relationship between cerebral and retinal microbleeds in cerebral amyloid angiopathy (CAA): A pilot study. J Neurol Sci 2021; 423:117383. [PMID: 33684655 DOI: 10.1016/j.jns.2021.117383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 01/26/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND The standard in vivo diagnostic imaging technique for cerebral amyloid angiopathy (CAA) is costly and thereby of limited utility for point-of-care diagnosis and monitoring of treatment efficacy. Recent recognition that retinal changes may reflect cerebral changes in neurodegenerative disease provides an ideal opportunity for development of accessible and cost-effective biomarkers for point-of-care use in the detection and monitoring of CAA. In this pilot study, we examined structural and angiographic retinal changes in CAA patients relative to a control group, and compared retinal and cerebral pathology in a group of CAA patients. METHODS We used spectral domain optical coherence tomography (SD-OCT) to image the retina and compared retinal microbleeds to both cerebral microbleeds and white matter hyperintensities (WMH) in CAA patients, as seen on MRI. We compared retinal angiographic changes, along with structural retinal neuronal layer changes in CAA patients and cognitively normal older adults, and examined the relationship between retinal and cerebral microbleeds and cognition in CAA patients. RESULTS We found a trend level correlation between retinal and cerebral microbleeds in CAA patients. Moreover, we found a significant correlation between retinal microbleeds and episodic memory performance in CAA patients. There were no significant group differences between CAA patients and cognitively normal older adults on retinal angiographic or structural measurements. CONCLUSION Retinal microbleeds may reflect degree of cerebral microbleed burden in CAA. This picture was complicated by systolic hypertension in the CAA group, which is a confounding factor for the interpretation of these data. Our results stimulate motivation for pursuit of a more comprehensive prospective study to determine the feasibility of retinal biomarkers in CAA.
Collapse
Affiliation(s)
- Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA; Memory & Aging Program, Butler Hospital, Providence, RI, USA.
| | - Edmund Arthur
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA; Memory & Aging Program, Butler Hospital, Providence, RI, USA
| | | | - Jonathan Drake
- Department of Neurology, Rhode Island Hospital, Providence, RI, USA; Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA
| | - Jerrold L Boxerman
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA; Department of Diagnostic Imaging, Rhode Island Hospital, Providence, RI, USA
| | - Brian Silver
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Brian R Ott
- Department of Neurology, Rhode Island Hospital, Providence, RI, USA; Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA
| | - Lenworth N Johnson
- Department of Surgery (Ophthalmology), Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter J Snyder
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA; Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA; Department of Surgery (Ophthalmology), Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
30
|
Graff-Radford J, Lesnick TG, Mielke MM, Constantopoulos E, Rabinstein A, Przybelski SA, Vemuri P, Botha H, Jones DT, Ramanan VK, Petersen RC, Knopman DS, Boeve BF, Murray ME, Dickson DW, Jack CR, Kantarci K, Reichard RR. Cerebral Amyloid Angiopathy Burden and Cerebral Microbleeds: Pathological Evidence for Distinct Phenotypes. J Alzheimers Dis 2021; 81:113-122. [PMID: 33720897 PMCID: PMC8113155 DOI: 10.3233/jad-201536] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The relationship between cerebral microbleeds (CMBs) on hemosiderin-sensitive MRI sequences and cerebral amyloid angiopathy (CAA) remains unclear in population-based participants or in individuals with dementia. OBJECTIVE To determine whether CMBs on antemortem MRI correlate with CAA. METHODS We reviewed 54 consecutive participants with antemortem T2*GRE-MRI sequences and subsequent autopsy. CMBs were quantified on MRIs closest to death. Autopsy CAA burden was quantified in each region including leptomeningeal/cortical and capillary CAA. By a clustering approach, we examined the relationship among CAA variables and performed principal component analysis (PCA) for dimension reduction to produce two scores from these 15 interrelated predictors. Hurdle models assessed relationships between principal components and lobar CMBs. RESULTS MRI-based CMBs appeared in 20/54 (37%). 10 participants had ≥2 lobar-only CMBs. The first two components of the PCA analysis of the CAA variables explained 74% variability. The first rotated component (RPC1) consisted of leptomeningeal and cortical CAA and the second rotated component of capillary CAA (RPC2). Both the leptomeningeal and cortical component and the capillary component correlated with lobar-only CMBs. The capillary CAA component outperformed the leptomeningeal and cortical CAA component in predicting lobar CMBs. Both capillary and the leptomeningeal/cortical components correlated with number of lobar CMBs. CONCLUSION Capillary and leptomeningeal/cortical scores correlated with lobar CMBs on MRI but lobar CMBs were more closely associated with the capillary component. The capillary component correlated with APOEɛ4, highlighting lobar CMBs as one aspect of CAA phenotypic diversity. More CMBs also increase the probability of underlying CAA.
Collapse
Affiliation(s)
| | | | - Michelle M. Mielke
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
- Department of Health Sciences Research
| | | | | | | | | | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Melissa E. Murray
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, Florida
| | - Dennis W. Dickson
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, Florida
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - R. Ross Reichard
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
31
|
Theodorou A, Tsantzali I, Kapaki E, Constantinides VC, Voumvourakis K, Tsivgoulis G, Paraskevas GP. Cerebrospinal fluid biomarkers and apolipoprotein E genotype in cerebral amyloid angiopathy. A narrative review. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100010. [PMID: 36324707 PMCID: PMC9616386 DOI: 10.1016/j.cccb.2021.100010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 11/26/2022]
Abstract
Lower Cerebrospinal Fluid Levels of Aβ40 and Aβ42 in Cerebral Amyloid Angiopathy compared with Alzheimer Disease and Healthy Controls. Cortical Superficial Siderosis-extent and lower levels of Aβ42 could be prognostic for the severity of Cerebral Amyloid Angiopathy. Apolipoprotein – E Genotype plays an important role in the pathophysiology of Cerebral Amyloid Angiopathy.
Sporadic cerebral amyloid angiopathy (CAA) is a cerebral small vessel disease, characterized by the deposition of β-amyloid within the cortical and leptomeningeal blood vessel walls. It has attracted interest concerning new therapeutic perspectives. However, there are scarce data regarding the cerebrospinal fluid biomarkers (CSF) and genetic factors in sporadic CAA. In this narrative review, we investigated the literature regarding the cerebrospinal fluid core biomarkers profile of patients with probable or possible CAA and its subtype, the CAA- related inflammation (CAA-ri), taking into account the clinical and radiological characteristics of the patients. We also analyzed the Apolipoprotein E (APOE) genotype differentiations among the different subtypes of cerebral amyloid angiopathy. Our results demonstrate specific CSF patterns of β-amyloid (Aβ42 and Aβ40) and tau-proteins (t-tau and p-tau) which may serve as molecular biomarkers for CAA/ CAA-ri and could prove helpful for novel therapeutic procedures. Specifically, decreased levels of Aβ40 and Aβ42 in both CAA and CAA-ri, mildly increased concentrations of tau protein in patients with CAA-ri and a strong association between APOE ε4/ε4 genotype and CAA-ri are the main findings.
Collapse
|
32
|
Li Z, Shue F, Zhao N, Shinohara M, Bu G. APOE2: protective mechanism and therapeutic implications for Alzheimer's disease. Mol Neurodegener 2020; 15:63. [PMID: 33148290 PMCID: PMC7640652 DOI: 10.1186/s13024-020-00413-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023] Open
Abstract
Investigations of apolipoprotein E (APOE) gene, the major genetic risk modifier for Alzheimer's disease (AD), have yielded significant insights into the pathogenic mechanism. Among the three common coding variants, APOE*ε4 increases, whereas APOE*ε2 decreases the risk of late-onset AD compared with APOE*ε3. Despite increased understanding of the detrimental effect of APOE*ε4, it remains unclear how APOE*ε2 confers protection against AD. Accumulating evidence suggests that APOE*ε2 protects against AD through both amyloid-β (Aβ)-dependent and independent mechanisms. In addition, APOE*ε2 has been identified as a longevity gene, suggesting a systemic effect of APOE*ε2 on the aging process. However, APOE*ε2 is not entirely benign; APOE*ε2 carriers exhibit increased risk of certain cerebrovascular diseases and neurological disorders. Here, we review evidence from both human and animal studies demonstrating the protective effect of APOE*ε2 against AD and propose a working model depicting potential underlying mechanisms. Finally, we discuss potential therapeutic strategies designed to leverage the protective effect of APOE2 to treat AD.
Collapse
Affiliation(s)
- Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan.
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
33
|
Xiong L, Charidimou A, Pasi M, Boulouis G, Pongpitakmetha T, Schirmer MD, Singh S, Benson E, Gurol EM, Rosand J, Greenberg SM, Biffi A, Viswanathan A. Predictors for Late Post-Intracerebral Hemorrhage Dementia in Patients with Probable Cerebral Amyloid Angiopathy. J Alzheimers Dis 2020; 71:435-442. [PMID: 31403947 DOI: 10.3233/jad-190346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Cerebral amyloid angiopathy (CAA) accounts for the majority of lobar intracerebral hemorrhage (ICH); however, the risk factors for dementia conversion after ICH occurrence in CAA patients are unknown, especially in the long-term period after ICH. Therefore, we aimed to unravel the predictors for late post-ICH dementia (6 months after ICH event) in probable CAA patients. METHODS From a large consecutive MRI prospective cohort of spontaneous ICH (2006-2017), we identified probable CAA patients (modified Boston criteria) without dementia 6 months post-ICH. Cognitive outcome during follow-up was determined based on the information from standardized clinical visit notes. We used Cox regression analysis to investigate the association between baseline demographic characteristics, past medical history, MRI biomarkers, and late post-ICH dementia conversion (dementia occurred after 6 months). RESULTS Among 97 non-demented lobar ICH patients with probable CAA, 25 patients (25.8%) developed dementia during a median follow-up time of 2.5 years (IQR 1.5-3.8 years). Pre-existing mild cognitive impairment, increased white matter hyperintensities (WMH) burden, the presence of disseminated cortical superficial siderosis (cSS), and higher total small vessel disease score for CAA were all independent predictors for late dementia conversion. CONCLUSION In probable CAA patients presenting with lobar ICH, high WMH burden and presence of disseminated cSS are useful neuroimaging biomarkers for dementia risk stratification. These findings have implications for clinical practice and future trial design.
Collapse
Affiliation(s)
- Li Xiong
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Andreas Charidimou
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Marco Pasi
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Gregoire Boulouis
- Centre Hospitalier Sainte-Anne, Université Paris Descartes, Paris, France
| | - Thanakit Pongpitakmetha
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA.,Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Markus D Schirmer
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA.,Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Boston, MA, USA.,Department of Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Germany
| | - Sanjula Singh
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Emily Benson
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Edip M Gurol
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Alessandro Biffi
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Lee JS, Lee H, Park S, Choe Y, Park YH, Cheon BK, Hahn A, Ossenkoppele R, Kim HJ, Kim S, Yoo H, Jang H, Cho SH, Kim SJ, Kim JP, Jung YH, Park KC, DeCarli C, Weiner MW, Na DL, Seo SW. Association between APOE ε2 and Aβ burden in patients with Alzheimer- and vascular-type cognitive impairment. Neurology 2020; 95:e2354-e2365. [PMID: 32928967 DOI: 10.1212/wnl.0000000000010811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 06/03/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the association between APOE genotype and β-amyloid (Aβ) burden, as measured by PET in patients with subcortical vascular cognitive impairment (SVCI) and those with Alzheimer disease-related cognitive impairment (ADCI). METHODS This was a cross-sectional study of 310 patients with SVCI and 999 with ADCI. To evaluate the effects of APOE genotype or diagnostic group on Aβ positivity, we performed multivariate logistic regression analyses. Further distinctive underlying features of latent subgroups were examined by employing a latent class cluster analysis approach. RESULTS In comparison with ε3 homozygotes, in the ADCI group, ε2 carriers showed a lower frequency of Aβ positivity (odds ratio [OR] 0.43, 95% confidence interval [CI] 0.23-0.79), while in the SVCI group, ε2 carriers showed a higher frequency of Aβ positivity (OR 2.26, 95% CI 1.02-5.01). In particular, we observed an interaction effect of ε2 carrier status and diagnostic group on Aβ positivity (OR 5.12, 95% CI 1.93-13.56), in that relative to ε3 homozygotes, there were more Aβ-positive ε2 carriers in the SVCI group than in the ADCI group. We also identified latent subgroups of Aβ-positive APOE ε2 carriers with SVCI and Aβ-positive APOE ε4 carriers with ADCI. CONCLUSIONS Our findings suggest that APOE ε2 is distinctly associated with Aβ deposition in patients with SVCI and those with ADCI. Our findings further suggest that there is a distinctive subgroup of Aβ-positive APOE ε2 carriers with SVCI among patients with cognitive impairment.
Collapse
Affiliation(s)
- Jin San Lee
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA.
| | - Hyejoo Lee
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA.
| | - Seongbeom Park
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Yeongsim Choe
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Yu Hyun Park
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Bo Kyoung Cheon
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Alice Hahn
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Rik Ossenkoppele
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Hee Jin Kim
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Seonwoo Kim
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Heejin Yoo
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Hyemin Jang
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Soo Hyun Cho
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Seung Joo Kim
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Jun Pyo Kim
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Young Hee Jung
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Key-Chung Park
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Charles DeCarli
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Michael W Weiner
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Duk L Na
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA
| | - Sang Won Seo
- From the Department of Neurology (J.S.L., H.L., S.P., Y.C., Y.H.P., B.K.C., A.H., H.J.K., H.J., J.P.K., D.L.N., S.W.S.), Samsung Alzheimer Research Center (H.J.K., H.J., J.P.K., D.L.N., S.W.S.), and Statistics and Data Center (S.K., H.Y.), Samsung Medical Center; Department of Intelligent Precision Healthcare Convergence (S.W.S.), Sungkyunkwan University School of Medicine; Department of Health Sciences and Technology (S.W.S.), SAIHST, Sungkyunkwan University; Department of Neurology (J.S.L., K.-C.P.), Kyung Hee University College of Medicine, Kyung Hee University Hospital, Seoul, Korea; Department of Neurology and Alzheimer Center (R.O.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Department of Neurology (S.H.C.), Chonnam National University Medical School, Gwangju; Department of Neurology (S.J.K.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon; Department of Neurology (Y.H.J.), Myungji Hospital, Goyang, Korea; Department of Neurology and Center for Neuroscience (C.D.), University of California, Davis; Department of Medicine (M.W.W.), University of California; and Department of Veterans Affairs Medical Center (M.W.W.), Center for Imaging of Neurodegenerative Diseases, San Francisco, CA.
| |
Collapse
|
35
|
The Role of HDL and HDL Mimetic Peptides as Potential Therapeutics for Alzheimer's Disease. Biomolecules 2020; 10:biom10091276. [PMID: 32899606 PMCID: PMC7563116 DOI: 10.3390/biom10091276] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
The role of high-density lipoproteins (HDL) in the cardiovascular system has been extensively studied and the cardioprotective effects of HDL are well established. As HDL particles are formed both in the systemic circulation and in the central nervous system, the role of HDL and its associated apolipoproteins in the brain has attracted much research interest in recent years. Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and the leading cause of dementia worldwide, for which there currently exists no approved disease modifying treatment. Multiple lines of evidence, including a number of large-scale human clinical studies, have shown a robust connection between HDL levels and AD. Low levels of HDL are associated with increased risk and severity of AD, whereas high levels of HDL are correlated with superior cognitive function. Although the mechanisms underlying the protective effects of HDL in the brain are not fully understood, many of the functions of HDL, including reverse lipid/cholesterol transport, anti-inflammation/immune modulation, anti-oxidation, microvessel endothelial protection, and proteopathy modification, are thought to be critical for its beneficial effects. This review describes the current evidence for the role of HDL in AD and the potential of using small peptides mimicking HDL or its associated apolipoproteins (HDL-mimetic peptides) as therapeutics to treat AD.
Collapse
|
36
|
Banerjee G, Wilson D, Ambler G, Hostettler IC, Shakeshaft C, Cohen H, Yousry T, Al-Shahi Salman R, Lip GYH, Houlden H, Muir KW, Brown MM, Jäger HR, Werring DJ. Longer term stroke risk in intracerebral haemorrhage survivors. J Neurol Neurosurg Psychiatry 2020; 91:840-845. [PMID: 32554800 DOI: 10.1136/jnnp-2020-323079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/16/2020] [Accepted: 05/05/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To evaluate the influence of intracerebral haemorrhage (ICH) location on stroke outcomes. METHODS We included patients recruited to a UK hospital-based, multicentre observational study of adults with imaging confirmed spontaneous ICH. The outcomes of interest were occurrence of a cerebral ischaemic event (either stroke or transient ischaemic attack) or a further ICH following study entry. Haematoma location was classified as lobar or non-lobar. RESULTS All 1094 patients recruited to the CROMIS-2 (Clinical Relevance of Microbleeds in Stroke) ICH study were included (mean age 73.3 years; 57.4% male). There were 45 recurrent ICH events (absolute event rate (AER) 1.88 per 100 patient-years); 35 in patients presenting with lobar ICH (n=447, AER 3.77 per 100 patient-years); and 9 in patients presenting with non-lobar ICH (n=580, AER 0.69 per 100 patient-years). Multivariable Cox regression found that lobar ICH was associated with ICH recurrence (HR 8.96, 95% CI 3.36 to 23.87, p<0.0001); similar results were found in multivariable completing risk analyses. There were 70 cerebral ischaemic events (AER 2.93 per 100 patient-years); 29 in patients presenting with lobar ICH (AER 3.12 per 100 patient-years); and 39 in patients with non-lobar ICH (AER 2.97 per 100 patient-years). Multivariable Cox regression found no association with ICH location (HR 1.13, 95% CI 0.66 to 1.92, p = 0.659). Similar results were seen in completing risk analyses. CONCLUSIONS In ICH survivors, lobar ICH location was associated with a higher risk of recurrent ICH events than non-lobar ICH; ICH location did not influence risk of subsequent ischaemic events. TRIAL REGISTRATION NUMBER NCT02513316.
Collapse
Affiliation(s)
- Gargi Banerjee
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Duncan Wilson
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Gareth Ambler
- Department of Statistical Science, University College London, London, United Kingdom
| | - Isabel Charlotte Hostettler
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Clare Shakeshaft
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Hannah Cohen
- Haemostasis Research Unit, Department of Haematology, University College London, London, United Kingdom
| | - Tarek Yousry
- Lysholm Department of Neuroradiology and the Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | | | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Henry Houlden
- Department of Molecular Neuroscience, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Keith W Muir
- Institute of Neuroscience & Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Martin M Brown
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Hans Rolf Jäger
- Lysholm Department of Neuroradiology and the Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - David J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | | |
Collapse
|
37
|
Xia Y, Prokop S, Gorion KMM, Kim JD, Sorrentino ZA, Bell BM, Manaois AN, Chakrabarty P, Davies P, Giasson BI. Tau Ser208 phosphorylation promotes aggregation and reveals neuropathologic diversity in Alzheimer's disease and other tauopathies. Acta Neuropathol Commun 2020; 8:88. [PMID: 32571418 PMCID: PMC7310041 DOI: 10.1186/s40478-020-00967-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Tau protein abnormally aggregates in tauopathies, a diverse group of neurologic diseases that includes Alzheimer’s disease (AD). In early stages of disease, tau becomes hyperphosphorylated and mislocalized, which can contribute to its aggregation and toxicity. We demonstrate that tau phosphorylation at Ser208 (pSer208) promotes microtubule dysfunction and tau aggregation in cultured cells. Comparative assessment of the epitopes recognized by antibodies AT8, CP13, and 7F2 demonstrates that CP13 and 7F2 are specific for tau phosphorylation at Ser202 and Thr205, respectively, independently of the phosphorylation state of adjacent phosphorylation sites. Supporting the involvement of pSer208 in tau pathology, a novel monoclonal antibody 3G12 specific for tau phosphorylation at Ser208 revealed strong reactivity of tau inclusions in the brains of PS19 and rTg4510 transgenic mouse models of tauopathy. 3G12 also labelled neurofibrillary tangles in brains of patients with AD but revealed differential staining compared to CP13 and 7F2 for other types of tau pathologies such as in neuropil threads and neuritic plaques in AD, tufted astrocytes in progressive supranuclear palsy and astrocytic plaques in corticobasal degeneration. These results support the hypothesis that tau phosphorylation at Ser208 strongly contributes to unique types of tau aggregation and may be a reliable marker for the presence of mature neurofibrillary tangles.
Collapse
|
38
|
Biffi A, Urday S, Kubiszewski P, Gilkerson L, Sekar P, Rodriguez-Torres A, Bettin M, Charidimou A, Pasi M, Kourkoulis C, Schwab K, DiPucchio Z, Behymer T, Osborne J, Morgan M, Moomaw CJ, James ML, Greenberg SM, Viswanathan A, Gurol ME, Worrall BB, Testai FD, McCauley JL, Falcone GJ, Langefeld CD, Anderson CD, Kamel H, Woo D, Sheth KN, Rosand J. Combining Imaging and Genetics to Predict Recurrence of Anticoagulation-Associated Intracerebral Hemorrhage. Stroke 2020; 51:2153-2160. [PMID: 32517581 DOI: 10.1161/strokeaha.120.028310] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE For survivors of oral anticoagulation therapy (OAT)-associated intracerebral hemorrhage (OAT-ICH) who are at high risk for thromboembolism, the benefits of OAT resumption must be weighed against increased risk of recurrent hemorrhagic stroke. The ε2/ε4 alleles of the apolipoprotein E (APOE) gene, MRI-defined cortical superficial siderosis, and cerebral microbleeds are the most potent risk factors for recurrent ICH. We sought to determine whether combining MRI markers and APOE genotype could have clinical impact by identifying ICH survivors in whom the risks of OAT resumption are highest. METHODS Joint analysis of data from 2 longitudinal cohort studies of OAT-ICH survivors: (1) MGH-ICH study (Massachusetts General Hospital ICH) and (2) longitudinal component of the ERICH study (Ethnic/Racial Variations of Intracerebral Hemorrhage). We evaluated whether MRI markers and APOE genotype predict ICH recurrence. We then developed and validated a combined APOE-MRI classification scheme to predict ICH recurrence, using Classification and Regression Tree analysis. RESULTS Cortical superficial siderosis, cerebral microbleed, and APOE ε2/ε4 variants were independently associated with ICH recurrence after OAT-ICH (all P<0.05). Combining APOE genotype and MRI data resulted in improved prediction of ICH recurrence (Harrell C: 0.79 versus 0.55 for clinical data alone, P=0.033). In the MGH (training) data set, CSS, cerebral microbleed, and APOE ε2/ε4 stratified likelihood of ICH recurrence into high-, medium-, and low-risk categories. In the ERICH (validation) data set, yearly ICH recurrence rates for high-, medium-, and low-risk individuals were 6.6%, 2.5%, and 0.9%, respectively, with overall area under the curve of 0.91 for prediction of recurrent ICH. CONCLUSIONS Combining MRI and APOE genotype stratifies likelihood of ICH recurrence into high, medium, and low risk. If confirmed in prospective studies, this combined APOE-MRI classification scheme may prove useful for selecting individuals for OAT resumption after ICH.
Collapse
Affiliation(s)
- Alessandro Biffi
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Center for Genomic Medicine (A.B., P.K., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston.,Henry and Allison McCance Center for Brain Health (A.B., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston
| | - Sebastian Urday
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston
| | - Patryk Kubiszewski
- Center for Genomic Medicine (A.B., P.K., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston
| | - Lee Gilkerson
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Padmini Sekar
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Axana Rodriguez-Torres
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Margaret Bettin
- Department of Neurology, University of Virginia Health System, Charlottesville (M.B., B.B.W.)
| | - Andreas Charidimou
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Marco Pasi
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Christina Kourkoulis
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Center for Genomic Medicine (A.B., P.K., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston.,Henry and Allison McCance Center for Brain Health (A.B., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston
| | - Kristin Schwab
- Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Zora DiPucchio
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Tyler Behymer
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Jennifer Osborne
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Misty Morgan
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Charles J Moomaw
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | - Michael L James
- Department of Anesthesiology, Duke University, Durham, NC (M.L.J.)
| | - Steven M Greenberg
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Anand Viswanathan
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - M Edip Gurol
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston
| | - Bradford B Worrall
- Department of Neurology, University of Virginia Health System, Charlottesville (M.B., B.B.W.)
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, Chicago (F.D.T.)
| | - Jacob L McCauley
- Center for Genome Technology and Biorepository Facility, University of Miami, Miller School of Medicine, FL (J.L.M.)
| | - Guido J Falcone
- Department of Neurology, Yale University School of Medicine, New Haven, CT (G.J.F.)
| | - Carl D Langefeld
- Department of Biostatistics and Data Sciences, Wake Forest University, Winston-Salem, NC (C.D.L.)
| | - Christopher D Anderson
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Center for Genomic Medicine (A.B., P.K., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston.,Henry and Allison McCance Center for Brain Health (A.B., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge (C.D.A., J.R.)
| | - Hooman Kamel
- Department of Neurology, Weill Cornell School of Medicine, New York, NY (H.K.)
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, OH (L.G., P.S., T.B., J.O., M.M., C.J.M., D.W.)
| | | | - Jonathan Rosand
- Department of Neurology (A.B., S.U., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Center for Genomic Medicine (A.B., P.K., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Hemorrhagic Stroke Research Program (A.B., A.R.-T., A.C., M.P., C.K., K.S., Z.D., S.M.G., A.V., M.E.G., C.D.A, J.R.), Massachusetts General Hospital, Boston.,Henry and Allison McCance Center for Brain Health (A.B., C.K., C.D.A., J.R.), Massachusetts General Hospital, Boston.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge (C.D.A., J.R.)
| |
Collapse
|
39
|
Shindo A, Ishikawa H, Ii Y, Niwa A, Tomimoto H. Clinical Features and Experimental Models of Cerebral Small Vessel Disease. Front Aging Neurosci 2020; 12:109. [PMID: 32431603 PMCID: PMC7214616 DOI: 10.3389/fnagi.2020.00109] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease (SVD) refers to a group of disease conditions affecting the cerebral small vessels, which include the small arteries, arterioles, capillaries, and postcapillary venules in the brain. SVD is the primary cause of vascular cognitive impairment and gait disturbances in aged people. There are several types of SVD, though arteriolosclerosis, which is mainly associated with hypertension, aging, and diabetes mellitus, and cerebral amyloid angiopathy (CAA) comprise most SVD cases. The pathology of arteriolosclerosis-induced SVD is characterized by fibrinoid necrosis and lipohyalinosis, while CAA-associated SVD is characterized by progressive deposition of amyloid beta (Aβ) protein in the cerebral vessels. Brain magnetic resonance imaging (MRI) has been used for examination of SVD lesions; typical lesions are characterized by white matter hyperintensity, lacunar infarcts, enlargement of perivascular spaces (EPVS), microbleeds, cortical superficial siderosis (cSS), and cortical microinfarcts. The microvascular changes that occur in the small vessels are difficult to identify clearly; however, these consequent image findings can represent the SVD. There are two main strategies for prevention and treatment of SVD, i.e., pharmacotherapy and lifestyle modification. In this review, we discuss clinical features of SVD, experimental models replicating SVD, and treatments to further understand the pathological and clinical features of SVD.
Collapse
Affiliation(s)
- Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Atsushi Niwa
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| |
Collapse
|
40
|
Pongpitakmetha T, Fotiadis P, Pasi M, Boulouis G, Xiong L, Warren AD, Schwab KM, Rosand J, Gurol ME, Greenberg SM, Viswanathan A, Charidimou A. Cortical superficial siderosis progression in cerebral amyloid angiopathy: Prospective MRI study. Neurology 2020; 94:e1853-e1865. [PMID: 32284360 DOI: 10.1212/wnl.0000000000009321] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/26/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To investigate the prevalence, predictors, and clinical relevance of cortical superficial siderosis (cSS) progression in cerebral amyloid angiopathy (CAA). METHODS Consecutive patients with symptomatic CAA meeting Boston criteria in a prospective cohort underwent baseline and follow-up MRI within 1 year. cSS progression was evaluated on an ordinal scale and categorized into mild (score 1-2 = cSS extension within an already present cSS focus or appearance of 1 new cSS focus) and severe progression (score 3-4 = appearance of ≥2 new cSS foci). Binominal and ordinal multivariable logistic regression were used to determine cSS progression predictors. We investigated future lobar intracerebral hemorrhage (ICH) risk in survival analysis models. RESULTS We included 79 patients with CAA (mean age, 69.2 years), 56 (71%) with lobar ICH at baseline. cSS progression was detected in 23 (29%) patients: 15 (19%) patients had mild and 8 (10%) severe progression. In binominal multivariable logistic regression, ICH presence (odds ratio [OR], 7.54; 95% confidence interval [CI], 1.75-53.52; p = 0.016) and baseline cSS (OR, 10.41; 95% CI, 2.84-52.83; p = 0.001) were independent predictors of cSS progression. In similar models, presence of disseminated (but not focal) cSS at baseline (OR, 5.58; 95% CI, 1.81-19.41; p = 0.004) was an independent predictor of cSS progression. Results were similar in ordinal multivariable logistic regression models. In multivariable Cox regression analysis, severe cSS progression was independently associated with increased future ICH risk (HR, 5.90; 95% CI, 1.30-26.68; p = 0.021). CONCLUSIONS cSS evolution on MRI is common in patients with symptomatic CAA and might be a potential biomarker for assessing disease severity and future ICH risk. External validation of these findings is warranted.
Collapse
Affiliation(s)
- Thanakit Pongpitakmetha
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Panagiotis Fotiadis
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Marco Pasi
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Gregoire Boulouis
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Li Xiong
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Andrew D Warren
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Kristin M Schwab
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Jonathan Rosand
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - M Edip Gurol
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Steven M Greenberg
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Anand Viswanathan
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand
| | - Andreas Charidimou
- From the Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology (T.P., P.F., M.P., G.B., L.X., A.D.W., K.M.S., J.R., M.E.G., S.M.G., A.V., A.C.), and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, and MIND Informatics, Massachusetts General Hospital Biomedical Informatics Core (J.R.), Harvard Medical School, Boston; and Department of Pharmacology, Faculty of Medicine (T.P.), Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
41
|
Baldacci F, Mazzucchi S, Della Vecchia A, Giampietri L, Giannini N, Koronyo-Hamaoui M, Ceravolo R, Siciliano G, Bonuccelli U, Elahi FM, Vergallo A, Lista S, Giorgi FS. The path to biomarker-based diagnostic criteria for the spectrum of neurodegenerative diseases. Expert Rev Mol Diagn 2020; 20:421-441. [PMID: 32066283 PMCID: PMC7445079 DOI: 10.1080/14737159.2020.1731306] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
Introduction: The postmortem examination still represents the reference standard for detecting the pathological nature of chronic neurodegenerative diseases (NDD). This approach displays intrinsic conceptual limitations since NDD represent a dynamic spectrum of partially overlapping phenotypes, shared pathomechanistic alterations that often give rise to mixed pathologies.Areas covered: We scrutinized the international clinical diagnostic criteria of NDD and the literature to provide a roadmap toward a biomarker-based classification of the NDD spectrum. A few pathophysiological biomarkers have been established for NDD. These are time-consuming, invasive, and not suitable for preclinical detection. Candidate screening biomarkers are gaining momentum. Blood neurofilament light-chain represents a robust first-line tool to detect neurodegeneration tout court and serum progranulin helps detect genetic frontotemporal dementia. Ultrasensitive assays and retinal scans may identify Aβ pathology early, in blood and the eye, respectively. Ultrasound also represents a minimally invasive option to investigate the substantia nigra. Protein misfolding amplification assays may accurately detect α-synuclein in biofluids.Expert opinion: Data-driven strategies using quantitative rather than categorical variables may be more reliable for quantification of contributions from pathophysiological mechanisms and their spatial-temporal evolution. A systems biology approach is suitable to untangle the dynamics triggering loss of proteostasis, driving neurodegeneration and clinical evolution.
Collapse
Affiliation(s)
- Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
| | - Sonia Mazzucchi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Giannini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fanny M. Elahi
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Sean Giorgi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
42
|
Yost M, Fiebelkorn CA, Rabinstein AA, Klaas J, Aakre JA, Brown RD, Mielke MM, Knopman DS, Lowe V, Petersen RC, Jack CR, Vemuri P, Graff-Radford J. Incidence of Convexal Subarachnoid Hemorrhage in the Elderly: The Mayo Clinic Study of Aging. J Stroke Cerebrovasc Dis 2019; 28:104451. [PMID: 31668581 PMCID: PMC6886710 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Nontraumatic convexal subarachnoid hemorrhages in the elderly can be a manifestation of cerebral amyloid angiopathy associated with a high risk of future intracerebral hemorrhage. The incidence in the elderly population is unknown. Our objectives were to: 1) determine the incidence of convexal subarachnoid hemorrhage in a population-based study, and, 2) to compare apopolipoprotein-E genotype and amyloid positron emission tomographic (PET) imaging for those with versus without hemorrhage. METHODS Between 11/29/2004 and 3/11/2017, 4462 individuals without hemorrhage at baseline participated in the population-based Mayo Clinic Study of Aging. We used the Rochester Epidemiology Project medical records-linkage system to identify intracerebral hemorrhages. Records and images were reviewed to identify convexal subarachnoid hemorrhage. Neuroimaging characteristics, demographics, medications, and apopolipoprotein-E genotype were recorded. RESULTS Four cases were identified. The incidence of convexal subarachnoid hemorrhage was 14.1 per 100,000 person years. Three occurred in women, median age, 79 (range: 71-84). One patient had coexisting cerebral microbleeds. Two participants developed a subsequent lobar intracerebral hemorrhage at a median of 4.75 years after convexal subarachnoid hemorrhage. The apopolipoprotein-E -allele combinations of the 4 were: 3/3, 3/3, 2/2, and 2/3. On Pittsburgh Compound B-PET imaging, median standardized uptake value ratio with convexal subarachnoid hemorrhage was 1.86 (range: 1.38-2.34). CONCLUSIONS Convexal subarachnoid hemorrhage is rare in the older population, occurring with an incidence of about 14 per 100,000 person years. Yet, when present, it may be associated with a high risk of future intracerebral hemorrhage.
Collapse
Affiliation(s)
- Micah Yost
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | - James Klaas
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Robert D Brown
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, Minnesota; Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Val Lowe
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Prashanthi Vemuri
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
43
|
Schreiber S, Wilisch-Neumann A, Schreiber F, Assmann A, Scheumann V, Perosa V, Jandke S, Mawrin C, Carare RO, Werring DJ. Invited Review: The spectrum of age-related small vessel diseases: potential overlap and interactions of amyloid and nonamyloid vasculopathies. Neuropathol Appl Neurobiol 2019; 46:219-239. [PMID: 31386773 DOI: 10.1111/nan.12576] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
Abstract
Deep perforator arteriopathy (DPA) and cerebral amyloid angiopathy (CAA) are the commonest known cerebral small vessel diseases (CSVD), which cause ischaemic stroke, intracebral haemorrhage (ICH) and vascular cognitive impairment (VCI). While thus far mainly considered as separate entities, we here propose that DPA and CAA share similarities, overlap and interact, so that 'pure' DPA or CAA are extremes along a continuum of age-related small vessel pathologies. We suggest blood-brain barrier (BBB) breakdown, endothelial damage and impaired perivascular β-amyloid (Aβ) drainage are hallmark common mechanisms connecting DPA and CAA. We also suggest a need for new biomarkers (e.g. high-resolution imaging) to deepen understanding of the complex relationships between DPA and CAA.
Collapse
Affiliation(s)
- S Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany.,Center for behavioral brain sciences (CBBS), Magdeburg, Germany
| | - A Wilisch-Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - F Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - A Assmann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - V Scheumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - V Perosa
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - S Jandke
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - C Mawrin
- Department of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - R O Carare
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - D J Werring
- Stroke Research Centre, Department of Brain Repair & Rehabilitation, UCL Institute of Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
44
|
Vales-Montero M, García-Pastor A, Iglesias-Mohedano AM, Esteban-de Antonio E, Salgado-Cámara P, García-Domínguez JM, Vázquez-Alén P, Díaz-Otero F, Fernández-Bullido Y, Gil-Núñez A. Cerebral amyloid angiopathy-related transient focal neurological episodes: A transient ischemic attack mimic with an increased risk of intracranial hemorrhage. J Neurol Sci 2019; 406:116452. [PMID: 31525529 DOI: 10.1016/j.jns.2019.116452] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 08/05/2019] [Accepted: 09/06/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Transient focal neurological episodes (TFNEs) are a recently recognized clinical presentation of cerebral amyloid angiopathy (CAA). Our aim was to describe the clinical and radiological features of a series of patients with AS. METHODS We included 11 patients presenting with recurrent transient focal neurological symptoms and radiological features related to CAA. RESULTS Mean age was 76,6 and 5 patients were women. All patients reported transient, stereotyped, and recurrent episodes (6 patients had >10 episodes). Gradual spread of the symptoms was recorded in 9 patients. Initially, 3 patients were misdiagnosed as having recurrent transient ischemic attack (TIA), 6 as having seizures, and 2 as having both. Two patients were prescribed antiplatelet therapy. A cerebral MRI with T2* gradient-recalled echo sequence revealed cortical superficial siderosis (cSS) in 5 patients, cortical microbleeds in 1 patient, and both features in 5 cases. After a median follow-up of 36 months, intracranial hemorrhage (ICH) was recorded in 4 patients. All 4 had cSS in the previous cerebral MRI, and 1 was on antiplatelet therapy. CONCLUSION CAA-related TFNEs are an underdiagnosed entity, often mimicking TIA, seizures, or migraine aura. This misdiagnosis can lead to the prescription of antiplatelet or anticoagulant therapy, which increases the risk of ICH. Our results suggest that cSS might be a radiological marker that is closely related to an increased risk of bleeding. A T2* gradient-recalled echo MRI should be performed in elderly patients with transient focal neurological symptoms suggestive of CAA.
Collapse
Affiliation(s)
- Marta Vales-Montero
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Andrés García-Pastor
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain.
| | - Ana María Iglesias-Mohedano
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Ester Esteban-de Antonio
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Paula Salgado-Cámara
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - José Manuel García-Domínguez
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Pilar Vázquez-Alén
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Fernando Díaz-Otero
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Yolanda Fernández-Bullido
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Antonio Gil-Núñez
- Stroke Centre - Department of Vascular Neurology, Hospital General Universitario Gregorio Marañón, C/ Dr. Esquerdo 46, 28007 Madrid, Spain
| |
Collapse
|
45
|
Carmona-Iragui M, Videla L, Lleó A, Fortea J. Down syndrome, Alzheimer disease, and cerebral amyloid angiopathy: The complex triangle of brain amyloidosis. Dev Neurobiol 2019; 79:716-737. [PMID: 31278851 DOI: 10.1002/dneu.22709] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/04/2019] [Accepted: 07/02/2019] [Indexed: 11/07/2022]
Abstract
Down syndrome (DS) is the main genetic cause of intellectual disability worldwide. The overexpression of the Amyloid Precursor Protein, present in chromosome 21, leads to β-amyloid deposition that results in Alzheimer disease (AD) and, in most cases, also to cerebral amyloid angiopathy (CAA) neuropathology. People with DS invariably develop the neuropathological hallmarks of AD at the age of 40, and they are at an ultra high risk for suffering AD-related cognitive impairment thereafter. In the general population, cerebrovascular disease is a significant contributor to AD-related cognitive impairment, while in DS remains understudied. This review describes the current knowledge on cerebrovascular disease in DS and reviews the potential biomarkers that could be useful in the future studies, focusing on CAA. We also discuss available evidence on sporadic AD or other genetically determined forms of AD. We highlight the urgent need of large biomarker-characterized cohorts, including neuropathological correlations, to study the exact contribution of CAA and related vascular factors that play a role in cognition and occur with aging, their characterization and interrelationships. DS represents a unique context in which to perform these studies as this population is relatively protected from some conventional vascular risk factors and they develop significant CAA, DS represents a particular atheroma-free model to study AD-related vascular pathologies. Only deepening on these underlying mechanisms, new preventive and therapeutic strategies could be designed to improve the quality of life of this population and their caregivers and lead to new avenues of treatment also in the general AD population.
Collapse
Affiliation(s)
- María Carmona-Iragui
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Videla
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
46
|
Charidimou A, Zonneveld HI, Shams S, Kantarci K, Shoamanesh A, Hilal S, Yates PA, Boulouis G, Na HK, Pasi M, Biffi A, Chai YL, Chong JR, Wahlund LO, Clifford JR, Chen C, Gurol ME, Goldstein JN, Na DL, Barkhof F, Seo SW, Rosand J, Greenberg SM, Viswanathan A. APOE and cortical superficial siderosis in CAA: Meta-analysis and potential mechanisms. Neurology 2019; 93:e358-e371. [PMID: 31243071 DOI: 10.1212/wnl.0000000000007818] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To assess potential mechanisms of cortical superficial siderosis (cSS), a central MRI biomarker in cerebral amyloid angiopathy (CAA), we performed a collaborative meta-analysis of APOE associations with cSS presence and severity. METHODS We pooled data from published studies reporting APOE genotype and MRI assessment of cSS in 3 distinct settings: (1) stroke clinic patients with symptomatic CAA (i.e., lobar intracerebral hemorrhage, transient focal neurologic episodes) according to the Boston criteria; (2) memory clinic patients; and (3) population-based studies. We compared cSS presence and severity (focal or disseminated vs no cSS) in participants with ε2+ or ε4+ genotype vs the ε3/ε3 genotype, by calculating study-specific and random effects pooled, unadjusted odds ratios (ORs). RESULTS Thirteen studies fulfilled inclusion criteria: 7 memory clinic cohorts (n = 2,587), 5 symptomatic CAA cohorts (n = 402), and 1 population-based study (n = 1,379). There was no significant overall association between APOE ε4+ and cSS presence or severity. When stratified by clinical setting, APOE ε4+ was associated with cSS in memory clinic (OR 2.10; 95% confidence interval [CI] 1.11-3.99) but not symptomatic CAA patients. The pooled OR showed significantly increased odds of having cSS for APOE ε2+ genotypes (OR 2.42, 95% CI 1.48-3.95) in both patient populations. This association was stronger for disseminated cSS in symptomatic CAA cohorts. In detailed subgroup analyses, APOE ε2/ε2 and APOE ε2/ε4 genotypes were most consistently and strongly associated with cSS presence and severity. CONCLUSION CAA-related vasculopathic changes and fragility associated with APOE ε2+ allele might have a biologically meaningful role in the pathophysiology and severity of cSS.
Collapse
Affiliation(s)
- Andreas Charidimou
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston.
| | - Hazel I Zonneveld
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sara Shams
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kejal Kantarci
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ashkan Shoamanesh
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Saima Hilal
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Paul A Yates
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Gregoire Boulouis
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Han Kyu Na
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Marco Pasi
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Allesandro Biffi
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Yuek Ling Chai
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Joyce Ruifen Chong
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lars-Olof Wahlund
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jack R Clifford
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Christopher Chen
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - M Edip Gurol
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Joshua N Goldstein
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Duk L Na
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Frederik Barkhof
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sang Won Seo
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jonathan Rosand
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Steven M Greenberg
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anand Viswanathan
- From the Hemorrhagic Stroke Research Program, Department of Neurology (A.C., M.P., A.B., M.E.G., J.N.G., J.R., S.M.G., A.V.), Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston; Alzheimer Center and the Neuroscience Campus Amsterdam and Departments of Radiology and Nuclear Medicine (H.I.Z., F.B.), VU University Medical Center, the Netherlands; Karolinska Institutet (S.S., L.-O.W.), Karolinska University Hospital, Stockholm, Sweden; Department of Radiology (K.K., J.R.C.), Mayo Clinic, Rochester, MN; Department of Medicine (Neurology) (A.S.), McMaster University and Population Health Research Institute, Hamilton, Canada; Memory, Aging and Cognition Center (S.H., Y.L.C., J.R.C., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., Y.L.C., J.R.C., C.C.), National University of Singapore; Department of Nuclear Medicine and Centre for PET (P.A.Y.), The University of Melbourne, Parkville, Australia; Department of Neuroradiology (G.B.), Université Paris-Descartes, INSERM U894, CH Sainte-Anne, Paris, France; Department of Neurology and Neuroscience Center (H.K.N., D.L.N., S.W.S.), Samsung Medical Center, Seoul, Republic of Korea; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; and Center for Genomic Medicine (J.R.) and Division of Neurocritical Care and Emergency Neurology (J.R.), Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
47
|
Kulesh AA, Drobakha VE, Shestakov VV. Cerebral small vessel disease: classification, clinical manifestations, diagnosis, and features of treatment. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2019. [DOI: 10.14412/2074-2711-2019-3s-4-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The paper considers the relevance of the problem of cerebral small vessel disease (CSVD) that is an important cause of ischemic and hemorrhagic stroke, associated with the development of cognitive impairment and complications of antithrombotic therapy. It presents briefly the current issues of etiology and pathogenesis of the disease. Sporadic non-amyloid microangiopathy, cerebral amyloid angiopathy, and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) are discussed in detail from the point of view of their clinical presentation, neuroimaging, and features of therapeutic tactics. An algorithm for diagnosing CSVD in patients admitted to hospital for stroke and a differentiated approach to their treatment are proposed. Consideration of the neuroimaging manifestations of CSVD is noted to be necessary for the safe and more effective treatment of patients with cerebrovascular diseases.
Collapse
Affiliation(s)
- A. A. Kulesh
- Acad. E.A. Vagner Perm State Medical University, Ministry of Health of Russia
| | - V. E. Drobakha
- Acad. E.A. Vagner Perm State Medical University, Ministry of Health of Russia
| | - V. V. Shestakov
- Acad. E.A. Vagner Perm State Medical University, Ministry of Health of Russia
| |
Collapse
|
48
|
Das AS, Regenhardt RW, Vernooij MW, Blacker D, Charidimou A, Viswanathan A. Asymptomatic Cerebral Small Vessel Disease: Insights from Population-Based Studies. J Stroke 2019; 21:121-138. [PMID: 30991799 PMCID: PMC6549070 DOI: 10.5853/jos.2018.03608] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/28/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is a common group of neurological conditions that confer a significant burden of morbidity and mortality worldwide. In most cases, CSVD is only recognized in its advanced stages once its symptomatic sequelae develop. However, its significance in asymptomatic healthy populations remains poorly defined. In population-based studies of presumed healthy elderly individuals, CSVD neuroimaging markers including white matter hyperintensities, lacunes, cerebral microbleeds, enlarged perivascular spaces, cortical superficial siderosis, and cerebral microinfarcts are frequently detected. While the presence of these imaging markers may reflect unique mechanisms at play, there are likely shared pathways underlying CSVD. Herein, we aim to assess the etiology and significance of these individual biomarkers by focusing in asymptomatic populations at an epidemiological level. By primarily examining population-based studies, we explore the risk factors that are involved in the formation and progression of these biomarkers. Through a critical semi-systematic review, we aim to characterize “asymptomatic” CSVD, review screening modalities, and draw associations from observational studies in clinical populations. Lastly, we highlight areas of research (including therapeutic approaches) in which further investigation is needed to better understand asymptomatic CSVD.
Collapse
Affiliation(s)
- Alvin S Das
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Deborah Blacker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andreas Charidimou
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Clinical significance of amyloid β positivity in patients with probable cerebral amyloid angiopathy markers. Eur J Nucl Med Mol Imaging 2019; 46:1287-1298. [DOI: 10.1007/s00259-019-04314-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
|
50
|
Camacho J, Moliné T, Bonaterra-Pastra A, Ramón Y Cajal S, Martínez-Sáez E, Hernández-Guillamon M. Brain ApoA-I, ApoJ and ApoE Immunodetection in Cerebral Amyloid Angiopathy. Front Neurol 2019; 10:187. [PMID: 30918495 PMCID: PMC6424885 DOI: 10.3389/fneur.2019.00187] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a common cause of lobar intracerebral hemorrhage (ICH) in elderly individuals and it is the result of the cerebrovascular deposition of beta-amyloid (Aβ) protein. CAA is frequently found in patients with Alzheimer's disease (AD), although it has an independent contribution to the cognitive deterioration associated with age. Specific apolipoproteins (Apo) have been associated with Aβ fibrillization and clearance from the brain. In this regard, in the present study, we analyzed the brain levels of ApoE, ApoA-I, and ApoJ/clusterin in autopsy brains from 20 post-mortem cases with CAA type I, CAA type II, with parenchymal Aβ deposits or without Aβ deposits. Our objective was to find a possible differential pattern of apolipoproteins distribution in the brain depending on the CAA pathological presentation. The protein expression levels were adjusted by the APOE genotype of the patients included in the study. We found that ApoE and ApoJ were abundantly present in meningeal, cortical, and capillary vessels of the brains with vascular Aβ accumulation. ApoE and ApoJ also deposited extracellularly in the parenchyma, especially in cases presenting Aβ diffuse and neuritic parenchymal deposits. In contrast, ApoA-I staining was only relevant in capillary walls in CAA type I cases. On the other hand, ICH was the principal cause of death among CAA patients in our cohort. We found that CAA patients with ICH more commonly had APOEε2 compared with CAA patients without ICH. In addition, patients who suffered an ICH presented higher vascular ApoE levels in brain. However, higher ApoE presence in cortical arteries was the only independent predictor of suffering an ICH in our cohort after adjusting by age and APOE genotype. In conclusion, while ApoE and ApoJ appear to be involved in both vascular and parenchymal Aβ pathology, ApoA-I seems to be mainly associated with CAA, especially in CAA type I pathology. We consider that our study helps to molecularly characterize the distribution subtypes of Aβ deposition within the brain.
Collapse
Affiliation(s)
- Jessica Camacho
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Moliné
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Martínez-Sáez
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|