1
|
Hu H, Wan S, Hu Y, Wang Q, Li H, Zhang N. Deciphering the role of APOE in cerebral amyloid angiopathy: from genetic insights to therapeutic horizons. Ann Med 2025; 57:2445194. [PMID: 39745195 DOI: 10.1080/07853890.2024.2445194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid-β (Aβ) peptides in the walls of medium and small vessels of the brain and leptomeninges, is a major cause of lobar hemorrhage in elderly individuals. Among the genetic risk factors for CAA that continue to be recognized, the apolipoprotein E (APOE) gene is the most significant and prevalent, as its variants have been implicated in more than half of all patients with CAA. While the presence of the APOE ε4 allele markedly increases the risk of CAA, the ε2 allele confers a protective effect relative to the common ε3 allele. These allelic variants encode three APOE isoforms that differ at two amino acid positions. The primary physiological role of APOE is to mediate lipid transport in the brain and periphery; however, it has also been shown to be involved in a wide array of biological functions, particularly those involving Aβ, in which it plays a known role in processing, production, aggregation, and clearance. The challenges posed by the reliance on postmortem histological analyses and the current absence of an effective intervention underscore the urgency for innovative APOE-targeted strategies for diagnosing CAA. This review not only deepens our understanding of the impact of APOE on the pathogenesis of CAA but can also help guide the exploration of targeted therapies, inspiring further research into the therapeutic potential of APOE.
Collapse
Affiliation(s)
- Hantian Hu
- Tianjin Medical University, Tianjin, China
| | - Siqi Wan
- Tianjin Medical University, Tianjin, China
| | - Yuetao Hu
- Tianjin Medical University, Tianjin, China
| | - Qi Wang
- Tianjin Medical University, Tianjin, China
| | - Hanyu Li
- Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Ban SY, Nam Y, Do TT, Kim BH, Shin SJ, Thi Nguyen MT, Kim J, Moon M, Park JT. Liver-X receptor β-selective agonist CE9A215 regulates Alzheimer's disease-associated pathology in a 3xTg-AD mouse model. Biomed Pharmacother 2025; 184:117895. [PMID: 39919463 DOI: 10.1016/j.biopha.2025.117895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
In Alzheimer's disease (AD), tau pathology is closely associated with disease progression. Therefore, therapeutics that alleviate tau pathology are essential. Liver-X receptor (LXR) has garnered interest as a potential target for the treatment of AD. We previously investigated the potent anti-allergic and anti-inflammatory effects of inotodiol, hereafter referred to as CE9A215, in various disease models. In this study, we explored the potential of CE9A215 as a treatment for AD. CE9A215 preferentially activated LXRβ (EC50 <10 nM), with no significant activation observed for LXRα at concentrations up to 1000 nM. Pharmacokinetic analysis confirmed that CE9A215 crosses the blood-brain barrier and accumulates in the brain. Moreover, CE9A215 modulated the expression of ABCA1, APOE, SREBP-1c and AQP4 in the brains of wild-type and LXR α/β knockout mice in LXRβ-dependent manner. The efficacy of CE9A215 on AD-related pathologies was evaluated using 3xTg-AD mice. CE9A215 exerted both prophylactic and therapeutic effects on AD-associated behaviors and pathologies, including reductions in amyloid-β, phosphorylated tau, and neuroinflammation in the hippocampus. Transcriptomic analysis revealed that CE9A215 induced significant changes in genes associated with tau pathology, particularly in pathways related to protein phosphorylation and PI3K/AKT signaling. Our findings suggest that CE9A215 could be a promising therapeutic candidate for AD, particularly in mitigating tau hyperphosphorylation and related AD pathologies.
Collapse
Affiliation(s)
- So Young Ban
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | | | - Byeong-Hyeon Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - My Tuyen Thi Nguyen
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Republic of Korea; Department of Food Technology, Can Tho University, Can Tho 94000, Viet Nam
| | - Jaehan Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea; Research Institute for Dementia Science, Konyang University, Daejeon 35365, Republic of Korea.
| | - Jong-Tae Park
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea.
| |
Collapse
|
3
|
Taghvaei M, Dolui S, Sadaghiani S, Shakibajahromi B, Brown C, Khandelwal P, Xie SX, Das S, Yushkevich PA, Wolk DA, Detre JA. Regional cerebral blood flow reflects both neurodegeneration and microvascular integrity across the Alzheimer's continuum. Alzheimers Dement 2025; 21:e14382. [PMID: 39625074 PMCID: PMC11772719 DOI: 10.1002/alz.14382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) typically involves both neurodegenerative and vascular pathologies, each associated with reductions in cerebral blood flow (CBF). However, it remains unclear whether vascular and neural contributions to regional CBF can be differentiated. METHODS Using 3D background-suppressed arterial spin labeled perfusion magnetic resonance imaging, we evaluated regional CBF in a cohort of 257 participants across the AD continuum and assessed the impact of risk factors for both AD and small vessel disease (SVD) on regional CBF. RESULTS Vascular risk factors (VRFs) were associated with reduced CBF in normal-appearing periventricular white matter, while amyloid positivity was associated with reduced CBF in the posterior cingulate cortex and precuneus. Putative SVD-sensitive regions in white matter exhibited diagnosis-related CBF changes comparable to those in typical AD cortical regions. DISCUSSION Spatial patterns of hypoperfusion may differentiate AD and VRF-related effects on regional CBF. Our findings also support the contribution of SVD in AD pathogenesis. HIGHLIGHTS We used 3D background-suppressed pCASL MRI to evaluate CBF across the AD continuum. Putative SVD-sensitive regions in white matter exhibited diagnosis-related CBF changes. AD and/or SVD risk correlated with reduced CBF in AD and/or SVD-related regions. VRFs were associated with more widespread CBF reductions than amyloid positivity. Spatial patterns of hypoperfusion may differentiate AD and VRF-related effects.
Collapse
Affiliation(s)
- Mohammad Taghvaei
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sudipto Dolui
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shokufeh Sadaghiani
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Christopher Brown
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Pulkit Khandelwal
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sharon X. Xie
- Department of BiostatisticsEpidemiology, and InformaticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sandhitsu Das
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Paul A. Yushkevich
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David A. Wolk
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - John A. Detre
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
4
|
Kang M, Li C, Mahajan A, Spat-Lemus J, Durape S, Chen J, Gurnani AS, Devine S, Auerbach SH, Ang TFA, Sherva R, Qiu WQ, Lunetta KL, Au R, Farrer LA, Mez J. Subjective Cognitive Decline Plus and Longitudinal Assessment and Risk for Cognitive Impairment. JAMA Psychiatry 2024; 81:993-1002. [PMID: 38959008 PMCID: PMC11223054 DOI: 10.1001/jamapsychiatry.2024.1678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/17/2024] [Indexed: 07/04/2024]
Abstract
Importance Subjective cognitive decline (SCD) is recognized to be in the Alzheimer disease (AD) cognitive continuum. The SCD Initiative International Working Group recently proposed SCD-plus (SCD+) features that increase risk for future objective cognitive decline but that have not been assessed in a large community-based setting. Objective To assess SCD risk for mild cognitive impairment (MCI), AD, and all-cause dementia, using SCD+ criteria among cognitively normal adults. Design, Setting, and Participants The Framingham Heart Study, a community-based prospective cohort study, assessed SCD between 2005 and 2019, with up to 12 years of follow-up. Participants 60 years and older with normal cognition at analytic baseline were included. Cox proportional hazards (CPH) models were adjusted for baseline age, sex, education, APOE ε4 status, and tertiles of AD polygenic risk score (PRS), excluding the APOE region. Data were analyzed from May 2021 to November 2023. Exposure SCD was assessed longitudinally using a single question and considered present if endorsed at the last cognitively normal visit. It was treated as a time-varying variable, beginning at the first of consecutive, cognitively normal visits, including the last, at which it was endorsed. Main Outcomes and Measures Consensus-diagnosed MCI, AD, and all-cause dementia. Results This study included 3585 participants (mean [SD] baseline age, 68.0 [7.7] years; 1975 female [55.1%]). A total of 1596 participants (44.5%) had SCD, and 770 (21.5%) were carriers of APOE ε4. APOE ε4 and tertiles of AD PRS status did not significantly differ between the SCD and non-SCD groups. MCI, AD, and all-cause dementia were diagnosed in 236 participants (6.6%), 73 participants (2.0%), and 89 participants (2.5%), respectively, during follow-up. On average, SCD preceded MCI by 4.4 years, AD by 6.8 years, and all-cause dementia by 6.9 years. SCD was significantly associated with survival time to MCI (hazard ratio [HR], 1.57; 95% CI, 1.22-2.03; P <.001), AD (HR, 2.98; 95% CI, 1.89-4.70; P <.001), and all-cause dementia (HR, 2.14; 95% CI, 1.44-3.18; P <.001). After adjustment for APOE and AD PRS, the hazards of SCD were largely unchanged. Conclusions and Relevance Results of this cohort study suggest that in a community setting, SCD reflecting SCD+ features was associated with an increased risk of future MCI, AD, and all-cause dementia with similar hazards estimated in clinic-based settings. SCD may be an independent risk factor for AD and other dementias beyond the risk incurred by APOE ε4 and AD PRS.
Collapse
Affiliation(s)
- Moonil Kang
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Clara Li
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arnav Mahajan
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Jessica Spat-Lemus
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Psychology, Montclair State University, Montclair, New Jersey
| | - Shruti Durape
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Jiachen Chen
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Ashita S. Gurnani
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Sherral Devine
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Sanford H. Auerbach
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Ting Fang Alvin Ang
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Richard Sherva
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Wei Qiao Qiu
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Kathryn L. Lunetta
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Rhoda Au
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Lindsay A. Farrer
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Jesse Mez
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| |
Collapse
|
5
|
Foley KE, Wilcock DM. Three major effects of APOE ε4 on Aβ immunotherapy induced ARIA. Front Aging Neurosci 2024; 16:1412006. [PMID: 38756535 PMCID: PMC11096466 DOI: 10.3389/fnagi.2024.1412006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
The targeting of amyloid-beta (Aβ) plaques therapeutically as one of the primary causes of Alzheimer's disease (AD) dementia has been an ongoing effort spanning decades. While some antibodies are extremely promising and have been moved out of clinical trials and into the clinic, most of these treatments show similar adverse effects in the form of cerebrovascular damage known as amyloid-related imaging abnormalities (ARIA). The two categories of ARIA are of major concern for patients, families, and prescribing physicians, with ARIA-E presenting as cerebral edema, and ARIA-H as cerebral hemorrhages (micro- and macro-). From preclinical and clinical trials, it has been observed that the greatest genetic risk factor for AD, APOEε4, is also a major risk factor for anti-Aβ immunotherapy-induced ARIA. APOEε4 carriers represent a large population of AD patients, and, therefore, limits the broad adoption of these therapies across the AD population. In this review we detail three hypothesized mechanisms by which APOEε4 influences ARIA risk: (1) reduced cerebrovascular integrity, (2) increased neuroinflammation and immune dysregulation, and (3) elevated levels of CAA. The effects of APOEε4 on ARIA risk is clear, however, the underlying mechanisms require more research.
Collapse
Affiliation(s)
- Kate E. Foley
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, United States
- Department of Neurology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, United States
- Department of Neurology, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
6
|
Edwards L, Thomas KR, Weigand AJ, Edmonds EC, Clark AL, Brenner EK, Banks SJ, Gilbert PE, Nation DA, Delano-Wood L, Bondi MW, Bangen KJ. Pulse pressure and APOE ε4 dose interact to affect cerebral blood flow in older adults without dementia. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100206. [PMID: 38328026 PMCID: PMC10847851 DOI: 10.1016/j.cccb.2024.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/20/2023] [Accepted: 01/14/2024] [Indexed: 02/09/2024]
Abstract
This study assessed whether the effect of vascular risk on cerebral blood flow (CBF) varies by gene dose of apolipoprotein (APOE) ε4 alleles. 144 older adults without dementia from the Alzheimer's Disease Neuroimaging Initiative underwent arterial spin labeling and T1-weighted MRI, APOE genotyping, fluorodeoxyglucose positron emission tomography (FDG-PET), lumbar puncture, and blood pressure (BP) assessment. Vascular risk was assessed using pulse pressure (systolic BP - diastolic BP). CBF was examined in six AD-vulnerable regions: entorhinal cortex, hippocampus, inferior temporal cortex, inferior parietal cortex, rostral middle frontal gyrus, and medial orbitofrontal cortex. Linear regressions tested the interaction between APOE ε4 dose and pulse pressure on CBF in each region, adjusting for age, sex, cognitive classification, antihypertensive medication use, FDG-PET, reference CBF region, and AD biomarker positivity. There was a significant interaction between pulse pressure and APOE ɛ4 dose on CBF in the entorhinal cortex, hippocampus, and inferior parietal cortex, such that higher pulse pressure was associated with lower CBF only among ε4 homozygous participants. These findings demonstrate that the association between pulse pressure and regional CBF differs by APOE ε4 dose, suggesting that targeting modifiable vascular risk factors may be particularly important for those genetically at risk for AD.
Collapse
Affiliation(s)
- Lauren Edwards
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Kelsey R. Thomas
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra J. Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Emily C. Edmonds
- Banner Alzheimer's Institute, Tucson, AZ, USA
- Departments of Neurology and Psychology, University of Arizona, Tucson, AZ, USA
| | - Alexandra L. Clark
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Einat K. Brenner
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Sarah J. Banks
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Paul E. Gilbert
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
- Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Daniel A. Nation
- Department of Psychology, University of California Irvine, Irvine, CA, USA
| | - Lisa Delano-Wood
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Mark W. Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Katherine J. Bangen
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Wang H, Zhang Z, Hongpaisan J. PKCε activator protects hippocampal microvascular disruption and memory defect in 3×Tg-Alzheimer's disease mice with cerebral microinfarcts. Front Aging Neurosci 2023; 15:1272361. [PMID: 38187357 PMCID: PMC10768563 DOI: 10.3389/fnagi.2023.1272361] [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: 08/03/2023] [Accepted: 11/23/2023] [Indexed: 01/09/2024] Open
Abstract
Background Current evidence suggests that microvessel disease is involved in Alzheimer's disease (AD). Cerebrovascular disease correlates with cardiovascular disease and is complicated in ≈40% of AD patients. The protein kinase C (PKC) ε activator DCPLA can stimulate human antigen (Hu) R that prevents degradation and promotes the translation of mitochondrial Mn-superoxide dismutase (MnSOD) and vascular endothelial growth factor-A (VEGF) mRNAs. Methods To induce brain microinfarcts, we injected triple transgenic (3×Tg) and wild-type (WT) control mice with microbeads (20 μm caliber) into common carotid arteries, with or without the DCPLA-ME (methyl-ester) for 2 weeks. After water maze training, mice at 16 months old were examined for confocal immunohistochemistry at a single cell or microvessel level in the hippocampal CA1 area, important for spatial memory storage, and in the dorsal hippocampus by western blots. Results In 3×Tg mice without cerebral microinfarcts, an accelerating age-related increase in (mild) oxidative stress and hypoxia inducible factor (HIF)-1α, but a reduction in VEGF, mitochondrial transcription factor A (TFAM), and MnSOD were associated with capillary loss. The change was less pronounced in arterioles. However, in 3×Tg mice with cerebral microinfarcts, increasing arteriolar diameter and their wall cells were related with the strong oxidative DNA damage 8-hydroxy-2'-deoxyguanosine (8-OHdG), apoptosis (cleaved caspase 3), and sustained hypoxia (increased HIF-1α and VEGF/PKCε/extracellular signal regulated kinase or ERK pathway). Microocclusion enhanced the loss of the synaptic marker spinophilin, astrocytic number, and astrocyte-vascular coupling areas and demyelination of axons. DCPLA-ME prevented spatial memory defect; strong oxidative stress-related apoptosis; sustained hypoxia (by reducing HIF-1α and VEGF); and exaggerated cell repair in arteriolar walls, pericapillary space dilation, neuro-glial-vascular disruption, and demyelination. Conclusion In conclusion, in 3×Tg mice with cerebral microinfarcts, sustained hypoxia (increased HIF-1α and VEGF signals) is dominant with arteriolar wall thickening, and DCPLA has a protective effect on sustained hypoxia.
Collapse
Affiliation(s)
| | | | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
8
|
Zedde M, Grisendi I, Assenza F, Vandelli G, Napoli M, Moratti C, Lochner P, Seiffge DJ, Piazza F, Valzania F, Pascarella R. The Venular Side of Cerebral Amyloid Angiopathy: Proof of Concept of a Neglected Issue. Biomedicines 2023; 11:2663. [PMID: 37893037 PMCID: PMC10604278 DOI: 10.3390/biomedicines11102663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Small vessel diseases (SVD) is an umbrella term including several entities affecting small arteries, arterioles, capillaries, and venules in the brain. One of the most relevant and prevalent SVDs is cerebral amyloid angiopathy (CAA), whose pathological hallmark is the deposition of amyloid fragments in the walls of small cortical and leptomeningeal vessels. CAA frequently coexists with Alzheimer's Disease (AD), and both are associated with cerebrovascular events, cognitive impairment, and dementia. CAA and AD share pathophysiological, histopathological and neuroimaging issues. The venular involvement in both diseases has been neglected, although both animal models and human histopathological studies found a deposition of amyloid beta in cortical venules. This review aimed to summarize the available information about venular involvement in CAA, starting from the biological level with the putative pathomechanisms of cerebral damage, passing through the definition of the peculiar angioarchitecture of the human cortex with the functional organization and consequences of cortical arteriolar and venular occlusion, and ending to the hypothesized links between cortical venular involvement and the main neuroimaging markers of the disease.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Ilaria Grisendi
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Federica Assenza
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Gabriele Vandelli
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Manuela Napoli
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Claudio Moratti
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - David J. Seiffge
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| |
Collapse
|
9
|
Wang H, Zhang Z, Sittirattanayeunyong S, Hongpaisan J. Association of Apolipoprotein E4-related Microvascular Disease in the Alzheimer's Disease Hippocampal CA1 Stratum Radiatum. Neuroscience 2023; 526:204-222. [PMID: 37385335 PMCID: PMC10528415 DOI: 10.1016/j.neuroscience.2023.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
Current data suggest a hypothesis of vascular pathogenesis for the development and progression of Alzheimer's disease (AD). To investigate this, we studied the association of apolipoprotein E4 (APOE4) gene on microvessels in human autopsy-confirmed AD with and without APOE4, compared with age/sex-matched control (AC) hippocampal CA1 stratum radiatum. AD arterioles (without APOE4 gene) had mild oxidative stress and loss of vascular endothelial growth factor (VEGF) and endothelial cell density, reflecting aging progression. In AD + APOE4, an increase in strong oxidative DNA damage marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), VEGF, and endothelial cell density were associated with increased diameter of arterioles and perivascular space dilation. In cultured human brain microvascular cells (HBMECs), treatment of ApoE4 protein plus amyloid-β (Aβ) oligomers increased superoxide production and the apoptotic marker cleaved caspase 3, sustained hypoxia inducible factor-1α (HIF-1α) stability that was associated with an increase in MnSOD, VEGF, and cell density. This cell over-proliferation was inhibited with the antioxidants N-acetyl cysteine and MnTMPyP, the HIF-1α inhibitor echinomycin, the VEGFR-2 receptor blocker SU1498, the protein kinase C (PKC) ε knock-down (KD) and the extracellular signal-regulated kinase 1/2 (ERK) inhibitor FR180204. The PKCε KD and echinomycin decreased VEGF and/or ERK. In conclusion, AD capillaries and arterioles in hippocampal CA1 stratum radiatum of non-APOE4 carriers are related with aging, while those in APOE4 carriers with AD are related with pathogenesis of cerebrovascular disease.
Collapse
Affiliation(s)
- Huaixing Wang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Zongxiu Zhang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sorawit Sittirattanayeunyong
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
10
|
Agrawal S, Schneider JA. Vascular pathology and pathogenesis of cognitive impairment and dementia in older adults. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100148. [PMID: 36324408 PMCID: PMC9616381 DOI: 10.1016/j.cccb.2022.100148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
It is well recognized that brains of older people often harbor cerebrovascular disease pathology including vessel disease and vascular-related tissue injuries and that this is associated with vascular cognitive impairment and contributes to dementia. Here we review vascular pathologies, cognitive impairment, and dementia. We highlight the importance of mixed co-morbid AD/non-AD neurodegenerative and vascular pathology that has been collected in multiple clinical pathologic studies, especially in community-based studies. We also provide an update of vascular pathologies from the Rush Memory and Aging Project and Religious Orders Study cohorts with special emphasis on the differences across age in persons with and without dementia. Finally, we discuss neuropathological perspectives on the interpretation of clinical-pathological studies and emerging data in community-based studies.
Collapse
Affiliation(s)
- Sonal Agrawal
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Julie A. Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
McCorkindale AN, Mundell HD, Guennewig B, Sutherland GT. Vascular Dysfunction Is Central to Alzheimer's Disease Pathogenesis in APOE e4 Carriers. Int J Mol Sci 2022; 23:7106. [PMID: 35806110 PMCID: PMC9266739 DOI: 10.3390/ijms23137106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and the leading risk factor, after age, is possession of the apolipoprotein E epsilon 4 allele (APOE4). Approximately 50% of AD patients carry one or two copies of APOE4 but the mechanisms by which it confers risk are still unknown. APOE4 carriers are reported to demonstrate changes in brain structure, cognition, and neuropathology, but findings have been inconsistent across studies. In the present study, we used multi-modal data to characterise the effects of APOE4 on the brain, to investigate whether AD pathology manifests differently in APOE4 carriers, and to determine if AD pathomechanisms are different between carriers and non-carriers. Brain structural differences in APOE4 carriers were characterised by applying machine learning to over 2000 brain MRI measurements from 33,384 non-demented UK biobank study participants. APOE4 carriers showed brain changes consistent with vascular dysfunction, such as reduced white matter integrity in posterior brain regions. The relationship between APOE4 and AD pathology was explored among the 1260 individuals from the Religious Orders Study and Memory and Aging Project (ROSMAP). APOE4 status had a greater effect on amyloid than tau load, particularly amyloid in the posterior cortical regions. APOE status was also highly correlated with cerebral amyloid angiopathy (CAA). Bulk tissue brain transcriptomic data from ROSMAP and a similar dataset from the Mount Sinai Brain Bank showed that differentially expressed genes between the dementia and non-dementia groups were enriched for vascular-related processes (e.g., "angiogenesis") in APOE4 carriers only. Immune-related transcripts were more strongly correlated with AD pathology in APOE4 carriers with some transcripts such as TREM2 and positively correlated with pathology severity in APOE4 carriers, but negatively in non-carriers. Overall, cumulative evidence from the largest neuroimaging, pathology, and transcriptomic studies available suggests that vascular dysfunction is key to the development of AD in APOE4 carriers. However, further studies are required to tease out non-APOE4-specific mechanisms.
Collapse
Affiliation(s)
- Andrew N. McCorkindale
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (A.N.M.); (H.D.M.)
| | - Hamish D. Mundell
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (A.N.M.); (H.D.M.)
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Boris Guennewig
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Greg T. Sutherland
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (A.N.M.); (H.D.M.)
| |
Collapse
|
12
|
Wang M, Shao W, Hao X, Huang S, Zhang D. Identify connectome between genotypes and brain network phenotypes via deep self-reconstruction sparse canonical correlation analysis. Bioinformatics 2022; 38:2323-2332. [PMID: 35143604 DOI: 10.1093/bioinformatics/btac074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION As a rising research topic, brain imaging genetics aims to investigate the potential genetic architecture of both brain structure and function. It should be noted that in the brain, not all variations are deservedly caused by genetic effect, and it is generally unknown which imaging phenotypes are promising for genetic analysis. RESULTS In this work, genetic variants (i.e. the single nucleotide polymorphism, SNP) can be correlated with brain networks (i.e. quantitative trait, QT), so that the connectome (including the brain regions and connectivity features) of functional brain networks from the functional magnetic resonance imaging data is identified. Specifically, a connection matrix is firstly constructed, whose upper triangle elements are selected to be connectivity features. Then, the PageRank algorithm is exploited for estimating the importance of different brain regions as the brain region features. Finally, a deep self-reconstruction sparse canonical correlation analysis (DS-SCCA) method is developed for the identification of genetic associations with functional connectivity phenotypic markers. This approach is a regularized, deep extension, scalable multi-SNP-multi-QT method, which is well-suited for applying imaging genetic association analysis to the Alzheimer's Disease Neuroimaging Initiative datasets. It is further optimized by adopting a parametric approach, augmented Lagrange and stochastic gradient descent. Extensive experiments are provided to validate that the DS-SCCA approach realizes strong associations and discovers functional connectivity and brain region phenotypic biomarkers to guide disease interpretation. AVAILABILITY AND IMPLEMENTATION The Matlab code is available at https://github.com/meimeiling/DS-SCCA/tree/main. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Meiling Wang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Xiaoke Hao
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Shuo Huang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| |
Collapse
|
13
|
Seto M, Mahoney ER, Dumitrescu L, Ramanan VK, Engelman CD, Deming Y, Albert M, Johnson SC, Zetterberg H, Blennow K, Vemuri P, Jefferson AL, Hohman TJ. Exploring common genetic contributors to neuroprotection from amyloid pathology. Brain Commun 2022; 4:fcac066. [PMID: 35425899 PMCID: PMC9006043 DOI: 10.1093/braincomms/fcac066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/13/2022] [Accepted: 03/15/2022] [Indexed: 01/25/2023] Open
Abstract
Preclinical Alzheimer's disease describes some individuals who harbour Alzheimer's pathologies but are asymptomatic. For this study, we hypothesized that genetic variation may help protect some individuals from Alzheimer's-related neurodegeneration. We therefore conducted a genome-wide association study using 5 891 064 common variants to assess whether genetic variation modifies the association between baseline beta-amyloid, as measured by both cerebrospinal fluid and positron emission tomography, and neurodegeneration defined using MRI measures of hippocampal volume. We combined and jointly analysed genotype, biomarker and neuroimaging data from non-Hispanic white individuals who were enrolled in four longitudinal ageing studies (n = 1065). Using regression models, we examined the interaction between common genetic variants (Minor Allele Frequency >0.01), including APOE-ɛ4 and APOE-ɛ2, and baseline cerebrospinal levels of amyloid (CSF Aβ42) on baseline hippocampal volume and the longitudinal rate of hippocampal atrophy. For targeted replication of top findings, we analysed an independent dataset (n = 808) where amyloid burden was assessed by Pittsburgh Compound B ([11C]-PiB) positron emission tomography. In this study, we found that APOE-ɛ4 modified the association between baseline CSF Aβ42 and hippocampal volume such that APOE-ɛ4 carriers showed more rapid atrophy, particularly in the presence of enhanced amyloidosis. We also identified a novel locus on chromosome 3 that interacted with baseline CSF Aβ42. Minor allele carriers of rs62263260, an expression quantitative trait locus for the SEMA5B gene (P = 1.46 × 10-8; 3:122675327) had more rapid neurodegeneration when amyloid burden was high and slower neurodegeneration when amyloid was low. The rs62263260 × amyloid interaction on longitudinal change in hippocampal volume was replicated in an independent dataset (P = 0.0112) where amyloid burden was assessed by positron emission tomography. In addition to supporting the established interaction between APOE and amyloid on neurodegeneration, our study identifies a novel locus that modifies the association between beta-amyloid and hippocampal atrophy. Annotation results may implicate SEMA5B, a gene involved in synaptic pruning and axonal guidance, as a high-quality candidate for functional confirmation and future mechanistic analysis.
Collapse
Affiliation(s)
- Mabel Seto
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Emily R. Mahoney
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726, USA
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Geriatric Education and Clinical Center, Wm.S.Middleton VA Hospital, Madison, WI 53705, USA
| | - Yuetiva Deming
- Department of Population Health Sciences, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726, USA
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Geriatric Education and Clinical Center, Wm.S.Middleton VA Hospital, Madison, WI 53705, USA
| | - Marilyn Albert
- Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sterling C. Johnson
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Geriatric Education and Clinical Center, Wm.S.Middleton VA Hospital, Madison, WI 53705, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal 413 90, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 413 45, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal 413 90, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 413 45, Sweden
| | | | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | |
Collapse
|
14
|
Mohseni-Moghaddam P, Ghobadian R, Khaleghzadeh-Ahangar H. Dementia in Diabetes mellitus and Atherosclerosis; Two Interrelated Systemic Diseases. Brain Res Bull 2022; 181:87-96. [DOI: 10.1016/j.brainresbull.2022.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/18/2021] [Accepted: 01/24/2022] [Indexed: 12/06/2022]
|
15
|
Park M, Lee HP, Kim J, Kim DH, Moon Y, Moon WJ. Brain myelin water fraction is associated with APOE4 allele status in patients with cognitive impairment. J Neuroimaging 2021; 32:521-529. [PMID: 34964524 DOI: 10.1111/jon.12960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Apolipoprotein E4 (APOE4) is a major genetic risk factor for Alzheimer's disease. However, the effect of APOE4 status on myelin remains unclear. This study investigated the effect of APOE4 on myelin content in cognitively impaired individuals using T2* gradient echo (GRE)-based myelin water fraction (MWF) imaging. METHODS Between August 2017 and January 2019, we evaluated 39 cognitively impaired patients (median age, 75 years; male:female = 8:31; Alzheimer's disease: mild cognitive impairment = 11:28). We obtained brain MWF values from white matter hyperintensities (WMHs) and normal-appearing white matter (NAWM). Linear regression analysis was performed to investigate the relationship between the APOE4 status and MWF and cognitive function and MWF. RESULTS Among the 39 cognitively impaired patients, nine (23.1%) were APOE4 carriers and 30 (76.9%) were noncarriers. APOE4 carriers had a lower hippocampal volume than noncarriers (p = .045), but other brain volume parameters were not differed. After age adjustment, the APOE4 status was significantly associated with reduced MWF in NAWM (β = -0.310 per allele; p = .049) but not in WMH (β = -0.258 per allele; p = .113). After age adjustment, MWF in NAWM was significantly associated with Mini-Mental State Examination score (β = 0.313, p = .031). CONCLUSIONS T2* GRE-based MWF imaging can reveal myelin loss, particularly in NAWM, in cognitively impaired patients among APOE4 carriers. In vivo MWF in NAWM might be a novel imaging marker of Alzheimer's disease, for clarifying the interactions between the white matter and cognitive dysfunction with respect to the APOE4 status.
Collapse
Affiliation(s)
- Mina Park
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea.,Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hong Pyo Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea
| | - Junghyeob Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea
| | - Dong Hyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea
| | - Yeonsil Moon
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Won-Jin Moon
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Dallaire-Théroux C, Saikali S, Richer M, Potvin O, Duchesne S. Histopathological Analysis of Cerebrovascular Lesions Associated With Aging. J Neuropathol Exp Neurol 2021; 81:97-105. [PMID: 34875082 DOI: 10.1093/jnen/nlab125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cerebrovascular disease (CVD) has been associated with cognitive impairment. Yet, our understanding of vascular contribution to cognitive decline has been limited by heterogeneity of definitions and assessment, as well as its occurrence in cognitively healthy aging. Therefore, we aimed to establish the natural progression of CVD associated with aging. We conducted a retrospective observational study of 63 cognitively healthy participants aged 19-84 years selected through the histological archives of the CHU de Québec. Assessment of CVD lesions was performed independently by 3 observers blinded to clinical data using the Vascular Cognitive Impairment Neuropathology Guidelines (VCING). We found moderate to almost perfect interobserver agreement for most regional CVD scores. Atherosclerosis (ρ = 0.758) and arteriolosclerosis (ρ = 0.708) showed the greatest significant association with age, followed by perivascular hemosiderin deposits (ρ = 0.432) and cerebral amyloid angiopathy (CAA; ρ = 0.392). Amyloid and tau pathologies were both associated with higher CVD load, but only CAA remained significantly associated with amyloid plaques after controlling for age. Altogether, these findings support the presence of multiple CVD lesions in the brains of cognitively healthy adults, the burden of which increases with age and can be quantified in a reproducible manner using standardized histological scales such as the VCING.
Collapse
Affiliation(s)
- Caroline Dallaire-Théroux
- From the CERVO Brain Research Center, Quebec City, Quebec, Canada (CD-T, OP, SD); Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (CD-T, SS, MR); Department of Neurological Sciences, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (CD-T); Department of Pathology, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (SS, MR); and Department of Radiology and nuclear medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (SD)
| | - Stephan Saikali
- From the CERVO Brain Research Center, Quebec City, Quebec, Canada (CD-T, OP, SD); Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (CD-T, SS, MR); Department of Neurological Sciences, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (CD-T); Department of Pathology, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (SS, MR); and Department of Radiology and nuclear medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (SD)
| | - Maxime Richer
- From the CERVO Brain Research Center, Quebec City, Quebec, Canada (CD-T, OP, SD); Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (CD-T, SS, MR); Department of Neurological Sciences, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (CD-T); Department of Pathology, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (SS, MR); and Department of Radiology and nuclear medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (SD)
| | - Olivier Potvin
- From the CERVO Brain Research Center, Quebec City, Quebec, Canada (CD-T, OP, SD); Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (CD-T, SS, MR); Department of Neurological Sciences, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (CD-T); Department of Pathology, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (SS, MR); and Department of Radiology and nuclear medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (SD)
| | - Simon Duchesne
- From the CERVO Brain Research Center, Quebec City, Quebec, Canada (CD-T, OP, SD); Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (CD-T, SS, MR); Department of Neurological Sciences, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (CD-T); Department of Pathology, Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada (SS, MR); and Department of Radiology and nuclear medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada (SD)
| |
Collapse
|
17
|
Paolini Paoletti F, Simoni S, Parnetti L, Gaetani L. The Contribution of Small Vessel Disease to Neurodegeneration: Focus on Alzheimer's Disease, Parkinson's Disease and Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22094958. [PMID: 34066951 PMCID: PMC8125719 DOI: 10.3390/ijms22094958] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 01/18/2023] Open
Abstract
Brain small vessel disease (SVD) refers to a variety of structural and functional changes affecting small arteries and micro vessels, and manifesting as white matter changes, microbleeds and lacunar infarcts. Growing evidence indicates that SVD might play a significant role in the neurobiology of central nervous system (CNS) neurodegenerative disorders, namely Alzheimer's disease (AD) and Parkinson's disease (PD), and neuroinflammatory diseases, such as multiple sclerosis (MS). These disorders share different pathophysiological pathways and molecular mechanisms (i.e., protein misfolding, derangement of cellular clearance systems, mitochondrial impairment and immune system activation) having neurodegeneration as biological outcome. In these diseases, the actual contribution of SVD to the clinical picture, and its impact on response to pharmacological treatments, is not known yet. Due to the high frequency of SVD in adult-aged patients, it is important to address this issue. In this review, we report preclinical and clinical data on the impact of SVD in AD, PD and MS, with the main aim of clarifying the predictability of SVD on clinical manifestations and treatment response.
Collapse
|
18
|
Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, Kalaria RN, Katsumata Y, Gold BT, Wang DJJ, Ma SJ, Shade LMP, Fardo DW, Hartz AMS, Jicha GA, Nelson KB, Magaki SD, Schmitt FA, Teylan MA, Ighodaro ET, Phe P, Abner EL, Cykowski MD, Van Eldik LJ, Nelson PT. Brain arteriolosclerosis. Acta Neuropathol 2021; 141:1-24. [PMID: 33098484 PMCID: PMC8503820 DOI: 10.1007/s00401-020-02235-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Brain arteriolosclerosis (B-ASC), characterized by pathologic arteriolar wall thickening, is a common finding at autopsy in aged persons and is associated with cognitive impairment. Hypertension and diabetes are widely recognized as risk factors for B-ASC. Recent research indicates other and more complex risk factors and pathogenetic mechanisms. Here, we describe aspects of the unique architecture of brain arterioles, histomorphologic features of B-ASC, relevant neuroimaging findings, epidemiology and association with aging, established genetic risk factors, and the co-occurrence of B-ASC with other neuropathologic conditions such as Alzheimer's disease and limbic-predominant age-related TDP-43 encephalopathy (LATE). There may also be complex physiologic interactions between metabolic syndrome (e.g., hypertension and inflammation) and brain arteriolar pathology. Although there is no universally applied diagnostic methodology, several classification schemes and neuroimaging techniques are used to diagnose and categorize cerebral small vessel disease pathologies that include B-ASC, microinfarcts, microbleeds, lacunar infarcts, and cerebral amyloid angiopathy (CAA). In clinical-pathologic studies that factored in comorbid diseases, B-ASC was independently associated with impairments of global cognition, episodic memory, working memory, and perceptual speed, and has been linked to autonomic dysfunction and motor symptoms including parkinsonism. We conclude by discussing critical knowledge gaps related to B-ASC and suggest that there are probably subcategories of B-ASC that differ in pathogenesis. Observed in over 80% of autopsied individuals beyond 80 years of age, B-ASC is a complex and under-studied contributor to neurologic disability.
Collapse
Affiliation(s)
- Brittney L Blevins
- Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Seth Love
- University of Bristol and Southmead Hospital, Bristol, BS10 5NB, UK
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- LIM-22, Department of Pathology, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Samantha J Ma
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Lincoln M P Shade
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University Kentucky, Lexington, KY, 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | | | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, University Washington, Seattle, WA, 98105, USA
| | | | - Panhavuth Phe
- Sanders-Brown Center on Aging, University Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, Department of Epidemiology, University Kentucky, Lexington, KY, 40536, USA
| | - Matthew D Cykowski
- Departments of Pathology and Genomic Medicine and Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA.
- Rm 311 Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
19
|
Kanagasingam S, Chukkapalli SS, Welbury R, Singhrao SK. Porphyromonas gingivalis is a Strong Risk Factor for Alzheimer's Disease. J Alzheimers Dis Rep 2020; 4:501-511. [PMID: 33532698 PMCID: PMC7835991 DOI: 10.3233/adr-200250] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is one of the several important bacterial pathogens associated with the sporadic Alzheimer’s disease (AD). Different serotypes are either capsulated or are non-capsulated. It has been demonstrated that P. gingivalis (non-capsulated) can reproduce the neurodegenerative AD-like changes in vitro, and a capsular P. gingivalis (strain W83) could reproduce the cardinal hallmark lesions of AD in a wild-type mouse model. All P. gingivalis forms express proteolytically active proteases that enable cleavage of the amyloid-β protin precursor (AβPP) and tau resulting in the formation of amyloid-β and neurofibrillary tangles. Tau is an established substrate for gingipains, which can cleave tau into various peptides. Some of the P. gingivalis fragmented tau protein peptides contain “VQIINK” and “VQIVYK” hexapeptide motifs which map to the flanking regions of the microtubule binding domains and are also found in paired helical filaments that form NFTs. P. gingivalis can induce peripheral inflammation in periodontitis and can also initiate signaling pathways that activate kinases, which in turn, phosphorylate neuronal tau. Periodontal disease related inflammation has metabolic implications for an individual’s peripheral and brain health as patients suffering from generalized periodontitis often have related co-morbidities and are “at risk” of developing AD. The aim here is to discuss the role of P. gingivalis behind such associations with the backdrop of huge efforts to test P. gingivalis virulence factors clinically (GAIN Trial: Phase 2/3 Study of COR388 in Subjects with AD) with inhibitors, which may lead to an intervention by reducing the pathogenic bacterial load.
Collapse
Affiliation(s)
- Shalini Kanagasingam
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Sasanka S Chukkapalli
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Richard Welbury
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Sim K Singhrao
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| |
Collapse
|
20
|
Lao PJ, Gutierrez J, Keator D, Rizvi B, Banerjee A, Igwe KC, Laing KK, Sathishkumar M, Moni F, Andrews H, Krinsky-McHale S, Head E, Lee JH, Lai F, Yassa MA, Rosas HD, Silverman W, Lott IT, Schupf N, Brickman AM. Alzheimer-Related Cerebrovascular Disease in Down Syndrome. Ann Neurol 2020; 88:1165-1177. [PMID: 32944999 PMCID: PMC7729262 DOI: 10.1002/ana.25905] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Adults with Down syndrome (DS) develop Alzheimer disease (AD) pathology by their 5th decade. Compared with the general population, traditional vascular risks in adults with DS are rare, allowing examination of cerebrovascular disease in this population and insight into its role in AD without the confound of vascular risk factors. We examined in vivo magnetic resonance imaging (MRI)-based biomarkers of cerebrovascular pathology in adults with DS, and determined their cross-sectional relationship with age, beta-amyloid pathology, and mild cognitive impairment or clinical AD diagnostic status. METHODS Participants from the Biomarkers of Alzheimer's Disease in Down Syndrome study (n = 138, 50 ± 7 years, 39% women) with MRI data and a subset (n = 90) with amyloid positron emission tomography (PET) were included. We derived MRI-based biomarkers of cerebrovascular pathology, including white matter hyperintensities (WMH), infarcts, cerebral microbleeds, and enlarged perivascular spaces (PVS), as well as PET-based biomarkers of amyloid burden. Participants were characterized as cognitively stable (CS), mild cognitive impairment-DS (MCI-DS), possible AD dementia, or definite AD dementia based on in-depth assessments of cognition, function, and health status. RESULTS There were detectable WMH, enlarged PVS, infarcts, and microbleeds as early as the 5th decade of life. There was a monotonic increase in WMH volume, enlarged PVS, and presence of infarcts across diagnostic groups (CS < MCI-DS < possible AD dementia < definite AD dementia). Higher amyloid burden was associated with a higher likelihood of an infarct. INTERPRETATION The findings highlight the prevalence of cerebrovascular disease in adults with DS and add to a growing body of evidence that implicates cerebrovascular disease as a core feature of AD and not simply a comorbidity. ANN NEUROL 2020;88:1165-1177.
Collapse
Affiliation(s)
- Patrick J Lao
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - José Gutierrez
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - David Keator
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Batool Rizvi
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Arit Banerjee
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Kay C Igwe
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Krystal K Laing
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Mithra Sathishkumar
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Fahmida Moni
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Howard Andrews
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY
| | - Sharon Krinsky-McHale
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY
| | - Elizabeth Head
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - Joseph H Lee
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | - H Diana Rosas
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA
- Department of Radiology, Athinoula Martinos Center, Massachusetts General Hospital, Harvard University, Charlestown, MA
| | - Wayne Silverman
- Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Ira T Lott
- Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Nicole Schupf
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Adam M Brickman
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
21
|
Wan X, Gan C, You C, Fan T, Zhang S, Zhang H, Wang S, Shu K, Wang X, Lei T. Association of APOE ε4 with progressive hemorrhagic injury in patients with traumatic intracerebral hemorrhage. J Neurosurg 2020; 133:496-503. [PMID: 31323634 DOI: 10.3171/2019.4.jns183472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/18/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The intracranial hematoma volume in patients with traumatic brain injury is a key parameter for the determination of the management approach and outcome. Apolipoprotein E (APOE) ε4 is reported to be a risk factor for larger hematoma volume, which might contribute to a poor outcome. However, whether APOE ε4 is related to progressive hemorrhagic injury (PHI), a common occurrence in the clinical setting, remains unclear. In this study, the authors aimed to investigate the association between the APOE genotype and occurrence of PHI. METHODS This prospective study included a cohort of 123 patients with traumatic intracerebral hemorrhage who initially underwent conservative treatment. These patients were assigned to the PHI or non-PHI group according to the follow-up CT scan. A polymerase chain reaction and sequencing method were carried out to determine the APOE genotype. Multivariate logistic regression analysis was applied to identify predictors of PHI. RESULTS The overall frequency of the alleles was as follows: E2/2, 0%; E2/3, 14.6%; E3/3, 57.8%; E2/4, 2.4%; E3/4, 22.8%; and E4/4, 2.4%. Thirty-four patients carried at least one allele of ε4. In this study 60 patients (48.8%) experienced PHI, and the distribution of the alleles was as follows: E2/2, 0%; E2/3, 5.7%; E3/3, 22.8%; E2/4, 2.4%; E3/4, 16.3%; and E4/4, 1.6%, which was significantly different from that in the non-PHI group (p = 0.008). Additionally, the late operation rate in the PHI group was significantly higher than that in the non-PHI group (24.4% vs 11.4%, p = 0.002). Multivariate logistic regression identified APOE ε4 (OR 5.14, 95% CI 2.40-11.62), an elevated international normalized ratio (OR 3.57, 95% CI 1.61-8.26), and higher glucose level (≥ 10 mmol/L) (OR 3.88, 95% CI 1.54-10.77) as independent risk factors for PHI. Moreover, APOE ε4 was not a risk factor for the coagulopathy and outcome of the patients with traumatic intracerebral hemorrhage. CONCLUSIONS The presence of APOE ε4, an elevated international normalized ratio, and a higher glucose level (≥ 10 mmol/L) are predictors of PHI. Additionally, APOE ε4 is not associated with traumatic coagulopathy and patient outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kai Shu
- 1Department of Neurosurgery and
| | - Xiong Wang
- 2Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | |
Collapse
|
22
|
Gurinovich A, Andersen SL, Puca A, Atzmon G, Barzilai N, Sebastiani P. Varying Effects of APOE Alleles on Extreme Longevity in European Ethnicities. J Gerontol A Biol Sci Med Sci 2020; 74:S45-S51. [PMID: 31724059 DOI: 10.1093/gerona/glz179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Indexed: 12/19/2022] Open
Abstract
APOE is a well-studied gene with multiple effects on aging and longevity. The gene has three alleles: e2, e3, and e4, whose frequencies vary by ethnicity. While the e2 is associated with healthy cognitive aging, the e4 allele is associated with Alzheimer's disease and early mortality and therefore its prevalence among people with extreme longevity (EL) is low. Using the PopCluster algorithm, we identified several ethnically different clusters in which the effect of the e2 and e4 alleles on EL changed substantially. For example, PopCluster discovered a large group of 1,309 subjects enriched of Southern Italian genetic ancestry with weaker protective effect of e2 (odds ratio [OR] = 1.27, p = .14) and weaker damaging effect of e4 (OR = 0.82, p = .31) on the phenotype of EL compared to other European ethnicities. Further analysis of this cluster suggests that the odds for EL in carriers of the e4 allele with Southern Italian genetic ancestry differ depending on whether they live in the United States (OR = 0.29, p = .009) or Italy (OR = 1.21, p = .38). PopCluster also found clusters enriched of subjects with Danish ancestry with varying effect of e2 on EL. The country of residence (Denmark or United States) appears to change the odds for EL in the e2 carriers.
Collapse
Affiliation(s)
- Anastasia Gurinovich
- Bioinformatics Program, Boston University, Massachusetts.,Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | | | - Annibale Puca
- Department of Medicine and Surgery, University of Salerno, Fisciano, SA, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Sesto San Giovanni, MI, Italy
| | - Gil Atzmon
- Faculty of Natural Science, University of Haifa, Israel.,Albert Einstein College of Medicine, Bronx, New York
| | - Nir Barzilai
- Albert Einstein College of Medicine, Bronx, New York
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| |
Collapse
|
23
|
Ii Y, Ishikawa H, Matsuyama H, Shindo A, Matsuura K, Yoshimaru K, Satoh M, Taniguchi A, Matsuda K, Umino M, Maeda M, Tomimoto H. Hypertensive Arteriopathy and Cerebral Amyloid Angiopathy in Patients with Cognitive Decline and Mixed Cerebral Microbleeds. J Alzheimers Dis 2020; 78:1765-1774. [PMID: 33185609 PMCID: PMC11062589 DOI: 10.3233/jad-200992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Hypertensive arteriopathy (HA) and cerebral amyloid angiopathy (CAA) may contribute to the development of mixed cerebral microbleeds (CMBs). Recently, the total small vessel disease (SVD) scores for HA and CAA were proposed, which are determined by a combination of MRI markers to reflect overall severity of these microangiopathies. OBJECTIVE We investigated whether or not total HA-SVD and CAA-SVD scores could be used to predict overlap of HA and CAA in patients with mixed CMBs. METHODS Fifty-three subjects with mixed CMBs were retrospectively analyzed. MRI markers (CMBs, lacunes, perivascular space, white matter hyperintensity [WMH] and cortical superficial siderosis [cSS]) were assessed. The HA-SVD score and CAA-SVD score were obtained for each subject. Anterior or posterior WMH was also assessed using the age-related white matter changes scale. RESULTS The two scores were positively correlated (ρ= 0.449, p < 0.001). The prevalence of lobar dominant CMB distribution (p < 0.001) and lacunes in the centrum semiovale (p < 0.001) and the severity of WMH in the parieto-occipital lobes (p = 0.004) were significantly higher in the high CAA-SVD score group. cSS was found in four patients with high CAA-SVD score who showed lobar-dominant CMB distribution and severe posterior WMH. CONCLUSION Mixed CMBs are mainly due to HA. Assessing both two scores may predict the overlap of HA and CAA in individuals with mixed CMBs. Patients with a high CAA-SVD score may have some degree of advanced CAA, especially when lobar predominant CMBs, severe posterior WMH, lobar lacunes, or cSS are observed.
Collapse
Affiliation(s)
- Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hirofumi Matsuyama
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Keita Matsuura
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Kimiko Yoshimaru
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayuki Satoh
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Mie, Japan
| | - Akira Taniguchi
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Kana Matsuda
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Maki Umino
- Department of Radiology, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayuki Maeda
- Department of Neuroradiology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
24
|
Volgman AS, Bairey Merz CN, Aggarwal NT, Bittner V, Bunch TJ, Gorelick PB, Maki P, Patel HN, Poppas A, Ruskin J, Russo AM, Waldstein SR, Wenger NK, Yaffe K, Pepine CJ. Sex Differences in Cardiovascular Disease and Cognitive Impairment: Another Health Disparity for Women? J Am Heart Assoc 2019; 8:e013154. [PMID: 31549581 PMCID: PMC6806032 DOI: 10.1161/jaha.119.013154] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Neelum T Aggarwal
- Departments of Neurological Sciences Rush Alzheimer's Disease Center Rush Medical College Chicago IL
| | - Vera Bittner
- Division of Cardiovascular Disease Department of Medicine University of Alabama at Birmingham AL
| | - T Jared Bunch
- Section of Cardiology Department of Medicine Stanford University Palo Alto California.,Department of Cardiology Intermountain Heart Institute Intermountain Medical Center Salt Lake City UT
| | - Philip B Gorelick
- Department of Translational Neuroscience Michigan State University College of Human Medicine Grand Rapids MI
| | - Pauline Maki
- Department of Psychiatry, Psychology and Obstetrics & Gynecology University of Illinois at Chicago IL
| | - Hena N Patel
- Section of Cardiology Department of Medicine Rush Medical College Chicago IL
| | - Athena Poppas
- Section of Cardiology Department of Medicine Brown University School of Medicine Providence RI
| | - Jeremy Ruskin
- Division of Cardiology Massachusetts General Hospital Boston MA
| | - Andrea M Russo
- Section of Cardiology Department of Medicine Cooper Medical School of Rowan University Camden NJ
| | - Shari R Waldstein
- Department of Psychology University of Maryland, Baltimore County Baltimore MD
| | - Nanette K Wenger
- Section of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Kristine Yaffe
- Department of Psychiatry, Neurology and Epidemiology University of California San Francisco San Francisco CA
| | - Carl J Pepine
- Division of Cardiovascular Medicine Department of Medicine University of Florida Gainesville FL
| |
Collapse
|
25
|
Ben-Moshe H, Luz I, Liraz O, Boehm-Cagan A, Salomon-Zimri S, Michaelson D. ApoE4 Exacerbates Hippocampal Pathology Following Acute Brain Penetration Injury in Female Mice. J Mol Neurosci 2019; 70:32-44. [DOI: 10.1007/s12031-019-01397-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/14/2019] [Indexed: 10/26/2022]
|
26
|
Bown CW, Liu D, Osborn KE, Gupta DK, Mendes LA, Pechman KR, Hohman TJ, Wang TJ, Gifford KA, Jefferson AL. Apolipoprotein E Genotype Modifies the Association Between Cardiac Output and Cognition in Older Adults. J Am Heart Assoc 2019; 8:e011146. [PMID: 31364446 PMCID: PMC6761646 DOI: 10.1161/jaha.118.011146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Subtle reductions in cardiac output relate to lower cerebral blood flow, especially in regions where Alzheimer's disease pathology first develops. Apolipoprotein E (APOE)‐ε4 is a genetic susceptibility risk factor for Alzheimer's disease that also moderates vascular damage. This study investigated whether APOE‐ε4 carrier status modifies the cross‐sectional association between cardiac output and cognition. Methods and Results Vanderbilt Memory & Aging Project participants free of clinical stroke and dementia (n=306, 73±7 years, 42% female) underwent echocardiography to determine cardiac output (L/min), comprehensive neuropsychological assessment, and venous blood draw to determine APOE genotype and ε4 carrier status. Linear regressions related cardiac output to neuropsychological test performance, adjusting for age, sex, education, race/ethnicity, body surface area, cognitive diagnosis, Framingham Stroke Risk Profile, and APOE‐ε4 status. Main effect models were null (P>0.19). With identical covariates, models were repeated testing a cardiac output×APOE‐ε4 status interaction and again stratified by ε4 carrier status. Cardiac output×APOE‐ε4 status related to naming (β=0.91, P=0.0009), category fluency (β=1.2, P=0.01), information processing speed (β=−5.4, P=0.001), visuospatial skill (β=0.85, P=0.003), and executive function performances (β=0.22, P=0.002). Stratified models suggested that lower cardiac output was associated with worse neuropsychological performances among APOE‐ε4 carriers. Conclusions APOE‐ε4 carrier status appears to modify the cross‐sectional association between cardiac output and neuropsychological performance such that lower cardiac output relates to poorer performances among carriers of the ε4 allele. These findings add to increasing evidence that APOE‐ε4 carrier status has important implications for associations between vascular and brain health in aging adults.
Collapse
Affiliation(s)
- Corey W Bown
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN
| | - Dandan Liu
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN.,Department of Biostatistics Vanderbilt University Medical Center Nashville TN
| | - Katie E Osborn
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN
| | - Deepak K Gupta
- Division of Cardiovascular Medicine Department of Medicine Vanderbilt University Medical Center Nashville TN.,Vanderbilt Heart Imaging Core Lab (V-HICL) Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University Medical Center Nashville TN
| | - Lisa A Mendes
- Division of Cardiovascular Medicine Department of Medicine Vanderbilt University Medical Center Nashville TN
| | - Kimberly R Pechman
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN
| | - Timothy J Hohman
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN.,Vanderbilt Genetics Institute Vanderbilt University Medical Center Nashville TN
| | - Thomas J Wang
- Division of Cardiovascular Medicine Department of Medicine Vanderbilt University Medical Center Nashville TN
| | - Katherine A Gifford
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN
| | - Angela L Jefferson
- Department of Neurology Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center Nashville TN.,Division of Cardiovascular Medicine Department of Medicine Vanderbilt University Medical Center Nashville TN
| |
Collapse
|
27
|
Polygenic impact of common genetic risk loci for Alzheimer's disease on cerebral blood flow in young individuals. Sci Rep 2019; 9:467. [PMID: 30679549 PMCID: PMC6345995 DOI: 10.1038/s41598-018-36820-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/15/2018] [Indexed: 11/15/2022] Open
Abstract
Genome-wide association studies (GWAS) show that many common alleles confer risk for developing Alzheimer’s disease (AD). These risk loci may contribute to MRI alterations in young individuals, preceding the clinical manifestations of AD. Prior evidence identifies vascular dysregulation as the earliest marker of disease progression. However, it remains unclear whether cerebrovascular function (measured via grey-matter cerebral blood flow (gmCBF)) is altered in young individuals with increased AD genetic risk. We establish relationships between gmCBF with APOE and AD polygenic risk score in a young cohort (N = 75; aged: 19–32). Genetic risk was assessed via a) possessing at least one copy of the APOE ɛ4 allele and b) a polygenic risk score (AD-PRS) estimated from AD-GWAS. We observed a reduction in gmCBF in APOE ɛ4 carriers and a negative relationship between AD-PRS and gmCBF. We further found regional reductions in gmCBF in individuals with higher AD-PRS across the frontal cortex (PFWE < 0.05). Our findings suggest that a larger burden of AD common genetic risk alleles is associated with attenuated cerebrovascular function, during young adulthood. These results suggest that cerebral vasculature is a mechanism by which AD risk alleles confer susceptibility.
Collapse
|
28
|
Sebastiani P, Gurinovich A, Nygaard M, Sasaki T, Sweigart B, Bae H, Andersen SL, Villa F, Atzmon G, Christensen K, Arai Y, Barzilai N, Puca A, Christiansen L, Hirose N, Perls TT. APOE Alleles and Extreme Human Longevity. J Gerontol A Biol Sci Med Sci 2019; 74:44-51. [PMID: 30060062 PMCID: PMC6298189 DOI: 10.1093/gerona/gly174] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
We assembled a collection of 28,297 participants from seven studies of longevity and healthy aging comprising New England Centenarian, Long Life Family, Longevity Gene Population, Southern Italian Centenarian, Japanese Centenarian, the Danish Longevity, and the Health and Retirement Studies to investigate the association between the APOE alleles ε2ε3 and ε4 and extreme human longevity and age at death. By using three different genetic models and two definitions of extreme longevity based on either a threshold model or age at death, we show that ε4 is associated with a substantially decreased odds for extreme longevity, and increased risk for death that persists even beyond ages reached by less than 1% of the population. We also show that carrying the ε2ε2 or ε2ε3 genotype is associated with significantly increased odds to reach extreme longevity, with decreased risk for death compared with carrying the genotype ε3ε3 but with only a modest reduction in risk for death beyond an age reached by less than 1% of the population.
Collapse
Affiliation(s)
- Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Anastasia Gurinovich
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- Bioinformatics Program, Boston University, Massachusetts
| | - Marianne Nygaard
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Takashi Sasaki
- Center for Supercentenarian Research, Keio University, Tokyo, Japan
| | - Benjamin Sweigart
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Harold Bae
- College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Stacy L Andersen
- Geriatric Section, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Francesco Villa
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
| | - Gil Atzmon
- Faculty of Natural Science, University of Haifa, Israel
- Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Kaare Christensen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Yasumichi Arai
- Center for Supercentenarian Research, Keio University, Tokyo, Japan
| | - Nir Barzilai
- Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Annibale Puca
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Lene Christiansen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Nobuyoshi Hirose
- Center for Supercentenarian Research, Keio University, Tokyo, Japan
| | - Thomas T Perls
- Geriatric Section, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| |
Collapse
|
29
|
Abstract
Population-based clinic-pathological studies have established that the most common pathological substrate of dementia in community-dwelling elderly people is mixed, especially Alzheimer's disease (AD) and cerebrovascular ischemic disease (CVID), rather than pure AD. While these could be just two frequent unrelated comorbidities in the elderly, epidemiological research has reinforced the idea that mid-life (age <65 years) vascular risk factors increase the risk of late-onset (age ≥ 65 years) dementia, and specifically AD. By contrast, healthy lifestyle choices such as leisure activities, physical exercise, and Mediterranean diet are considered protective against AD. Remarkably, several large population-based longitudinal epidemiological studies have recently indicated that the incidence and prevalence of dementia might be decreasing in Western countries. Although it remains unclear whether these positive trends are attributable to neuropathologically definite AD versus CVID, based on these epidemiological data it has been estimated that a sizable proportion of AD cases could be preventable. In this review, we discuss the current evidence about modifiable risk factors for AD derived from epidemiological, preclinical, and interventional studies, and analyze the opportunities for therapeutic and preventative interventions.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John H. Growdon
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Lamar M, Yu L, Rubin LH, James BD, Barnes LL, Farfel JM, Gaiteri C, Buchman AS, Bennett DA, Schneider JA. APOE genotypes as a risk factor for age-dependent accumulation of cerebrovascular disease in older adults. Alzheimers Dement 2018; 15:258-266. [PMID: 30321502 PMCID: PMC6368888 DOI: 10.1016/j.jalz.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/15/2018] [Accepted: 08/21/2018] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Apolipoprotein E (APOE) is a susceptibility gene for late-onset Alzheimer's disease neuropathology; less is known about the relationship between APOE and cerebrovascular disease (CVD) neuropathology. METHODS We investigated associations of APOE status with arteriolosclerosis, macroinfarcts and microinfarcts, and atherosclerosis in 1383 adults (65.9-108.2 years at death) with and without dementia. Excluding ε2/ε4 carriers, multivariable regressions for each CVD-related neuropathology compared ε4 and ε2 carriers to ε3/ε3 carriers adjusting for confounders including age and Alzheimer's neuropathology. RESULTS Three hundred forty-two individuals (24.7%; ∼87.7 years at death; 39.9% nondemented) were ε3/ε4 or ε4/ε4, and 180 (13.0%; ∼89.9 years at death; 66.6% nondemented) were ε2/ε3 or ε2/ε2. ε4 carriers had higher odds of macroinfarcts (odds ratio = 1.41, 95% confidence interval: 1.02-1.94, P = .03), whereas ε2 carriers had higher odds of moderate-to-severe arteriolosclerosis (odds ratio = 1.68, 95% confidence interval: 1.15-2.45, P = .006) compared to ε3/ε3 carriers. Age-stratified analyses suggested that these relationships were driven by ε4 carriers <90 years at death and ε2 carriers ≥90 years at death, respectively. DISCUSSION APOE differentially affects type and timing of CVD-related neuropathology.
Collapse
Affiliation(s)
- Melissa Lamar
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA.
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Leah H Rubin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bryan D James
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Jose Marcelo Farfel
- Department of Geriatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
31
|
Rastogi P, Kumar N, Ahluwalia J, Das R, Varma N, Suri V, Senee H. Thrombophilic risk factors are laterally associated with Apolipoprotein E gene polymorphisms in deep vein thrombosis patients: An Indian study. Phlebology 2018; 34:324-335. [PMID: 30282515 DOI: 10.1177/0268355518802693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Deep vein thrombosis is a multifactorial disease with many acquired and genetic risk factors. Polymorphism in the APOE gene is an upcoming potential pathogenic factor whose role is unclear in deep vein thrombosis. METHODS An equal number of deep vein thrombosis cases and controls (N = 100, each) were investigated for APOE gene polymorphisms along with known acquired and hereditable thrombophilic risk factors. APOE genotyping was done by polymerase chain reaction. RESULTS The ε3/ε4 and ε2/ε3 APOE genotypes were commoner in deep vein thrombosis cases than controls but not statistically significant ( ε3/ε4 → 18% versus 11%, OR = 1.776, CI = 0.792-3.984, p = 0.16; ε2/ε3 →10% versus 9%, OR = 1.123, CI = 0.436-2.895, p = 0.809). However, the following risk factors were found to be laterally associated with APOE genotypes in cases of deep vein thrombosis: pregnancy with ε2/ε3 genotype positivity (N = 29; p = 0.019), recurrent pregnancy loss with ε3/ε3 genotype (N = 29; p = 0.016), normal antithrombin levels with ε3/ε3 genotype (N = 62; p = 0.03) and non-O blood group with ε3/ε4 genotype (N = 100; p = 0.023). CONCLUSION APOE genotypes have shown only a modest association with deep vein thrombosis and were not statistically significant. A lateral association of these genotypes with thrombophilic risk factors was observed which may be investigated further for the possible pathogenetic mechanisms and their therapeutic implications.
Collapse
Affiliation(s)
- Pulkit Rastogi
- 1 Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Narender Kumar
- 2 Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jasmina Ahluwalia
- 2 Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reena Das
- 2 Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- 2 Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vikas Suri
- 3 Department of Internal Medicine, Nehru Hospital, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harikishan Senee
- 2 Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
32
|
Hartmann DA, Hyacinth HI, Liao FF, Shih AY. Does pathology of small venules contribute to cerebral microinfarcts and dementia? J Neurochem 2018; 144:517-526. [PMID: 28950410 PMCID: PMC5869083 DOI: 10.1111/jnc.14228] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 09/08/2017] [Accepted: 09/18/2017] [Indexed: 12/28/2022]
Abstract
Microinfarcts are small, but strikingly common, ischemic brain lesions in the aging human brain. There is mounting evidence that microinfarcts contribute to vascular cognitive impairment and dementia, but the origins of microinfarcts are unclear. Understanding the vascular pathologies that cause microinfarcts may yield strategies to prevent their occurrence and reduce their deleterious effects on brain function. Current thinking suggests that cortical microinfarcts arise from the occlusion of penetrating arterioles, which are responsible for delivering oxygenated blood to small volumes of tissue. Unexpectedly, pre-clinical studies have shown that the occlusion of penetrating venules, which drain deoxygenated blood from cortex, lead to microinfarcts that appear identical to those resulting from arteriole occlusion. Here we discuss the idea that cerebral venule pathology could be an overlooked source for brain microinfarcts in humans. This article is part of the Special Issue "Vascular Dementia". Cover Image for this Issue: doi: 10.1111/jnc.14167.
Collapse
Affiliation(s)
- David A. Hartmann
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Hyacinth I. Hyacinth
- Aflac Cancer and Blood Disorder Center, Children’s Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Andy Y. Shih
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
33
|
Kresge HA, Khan OA, Wagener MA, Liu D, Terry JG, Nair S, Cambronero FE, Gifford KA, Osborn KE, Hohman TJ, Pechman KR, Bell SP, Wang TJ, Carr JJ, Jefferson AL. Subclinical Compromise in Cardiac Strain Relates to Lower Cognitive Performances in Older Adults. J Am Heart Assoc 2018; 7:e007562. [PMID: 29440034 PMCID: PMC5850190 DOI: 10.1161/jaha.117.007562] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/03/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Global longitudinal strain (GLS), reflecting total shortening of the myocardium during the cardiac cycle, has emerged as a more precise myocardial function measure than left ventricular ejection fraction (LVEF). Longitudinal strain may be selectively affected in subclinical heart disease, even in the presence of normal LVEF. This study examines subclinical cardiac dysfunction, assessed by GLS and LVEF, and cognition among older adults. METHODS AND RESULTS Vanderbilt Memory and Aging Project participants who were free of clinical dementia, stroke, and heart failure (n=318, 73±7 years, 58% male) completed neuropsychological assessment and cardiac magnetic resonance to quantify GLS and LVEF. Linear regression models related GLS and LVEF to neuropsychological performances, adjusting for age, sex, race/ethnicity, education, Framingham Stroke Risk Profile, cognitive diagnosis, and APOE*ε4 status. Models were repeated with a cardiac×cognitive diagnosis interaction term. Compromised GLS (reflected by higher values) related to worse naming (β=-0.07, P=0.04), visuospatial immediate recall (β=-0.83, P=0.03), visuospatial delayed recall (β=-0.22, P=0.03), and verbal delayed recall (β=-0.11, P=0.007). LVEF did not relate to worse performance on any measure (P>0.18). No diagnostic interactions were observed. CONCLUSIONS Our study results are among the first to suggest that compromised GLS relates to worse episodic memory and language performance among older adults who are free of clinical dementia, stroke, and heart failure. Subclinical cardiac dysfunction may correlate with cognitive health in late life, even when LVEF remains normal. The results add to growing evidence that GLS may be a more sensitive and preferred method for quantifying subclinical changes in cardiac function.
Collapse
Affiliation(s)
- Hailey A Kresge
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Omair A Khan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Madison A Wagener
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
- Department of Psychology, Vanderbilt University, Nashville, TN
| | - Dandan Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - James G Terry
- Radiology& Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Sangeeta Nair
- Radiology& Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Francis E Cambronero
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Katherine A Gifford
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Katie E Osborn
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Kimberly R Pechman
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
| | - Susan P Bell
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of General Internal Medicine, Department of Medicine, Center for Quality Aging, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - John Jeffrey Carr
- Radiology& Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Angela L Jefferson
- Department of Neurology, Vanderbilt Memory & Alzheimer's Center Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
34
|
Wijesinghe P, Shankar SK, Yasha TC, Gorrie C, Amaratunga D, Hulathduwa S, Kumara KS, Samarasinghe K, Suh YH, Steinbusch HWM, De Silva KRD. Vascular Contributions in Alzheimer's Disease-Related Neuropathological Changes: First Autopsy Evidence from a South Asian Aging Population. J Alzheimers Dis 2018; 54:1607-1618. [PMID: 27589527 DOI: 10.3233/jad-160425] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Evidence from various consortia on vascular contributions has been inconsistent in determining the etiology of sporadic Alzheimer's disease (AD). OBJECTIVE To investigate vascular risk factors and cerebrovascular pathologies associated in manifestation of AD-related neuropathological changes of an elderly population. METHODS Postmortem brain samples from 76 elderly subjects (≥50 years) were used to study genetic polymorphisms, intracranial atherosclerosis of the circle of Willis (IASCW), and microscopic infarcts in deep white matters. From this cohort, 50 brains (≥60 years) were subjected to neuropathological diagnosis using immunohistopathological techniques. RESULTS Besides the association with age, the apolipoprotein E ɛ4 allele was significantly and strongly associated with Thal amyloid-β phases ≥1 [odds ratio (OR) = 6.76, 95% confidence interval (CI) 1.37-33.45] and inversely with Braak neurofibrillary tangle (NFT) stages ≥III (0.02, 0.0-0.47). Illiterates showed a significant positive association for Braak NFT stages ≥IV (14.62, 1.21-176.73) and a significant negative association for microscopic infarcts (0.15, 0.03-0.71) in deep white matters. With respect to cerebrovascular pathologies, cerebral small vessel lesions (white matter hyperintensities and cerebral amyloid angiopathy) showed a higher degree of associations among them and with AD-related neuropathological changes (p < 0.05) compared to large vessel pathology (IASCW), which showed a significant association only with Braak NFT stages ≥I (p = 0.050). CONCLUSION These findings suggest that besides age, education, and genetic factors, other vascular risk factors were not associated with AD-related neuropathological changes and urge prompt actions be taken against cerebral small vessel diseases since evidence for effective prevention is still lacking.
Collapse
Affiliation(s)
- Printha Wijesinghe
- Interdisciplinary Center for Innovation in Biotechnology & Neuroscience, Genetic Diagnostic and Research Laboratory, Department of Anatomy, Faculty of Medical Sciences, University of Srijayewardenepura, Nugegoda, Sri Lanka
| | - S K Shankar
- Department of Neuropathology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - T C Yasha
- Department of Neuropathology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Catherine Gorrie
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, Australia
| | | | - Sanjayah Hulathduwa
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Srijayewardenepura, Nugegoda, Sri Lanka
| | - K Sunil Kumara
- Department of Judicial Medical Office, Colombo South Teaching Hospital, Colombo, Sri Lanka
| | - Kamani Samarasinghe
- Department of Pathology, University of Srijayewardenepura, Nugegoda, Sri Lanka
| | - Yoo-Hun Suh
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, Korea.,NRI, Gachon University, Incheon, South Korea
| | - Harry W M Steinbusch
- Department of Translational Neuroscience, Faculty Health, Medicine & Life Sciences, Maastricht University, Maastricht, Netherlands
| | - K Ranil D De Silva
- Interdisciplinary Center for Innovation in Biotechnology & Neuroscience, Genetic Diagnostic and Research Laboratory, Department of Anatomy, Faculty of Medical Sciences, University of Srijayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
35
|
Neuropathologic features of TOMM40 '523 variant on late-life cognitive decline. Alzheimers Dement 2017; 13:1380-1388. [PMID: 28624335 PMCID: PMC5723540 DOI: 10.1016/j.jalz.2017.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/03/2017] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The study investigated the role of neuropathologies in the relationship between TOMM40 '523 genotype and late-life cognitive decline. METHODS Participants were community-dwelling older persons who had annual cognitive assessments and brain autopsies after death. Genotyping used DNA from peripheral blood or postmortem brain tissue. Linear mixed models assessed the extent to which the association of '523 genotype with cognitive decline is attributable to neuropathologies. RESULTS Relative to ε3/ε3 homozygotes with '523-S/VL or '523-VL/VL genotype, both '523-L carriers and ε3/ε3 homozygotes with '523-S/S genotype had faster cognitive decline. The association of '523-L with cognitive decline was attenuated and no longer significant after controlling for Alzheimer's and other neuropathologies. By contrast, the association of '523-S/S was unchanged. DISCUSSION There are two distinct TOMM40 '523 signals in relation to late-life cognitive decline. One signal primarily acts through AD and other common neuropathologies, whereas the other operates through a different mechanism.
Collapse
|
36
|
Hohman TJ, Dumitrescu L, Oksol A, Wagener M, Gifford KA, Jefferson AL. APOE allele frequencies in suspected non-amyloid pathophysiology (SNAP) and the prodromal stages of Alzheimer's Disease. PLoS One 2017; 12:e0188501. [PMID: 29190651 PMCID: PMC5708777 DOI: 10.1371/journal.pone.0188501] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/08/2017] [Indexed: 01/14/2023] Open
Abstract
Biomarker definitions for preclinical Alzheimer’s disease (AD) have identified individuals with neurodegeneration (ND+) without β-amyloidosis (Aβ-) and labeled them with suspected non-AD pathophysiology (SNAP). We evaluated Apolipoprotein E (APOE) ε2 and ε4 allele frequencies across biomarker definitions—Aβ-/ND- (n = 268), Aβ+/ND- (n = 236), Aβ-/ND+ or SNAP (n = 78), Aβ+/ND+ (n = 204)—hypothesizing that SNAP would have an APOE profile comparable to Aβ-/ND-. Using AD Neuroimaging Initiative data (n = 786, 72±7 years, 48% female), amyloid status (Aβ+ or Aβ-) was defined by cerebrospinal fluid (CSF) Aβ-42 levels, and neurodegeneration status (ND+ or ND-) was defined by hippocampal volume from MRI. Binary logistic regression related biomarker status to APOE ε2 and ε4 allele carrier status, adjusting for age, sex, education, and cognitive diagnosis. Compared to the biomarker negative (Aβ-/ND-) participants, higher proportions of ε4 and lower proportions of ε2 carriers were observed among Aβ+/ND- (ε4: OR = 6.23, p<0.001; ε2: OR = 0.53, p = 0.03) and Aβ+/ND+ participants (ε4: OR = 12.07, p<0.001; ε2: OR = 0.29, p = 0.004). SNAP participants were statistically comparable to biomarker negative participants (p-values>0.30). In supplemental analyses, comparable results were observed when coding SNAP using amyloid imaging and when using CSF tau levels. In contrast to APOE, a polygenic risk score for AD that excluded APOE did not show an association with amyloidosis or neurodegeneration (p-values>0.15), but did show an association with SNAP defined using CSF tau (β = 0.004, p = 0.02). Thus, in a population with low levels of cerebrovascular disease and a lower prevalence of SNAP than the general population, APOE and known genetic drivers of AD do not appear to contribute to the neurodegeneration observed in SNAP. Additional work in population based samples is needed to better elucidate the genetic contributors to various etiological drivers of SNAP.
Collapse
Affiliation(s)
- Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- * E-mail:
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Amy Oksol
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Madison Wagener
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Katherine A. Gifford
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | | |
Collapse
|
37
|
White CC, Yang HS, Yu L, Chibnik LB, Dawe RJ, Yang J, Klein HU, Felsky D, Ramos-Miguel A, Arfanakis K, Honer WG, Sperling RA, Schneider JA, Bennett DA, De Jager PL. Identification of genes associated with dissociation of cognitive performance and neuropathological burden: Multistep analysis of genetic, epigenetic, and transcriptional data. PLoS Med 2017; 14:e1002287. [PMID: 28441426 PMCID: PMC5404753 DOI: 10.1371/journal.pmed.1002287] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The molecular underpinnings of the dissociation of cognitive performance and neuropathological burden are poorly understood, and there are currently no known genetic or epigenetic determinants of the dissociation. METHODS AND FINDINGS "Residual cognition" was quantified by regressing out the effects of cerebral pathologies and demographic characteristics on global cognitive performance proximate to death. To identify genes influencing residual cognition, we leveraged neuropathological, genetic, epigenetic, and transcriptional data available for deceased participants of the Religious Orders Study (n = 492) and the Rush Memory and Aging Project (n = 487). Given that our sample size was underpowered to detect genome-wide significance, we applied a multistep approach to identify genes influencing residual cognition, based on our prior observation that independent genetic and epigenetic risk factors can converge on the same locus. In the first step (n = 979), we performed a genome-wide association study with a predefined suggestive p < 10-5, and nine independent loci met this threshold in eight distinct chromosomal regions. Three of the six genes within 100 kb of the lead SNP are expressed in the dorsolateral prefrontal cortex (DLPFC): UNC5C, ENC1, and TMEM106B. In the second step, in the subset of participants with DLPFC DNA methylation data (n = 648), we found that residual cognition was related to differential DNA methylation of UNC5C and ENC1 (false discovery rate < 0.05). In the third step, in the subset of participants with DLPFC RNA sequencing data (n = 469), brain transcription levels of UNC5C and ENC1 were evaluated for their association with residual cognition: RNA levels of both UNC5C (estimated effect = -0.40, 95% CI -0.69 to -0.10, p = 0.0089) and ENC1 (estimated effect = 0.0064, 95% CI 0.0033 to 0.0096, p = 5.7 × 10-5) were associated with residual cognition. In secondary analyses, we explored the mechanism of these associations and found that ENC1 may be related to the previously documented effect of depression on cognitive decline, while UNC5C may alter the composition of presynaptic terminals. Of note, the TMEM106B allele identified in the first step as being associated with better residual cognition is in strong linkage disequilibrium with rs1990622A (r2 = 0.66), a previously identified protective allele for TDP-43 proteinopathy. Limitations include the small sample size for the genetic analysis, which was underpowered to detect genome-wide significance, the evaluation being limited to a single cortical region for epigenetic and transcriptomic data, and the use of categorical measures for certain non-amyloid-plaque, non-neurofibrillary-tangle neuropathologies. CONCLUSIONS Through a multistep analysis of cognitive, neuropathological, genomic, epigenomic, and transcriptomic data, we identified ENC1 and UNC5C as genes with convergent genetic, epigenetic, and transcriptomic evidence supporting a potential role in the dissociation of cognition and neuropathology in an aging population, and we expanded our understanding of the TMEM106B haplotype that is protective against TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Charles C. White
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Hyun-Sik Yang
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lei Yu
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Lori B. Chibnik
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Robert J. Dawe
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Jingyun Yang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Hans-Ulrich Klein
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel Felsky
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alfredo Ramos-Miguel
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
| | - William G. Honer
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reisa A. Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America
- Center for Translational & Systems Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
38
|
Coutu JP, Lindemer ER, Konukoglu E, Salat DH. Two distinct classes of degenerative change are independently linked to clinical progression in mild cognitive impairment. Neurobiol Aging 2017; 54:1-9. [PMID: 28286328 DOI: 10.1016/j.neurobiolaging.2017.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 01/15/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022]
Abstract
We previously demonstrated 2 statistically distinct factors of degeneration in Alzheimer's disease: one strongly related to white matter damage and age interpreted as "age- and vascular-related", and the other related to cortical atrophy thought to represent "neurodegenerative changes associated with Alzheimer's disease". Those factors are now replicated in a distinct cross-sectional data set of 364 participants from the Alzheimer's Disease Neuroimaging Initiative and their interpretation is improved using correlations with CSF biomarkers. Furthermore, we now show that changes in both factors over 2 years are independently associated with decline in Mini-Mental State Examination score in a longitudinal subset of 116 individuals with mild cognitive impairment. Progression in the "age- and vascular-related" factor was greater for individuals with 2 APOE ε4 alleles and linked to a greater attributable change in Mini-Mental State Examination than the "neurodegenerative" factor. These results suggest benefits of targeting white matter and vascular health to complement interventions focused on the neurodegenerative aspect of the disease, even in individuals with little discernable vascular comorbidity.
Collapse
Affiliation(s)
- Jean-Philippe Coutu
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Emily R Lindemer
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ender Konukoglu
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David H Salat
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Departments of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston, MA, USA
| | | |
Collapse
|
39
|
Kalheim LF, Selnes P, Bjørnerud A, Coello C, Vegge K, Fladby T. Amyloid Dysmetabolism Relates to Reduced Glucose Uptake in White Matter Hyperintensities. Front Neurol 2016; 7:209. [PMID: 27917152 PMCID: PMC5116462 DOI: 10.3389/fneur.2016.00209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/08/2016] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and cause of dementia and is characterized by amyloid plaques and neurofibrillary tangles. AD has traditionally been considered to primarily affect gray matter, but multiple lines of evidence also indicate white matter (WM) pathology and associated small-vessel cerebrovascular disease. WM glucose delivery and metabolism may have implications for local tissue integrity, and [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) may be helpful to assess neuroglial and axonal function in WM. Hypothesizing that affection of oligodendroglia will be associated with loss of glucose uptake, we aimed to investigate glucose metabolism in magnetic resonance imaging (MRI) white matter hyperintensities (WMHs) and normal-appearing WM in patients with and without evidence of amyloid plaques. Subjects with mild cognitive impairment or subjective cognitive decline were included and dichotomized according to pathological (Aβ+) or normal (Aβ−) concentrations of cerebrospinal fluid amyloid-β 1–42. A total of 50 subjects were included, of whom 30 subjects were classified as Aβ(+) and 20 subjects as Aβ(−). All subjects were assessed with MRI and FDG-PET. FDG-PET images were corrected for effects of partial voluming and normalized to cerebellar WM, before determining WMH FDG-uptake. Although there were no significant differences between the groups in terms of age, WMH volume, number of individual WMHs, or WMH distribution, we found significantly lower (p = 0.021) FDG-uptake in WMHs in Aβ(+) subjects (mean = 0.662, SD = 0.113) compared to Aβ(−) subjects (mean = 0.596, SD = 0.073). There were no significant group differences in the FDG-uptake in normal-appearing WM. Similar results were obtained without correction for effects of partial voluming. Our findings add to the evidence for a link between Aβ dysmetabolism and WM pathology in AD.
Collapse
Affiliation(s)
- Lisa Flem Kalheim
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Per Selnes
- Department of Neurology, Akershus University Hospital , Lørenskog , Norway
| | - Atle Bjørnerud
- The Intervention Centre, Oslo University Hospital , Oslo , Norway
| | - Christopher Coello
- Preclinical PET/CT, Institute of Basic Medical Sciences, University of Oslo , Oslo , Norway
| | - Kjetil Vegge
- Department of Radiology, Akershus University Hospital , Lørenskog , Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Salameh TS, Rhea EM, Banks WA, Hanson AJ. Insulin resistance, dyslipidemia, and apolipoprotein E interactions as mechanisms in cognitive impairment and Alzheimer's disease. Exp Biol Med (Maywood) 2016; 241:1676-83. [PMID: 27470930 PMCID: PMC4999626 DOI: 10.1177/1535370216660770] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An increased risk for Alzheimer's disease is associated with dyslipidemia and insulin resistance. A separate literature shows the genetic risk for developing Alzheimer's disease is strongly correlated to the presence of the E4 isoform of the apolipoprotein E carrier protein. Understanding how apolipoprotein E carrier protein, lipids, amyloid β peptides, glucose, central nervous system insulin, and peripheral insulin interact with one another in Alzheimer's disease is an area of increasing interest. Here, we will review the evidence relating apolipoprotein E carrier protein, lipids, and insulin action to Alzheimer's disease and Aβ peptides and then propose mechanisms as to how these factors might interact with one another to impair cognition and promote Alzheimer's disease.
Collapse
Affiliation(s)
- Therese S Salameh
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth M Rhea
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William A Banks
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Angela J Hanson
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
41
|
Zhang XX, Pan YH, Huang YM, Zhao HL. Neuroendocrine hormone amylin in diabetes. World J Diabetes 2016; 7:189-97. [PMID: 27162583 PMCID: PMC4856891 DOI: 10.4239/wjd.v7.i9.189] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/16/2016] [Accepted: 04/05/2016] [Indexed: 02/05/2023] Open
Abstract
The neuroendocrine hormone amylin, also known as islet amyloid polypeptide, is co-localized, co-packaged and co-secreted with insulin from adult pancreatic islet β cells to maintain glucose homeostasis. Specifically, amylin reduces secretion of nutrient-stimulated glucagon, regulates blood pressure with an effect on renin-angiotensin system, and delays gastric emptying. The physiological actions of human amylin attribute to the conformational α-helix monomers whereas the misfolding instable oligomers may be detrimental to the islet β cells and further transform to β-sheet fibrils as amyloid deposits. No direct evidence proves that the amylin fibrils in amyloid deposits cause diabetes. Here we also have performed a systematic review of human amylin gene changes and reported the S20G mutation is minor in the development of diabetes. In addition to the metabolic effects, human amylin may modulate autoimmunity and innate inflammation through regulatory T cells to impact on both human type 1 and type 2 diabetes.
Collapse
|
42
|
Kapasi A, Schneider JA. Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:878-86. [PMID: 26769363 PMCID: PMC11062590 DOI: 10.1016/j.bbadis.2015.12.023] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/29/2015] [Accepted: 12/29/2015] [Indexed: 12/27/2022]
Abstract
There is growing evidence suggesting that vascular pathologies and dysfunction play a critical role in cognitive impairment, clinical Alzheimer's disease, and dementia. Vascular pathologies such as macroinfarcts, microinfarcts, microbleeds, small and large vessel cerebrovascular disease, and white matter disease are common especially in the brains of older persons where they contribute to cognitive impairment and lower the dementia threshold. Vascular dysfunction resulting in decreased cerebral blood flow, and abnormalities in the blood brain barrier may also contribute to the Alzheimer's disease (AD) pathophysiologic process and AD dementia. This review provides a clinical-pathological perspective on the role of vessel disease, vascular brain injury, alterations of the neurovascular unit, and mixed pathologies in the Alzheimer's disease pathophysiologic process and Alzheimer's dementia. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- A Kapasi
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S. Paulina Street, IL 60612, Chicago, USA.
| | - J A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S. Paulina Street, IL 60612, Chicago, USA.
| |
Collapse
|
43
|
Therkelsen KE, Preis SR, Beiser A, DeCarli C, Seshadri S, Wolf P, Au R, Fox CS. Neck Circumference, Brain Imaging Measures, and Neuropsychological Testing Measures. J Stroke Cerebrovasc Dis 2016; 25:1570-1581. [PMID: 27056441 DOI: 10.1016/j.jstrokecerebrovasdis.2016.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 02/10/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Perivascular fat may have direct effects on local vascularity. Neck fat is associated with carotid intimal thickness, a predictor of brain aging outcomes. This study investigated whether neck circumference, an estimation of neck fat, has unique associations with brain aging outcomes. METHODS The study sample (n = 2082, 53.5% women, mean age 60.9 years) was derived from Framingham Heart Study participants with brain magnetic resonance imaging (MRI) and neuropsychological (NP) test measures. Multivariable-adjusted regressions examined cross-sectional associations of neck circumference with brain MRI and NP test measures. Models were also constructed with waist circumference and body mass index (BMI) as exposures. RESULTS A 1 standard deviation (2.8 cm [women]; 2.9 cm [men]) increment in neck circumference was associated with lower total cerebral brain volume (β = -.22, P = .0006) and lower frontal brain volume (β = -.55, P < .0001). However, a similar association was observed for both waist circumference and BMI. There were no associations between neck circumference and NP test measures after full covariate adjustment. CONCLUSIONS There were no unique associations between neck circumference and brain MRI or NP measures. Consistent with prior observations, all adiposity measures showed associations with more adverse brain MRI and NP measures, suggesting a global association of generalized adiposity.
Collapse
Affiliation(s)
- Kate E Therkelsen
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Boston University School of Medicine, Boston, Massachusetts
| | - Sarah R Preis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, California
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Philip Wolf
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Caroline S Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Division of Endocrinology and Metabolism, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
44
|
Hays CC, Zlatar ZZ, Wierenga CE. The Utility of Cerebral Blood Flow as a Biomarker of Preclinical Alzheimer's Disease. Cell Mol Neurobiol 2016; 36:167-79. [PMID: 26898552 PMCID: PMC5278904 DOI: 10.1007/s10571-015-0261-z] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/25/2015] [Indexed: 12/20/2022]
Abstract
There is accumulating evidence suggesting that changes in brain perfusion are present long before the clinical symptoms of Alzheimer's disease (AD), perhaps even before amyloid-β accumulation or brain atrophy. This evidence, consistent with the vascular hypothesis of AD, implicates cerebral blood flow (CBF) in the pathogenesis of AD and suggests its utility as a biomarker of preclinical AD. The extended preclinical phase of AD holds particular significance for disease modification, as treatment would likely be most effective in this early asymptomatic stage of disease. This highlights the importance of identifying reliable and accurate biomarkers of AD that can differentiate normal aging from preclinical AD prior to clinical symptom manifestation. Cerebral perfusion, as measured by arterial spin labeling magnetic resonance imaging (ASL-MRI), has been shown to distinguish between normal controls and adults with AD. In addition to demonstrating diagnostic utility, CBF has shown usefulness as a tool for identifying those who are at risk for AD and for predicting subtle cognitive decline and conversion to mild cognitive impairment and AD. Taken together, this evidence not only implicates CBF as a useful biomarker for tracking disease severity and progression, but also suggests that ASL-measured CBF may be useful for identifying candidates for future AD treatment trials, especially in the preclinical, asymptomatic phases of the disease.
Collapse
Affiliation(s)
- Chelsea C Hays
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 92161, USA
- SDSU/UC San Diego Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Court, Suite 103, San Diego, CA, 92120, USA
| | - Zvinka Z Zlatar
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Christina E Wierenga
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 92161, USA.
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA.
| |
Collapse
|
45
|
Cacciottolo M, Christensen A, Moser A, Liu J, Pike CJ, Smith C, LaDu MJ, Sullivan PM, Morgan TE, Dolzhenko E, Charidimou A, Wahlund LO, Wiberg MK, Shams S, Chiang GCY, Finch CE. The APOE4 allele shows opposite sex bias in microbleeds and Alzheimer's disease of humans and mice. Neurobiol Aging 2016; 37:47-57. [PMID: 26686669 PMCID: PMC4687024 DOI: 10.1016/j.neurobiolaging.2015.10.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/06/2015] [Accepted: 10/11/2015] [Indexed: 12/21/2022]
Abstract
The apolipoprotein APOE4 allele confers greater risk of Alzheimer's disease (AD) for women than men, in conjunction with greater clinical deficits per unit of AD neuropathology (plaques, tangles). Cerebral microbleeds, which contribute to cognitive dysfunctions during AD, also show APOE4 excess, but sex-APOE allele interactions are not described. We report that elderly men diagnosed for mild cognitive impairment and AD showed a higher risk of cerebral cortex microbleeds with APOE4 allele dose effect in 2 clinical cohorts (ADNI and KIDS). Sex-APOE interactions were further analyzed in EFAD mice carrying human APOE alleles and familial AD genes (5XFAD (+/-) /human APOE(+/+)). At 7 months, E4FAD mice had cerebral cortex microbleeds with female excess, in contrast to humans. Cerebral amyloid angiopathy, plaques, and soluble Aβ also showed female excess. Both the cerebral microbleeds and cerebral amyloid angiopathy increased in proportion to individual Aβ load. In humans, the opposite sex bias of APOE4 allele for microbleeds versus the plaques and tangles is the first example of organ-specific, sex-linked APOE allele effects, and further shows AD as a uniquely human condition.
Collapse
Affiliation(s)
- Mafalda Cacciottolo
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Christensen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Alexandra Moser
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jiahui Liu
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Conor Smith
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University, Durham VA Medical Center and GRECC, Durham, NC, USA
| | - Todd E Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Egor Dolzhenko
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Andreas Charidimou
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Division of Clinical Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Kristofferson Wiberg
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Shams
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Caleb E Finch
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA; Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Moser VA, Pike CJ. Obesity and sex interact in the regulation of Alzheimer's disease. Neurosci Biobehav Rev 2015; 67:102-18. [PMID: 26708713 DOI: 10.1016/j.neubiorev.2015.08.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, for which a number of genetic, environmental, and lifestyle risk factors have been identified. A significant modifiable risk factor is obesity in mid-life. Interestingly, both obesity and AD exhibit sex differences and are regulated by sex steroid hormones. Accumulating evidence suggests interactions between obesity and sex in regulation of AD risk, although the pathways underlying this relationship are unclear. Inflammation and the E4 allele of apolipoprotein E have been identified as independent risk factors for AD and both interact with obesity and sex steroid hormones. We review the individual and cooperative effects of obesity and sex on development of AD and examine the potential contributions of apolipoprotein E, inflammation, and their interactions to this relationship.
Collapse
Affiliation(s)
- V Alexandra Moser
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA.
| | - Christian J Pike
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
47
|
Vascular disease and risk factors are associated with cognitive decline in the alzheimer disease spectrum. Alzheimer Dis Assoc Disord 2015; 29:18-25. [PMID: 24787033 DOI: 10.1097/wad.0000000000000043] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We investigated the relationship between vascular disease and risk factors versus cognitive decline cross-sectionally and longitudinally in normal older control, mild cognitive impairment, and mild Alzheimer disease (AD) dementia subjects. A total of 812 participants (229 normal older control, 395 mild cognitive impairment, 188 AD) underwent cognitive testing, brain magnetic resonance imaging, and clinical evaluations at baseline and over a period of 3 years. General linear, longitudinal mixed-effects, and Cox proportional hazards models were used. Greater homocysteine level and white matter hyperintensity volume were associated with processing speed impairment (homocysteine: P=0.02; white matter hyperintensity: P<0.0001); greater Vascular Index score was associated with memory impairment (P=0.007); and greater number of apolipoprotein E ε4 (APOE4) alleles was associated with global cognitive impairment (P=0.007) at baseline. Apolipoprotein E ε4 was associated with greater rate of increase in global cognitive impairment (P=0.002) and processing speed impairment (P=0.001) over time, whereas higher total cholesterol was associated with greater rate of increase in global cognitive impairment (P=0.02) and memory impairment (P=0.06) over time. These results suggest a significant association of increased vascular disease and risk factors with cognitive impairment at baseline and over time in the AD spectrum in a sample that was selected to have low vascular burden at baseline.
Collapse
|
48
|
Serrano-Pozo A, Qian J, Monsell SE, Betensky RA, Hyman BT. APOEε2 is associated with milder clinical and pathological Alzheimer disease. Ann Neurol 2015; 77:917-29. [PMID: 25623662 DOI: 10.1002/ana.24369] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/19/2015] [Accepted: 06/06/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The Alzheimer disease (AD) APOEε4 risk allele associates with an earlier age at onset and increased amyloid-β deposition, whereas the protective APOEε2 allele delays the onset and appears to prevent amyloid-β deposition. Yet the clinical and pathological effects of APOEε2 remain uncertain because of its relative rarity. We investigated the effects of APOEε2 and ε4 alleles on AD pathology and cognition in a large US data set of well-characterized AD patients. METHODS We studied individuals from the National Alzheimer's Coordinating Center autopsy cohort across the entire clinicopathological continuum of AD. Multivariate models were built to examine the associations between APOE alleles and AD neuropathological changes, using the APOEε3/ε3 group as comparator. Mediation analysis was used to estimate the direct and indirect effects of APOE alleles on AD pathology and cognition (Clinical Dementia Rating Sum of Boxes and Mini-Mental State Examination). RESULTS Compared to APOEε3/ε3, APOEε2 is independently associated with lower Braak neurofibrillary tangle (NFT) stages and possibly fewer neuritic plaques, but has no direct effect on cerebral amyloid angiopathy (CAA) severity, whereas APOEε4 is associated with more neuritic plaques and CAA, but has no independent effect on Braak NFT stage. Unadjusted analyses showed marked differences among APOE genotypes with respect to cognitive performance (ε2 > ε3 > ε4). Mediation analysis suggests that this is largely explained through effects on pathology. INTERPRETATION Even when adjusted for age at onset, symptom duration, and other demographic variables, APOEε2 is associated with milder AD pathology and less severe antemortem cognitive impairment compared to APOEε3 and ε4 alleles, suggesting a relative neuroprotective effect of APOEε2 in AD.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA.,Massachusetts Alzheimer Disease Research Center, Charlestown, MA.,Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA
| | - Jing Qian
- Division of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA
| | - Sarah E Monsell
- National Alzheimer's Coordinating Center and Department of Epidemiology, University of Washington, Seattle, WA
| | - Rebecca A Betensky
- Massachusetts Alzheimer Disease Research Center, Charlestown, MA.,Department of Biostatistics, Harvard School of Public Health, Boston, MA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA.,Massachusetts Alzheimer Disease Research Center, Charlestown, MA
| |
Collapse
|
49
|
Wierenga CE, Hays CC, Zlatar ZZ. Cerebral blood flow measured by arterial spin labeling MRI as a preclinical marker of Alzheimer's disease. J Alzheimers Dis 2015; 42 Suppl 4:S411-9. [PMID: 25159672 DOI: 10.3233/jad-141467] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is growing recognition that cerebral hypoperfusion is related to the pathogenesis of Alzheimer's disease (AD), implicating the measurement of cerebral blood flow (CBF) as a possible biomarker of AD. The ability to identify the earliest and most reliable markers of incipient cognitive decline and clinical symptoms is critical to develop effective preventive strategies and interventions for AD. Arterial spin labeling (ASL) magnetic resonance imaging (MRI) measures CBF by magnetically labeling arterial water and using it as an endogenous tracer. Studies using ASL MRI in humans indicate that CBF changes are present several years before the development of the clinical symptoms of AD. Moreover, ASL-measured CBF has been shown to distinguish between cognitively normal individuals, adults at risk for AD, and persons diagnosed with AD. Some studies indicate that CBF may even be sensitive for predicting cognitive decline and conversion to mild cognitive impairment and AD over time. Taken together, evidence suggests that the current staging models of AD biomarker pathology should incorporate early changes in CBF as a useful biomarker, possibly present even earlier than amyloid-β accumulation. Though still a research tool, ASL imaging is a promising non-invasive and reliable method with the potential to serve as a future clinical tool for the measurement of CBF in preclinical AD.
Collapse
Affiliation(s)
- Christina E Wierenga
- VA San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | | | - Zvinka Z Zlatar
- VA San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
50
|
Jefferson AL, Gifford KA, Damon S, Chapman GW, Liu D, Sparling J, Dobromyslin V, Salat D. Gray & white matter tissue contrast differentiates Mild Cognitive Impairment converters from non-converters. Brain Imaging Behav 2015; 9:141-8. [PMID: 24493370 PMCID: PMC4146750 DOI: 10.1007/s11682-014-9291-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The clinical relevance of gray/white matter contrast ratio (GWR) in mild cognitive impairment (MCI) remains unknown. This study examined baseline GWR and 3-year follow-up diagnostic status in MCI. Alzheimer's Disease Neuroimaging Initiative MCI participants with baseline 1.5 T MRI and 3-year follow-up clinical data were included. Participants were categorized into two groups based on 3-year follow-up diagnoses: 1) non-converters (n = 69, 75 ± 7, 26 % female), and 2) converters (i.e., dementia at follow-up; n = 69, 75 ± 7, 30 % female) who were matched on baseline age and Mini-Mental State Examination scores. Groups were compared on FreeSurfer generated baseline GWR from structural images in which higher values represent greater tissue contrast. A general linear model, adjusting for APOE-status, scanner type, hippocampal volume, and cortical thickness, revealed that converters evidenced lower GWR values than non-converters (i.e., more degradation in tissue contrast; p = 0.03). Individuals with MCI who convert to dementia have lower baseline GWR values than individuals who remain diagnostically stable over a 3-year period, statistically independent of cortical thickness or hippocampal volume.
Collapse
Affiliation(s)
- Angela L Jefferson
- Vanderbilt Memory & Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, 2525 West End Avenue, 12th Floor - Suite 1200, Nashville, TN, 37203, USA,
| | | | | | | | | | | | | | | |
Collapse
|