1
|
Bruce SS, Zhang C, Liberman AL, Merkler AE, Navi BB, Chiang GC, Iadecola C, Kamel H, Murthy SB. Prevalence of Cerebral Amyloid Angiopathy and Associated Risk of Subsequent Ischemic and Hemorrhagic Stroke and Mortality in a Nationwide Cohort. Ann Neurol 2025. [PMID: 40309957 DOI: 10.1002/ana.27253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE There are limited population-based data regarding the prevalence of cerebral amyloid angiopathy (CAA) and associated risks of mortality and incident cerebrovascular events. METHODS We performed a retrospective cohort study using inpatient and outpatient claims from 2008 to 2022 from a 5% national sample of Medicare beneficiaries. CAA and ischemic and hemorrhagic stroke were identified using validated International Classification of Diseases 10th Revision (ICD-10) codes. We ascertained CAA from October 1, 2015 through 2022, and used data from 2008 through September 30, 2015 to ascertain comorbidities including prevalent stroke. We used Cox regression to examine the association of CAA with subsequent death and incident stroke subtypes after adjustment for demographics, vascular risk factors, and Charlson comorbidities. RESULTS Among 1,920,312 Medicare beneficiaries in our sample, 2,161 (11.3 per 10,000) had a diagnosis of CAA. In adjusted Cox regression analysis, there was an association between CAA and subsequent mortality (HR 4.9; 95% CI 4.6-5.2). Among 1,872,474 patients without prevalent stroke, including 900 of the CAA patients, there was a significant association between CAA and an increased risk of any stroke (HR 8.0; 95% CI 6.7-9.6), ischemic stroke (HR 4.6; 95% CI 3.6-6.0), intracerebral hemorrhage (HR 26.9; 95% CI 20.3-35.6), and subarachnoid hemorrhage (HR 21.6; 95% CI 12.2-38.1). After a diagnosis of CAA, absolute risks of ischemic stroke and intracerebral hemorrhage were broadly similar. INTERPRETATION In a large, nationwide cohort of Medicare beneficiaries, the prevalence of clinically diagnosed CAA was approximately 11 per 10,000. CAA was associated with an increased risk of mortality and incident stroke, both hemorrhagic and ischemic. ANN NEUROL 2025.
Collapse
Affiliation(s)
- Samuel S Bruce
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Cenai Zhang
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ava L Liberman
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Alexander E Merkler
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Babak B Navi
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Gloria C Chiang
- Brain Health Imaging Institute and Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Costantino Iadecola
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Santosh B Murthy
- Clinical and Translational Neuroscience Unit, Department of Neurology and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
2
|
Starmans NL, Leeuwis AE, Bennink E, Meyer Viol SL, Golla SS, Dankbaar JW, Bron EE, Biessels GJ, Kappelle LJ, van der Flier WM, Tolboom N. Dynamic PET imaging in patients with unilateral carotid occlusion shows lateralized cerebral hypoperfusion, but no amyloid binding. J Alzheimers Dis 2025:13872877251329593. [PMID: 40241519 DOI: 10.1177/13872877251329593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BackgroundCarotid occlusive disease is a risk factor for cognitive decline. A possible underlying etiology is that hemodynamic impairment results in decreased cerebral perfusion, exacerbated amyloid-β accumulation (Aβ) and poorer cognitive performance.ObjectiveWe aimed to determine whether patients with unilateral internal carotid artery (ICA) occlusion have less cerebral perfusion and more Aβ in the ipsilateral than in the contralateral hemisphere, and whether perfusion and Aβ are associated with cognitive functioning.MethodsWe included 20 patients (age 67.2 ± 7.0 years, 8 females, MMSE 29 [27-29]) with unilateral ICA occlusion, which underwent neuropsychological assessment and dynamic 18F-Florbetaben positron emission tomography (PET). Global and regional relative perfusion (R1) and binding potential (BPND) were obtained from the PET-images using a simplified reference tissue model. We performed Wilcoxon signed-rank tests to examine differences between hemispheres within subjects and linear regression to investigate associations with cognitive functioning.ResultsMedian global R1 was 0.911 (0.883-0.950) and global BPND was 0.172 (0.129-0.187). R1 was lower in the hemisphere ipsilateral to the ICA occlusion than in the contralateral hemisphere (0.899 [0.876-0.921] versus 0.935 [0.889-0.970]). BPND did not differ significantly between hemispheres (ipsilateral 0.172 [0.124-0.181] versus contralateral 0.168 [0.137-0.191]). Neither cerebral perfusion nor Aβ burden were associated with cognitive functioning.ConclusionsPatients with unilateral ICA occlusion did not have more Aβ in the ipsilateral hemisphere than in the contralateral hemisphere despite ipsilateral hypoperfusion. Perfusion and Aβ were unrelated to cognitive functioning. This indicates that cognitive impairment in patients with ICA occlusion is not due to exacerbated Aβ accumulation.
Collapse
Affiliation(s)
- Naomi Lp Starmans
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna E Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Medical Psychology, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Edwin Bennink
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sebastiaan L Meyer Viol
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Sandeep Sv Golla
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Jan Willem Dankbaar
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esther E Bron
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Khowdiary MM, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhenawy AA, Rashwan EK, Alexiou A, Papadakis M, Fetoh MEAE, Batiha GES. The Peripheral Amyloid-β Nexus: Connecting Alzheimer's Disease with Atherosclerosis through Shared Pathophysiological Mechanisms. Neuromolecular Med 2025; 27:20. [PMID: 40032716 DOI: 10.1007/s12017-025-08836-2] [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: 11/13/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two chronic diseases with seemingly distinct pathologies. However, emerging research points to a bidirectional relationship driven by common mechanisms, such as inflammation, oxidative stress, and dysregulation of Amyloid-Beta (Aβ). This review focuses on the role of Aβ as a critical molecular link between AD and AS, emphasizing its contribution to neuronal impairment and vascular damage. Specifically, peripheral Aβ produced in the pancreas and skeletal muscle tissues exacerbates AS by promoting endothelial dysfunction and insulin resistance (IR). Furthermore, AS accelerates AD progression by impairing cerebral blood flow and inducing chronic hypoxia, causing Aβ accumulation. This review critically evaluates recent findings, highlighting inconsistencies in clinical studies and suggesting future research directions. Understanding the bidirectional influence of AD and AS could pave the way for novel therapeutic approaches targeting shared molecular pathways, particularly emphasizing Aβ clearance and inflammation.
Collapse
Affiliation(s)
- Manal M Khowdiary
- Department of Chemistry, Faculty of Applied Science, Lieth Collage, Umm Al-Qura University, 24382, Makkah, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO. Box13, Kufa, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
- Chemistry Department, Faculty of Science, AlBaha University, 65731, Al Bahah, Saudi Arabia
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Jouf University, Akaka, Saudi Arabia
| | - Athanasios Alexiou
- Department of Research & Development, Funogen, 11741, Athens, Attiki, Greece
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
| | - Marios Papadakis
- University Hospital, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammed E Abo-El Fetoh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
4
|
Huang S, Nunez J, Toresco DL, Wen C, Slotabec L, Wang H, Zhang H, Rouhi N, Adenawoola MI, Li J. Alterations in the inflammatory homeostasis of aging-related cardiac dysfunction and Alzheimer's diseases. FASEB J 2025; 39:e70303. [PMID: 39758048 DOI: 10.1096/fj.202402725rr] [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: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Alzheimer's disease (AD) is well known among the elderly and has a profound impact on both patients and their families. Increasing research indicates that AD is a systemic disease, with a strong connection to cardiovascular disease. They share common genetic factors, such as mutations in the presenilin (PS1 and PS2) and the apolipoprotein E (APOE) genes. Cardiovascular conditions can lead to reduced cerebral blood flow and increased oxidative stress. These factors contribute to the accumulation of Aβ plaques and the formation of abnormal tau protein tangles, which are both key pathological features of AD. Additionally, Aβ deposits and abnormal protein responses have been observed in cardiomyocytes as well as in peripheral tissues. The toxic Aβ deposition intensifies damage to the microvascular structure associated with blood-brain barrier disruption and the initiation of neuroinflammation, which may accelerate the onset of neurocognitive deficits and cardiovascular dysfunction. Thus, we discuss the main mechanisms linking AD and cardiac dysfunction to enhance our understanding of these conditions. Ultimately, insights into the brain-heart axis may help us develop effective treatment strategies in the future.
Collapse
Affiliation(s)
- Shuli Huang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeremiah Nunez
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Dai Lan Toresco
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Haibei Zhang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
5
|
Takahashi MKN, Paradela RS, Grinberg LT, Leite REP, Farias-Itao DS, Paes VR, Braga ME, Naslavsky MS, Zatz M, Jacob-Filho W, Nitrini R, Pasqualucci CA, Suemoto CK. Hypertension may associate with cerebral small vessel disease and infarcts through the pathway of intracranial atherosclerosis. Neurobiol Aging 2025; 145:84-95. [PMID: 39541803 PMCID: PMC11864294 DOI: 10.1016/j.neurobiolaging.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Hypertension, a major modifiable risk factor for cardiovascular diseases, is linked to late-life neurocognitive disorders such as vascular dementia and Alzheimer's disease (AD). This study explores the associations between hypertension, intracranial atherosclerotic disease (ICAD), cerebral small vessel disease (cSVD), and Alzheimer's disease neuropathologic change (ADNC) in a large community-based autopsy study. This cross-sectional study used data from the Biobank for Aging Studies of the University of São Paulo Medical School. Sociodemographic and clinical information was gathered from a reliable next-of-kin informant. Neurofibrillary tangles, neuritic plaques, lacunar infarcts, hyaline arteriolosclerosis, and cerebral amyloid angiopathy were evaluated. Causal mediation analyses with natural effect models were performed to examine indirect associations of hypertension with cerebrovascular pathologies and ADNC through morphometric measurements of intracranial artery lumen obstruction. Hypertensive participants (n = 354) presented a higher rate of stenosed arteries (obstruction ≥ 50 %), critically stenosed arteries (obstruction ≥ 70 %), and more severe ICAD, shown by higher maximum and mean obstruction indexes compared to nonhypertensive participants (n = 166). These measurements of atherosclerosis were associated with neurofibrillary tangles and cSVD lesions. Hypertension was indirectly associated with hyaline arteriolosclerosis and lacunar infarcts through the pathway of ICAD. Presenting hypertension indirectly increased the odds of displaying hyaline arteriolosclerosis by 26 % (95 % CI: 1.08, 1.45, p = 0.002) and lacunar infarcts by 17 % (95 % CI: 1.01, 1.35, p = 0.029). Cognitive and APOE ε4 carrier status did not alter the investigated associations. In this community sample, hypertension was indirectly associated with cSVD through ICAD.
Collapse
Affiliation(s)
| | - Regina Silva Paradela
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Lea Tenenholz Grinberg
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, SP 01246-903, Brazil; Memory and Aging Center, Weill Institute for Neurosciences, Dept. of Neurology, University of California San Francisco, CA 94158, USA
| | | | | | - Vitor Ribeiro Paes
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, SP 01246-903, Brazil
| | - Maria Eduarda Braga
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Michel Satya Naslavsky
- Human Genome and Stem Cell Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Wilson Jacob-Filho
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Ricardo Nitrini
- Department of Neurology, University of Sao Paulo Medical School, SP 01246-903, Brazil
| | | | - Claudia Kimie Suemoto
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil.
| |
Collapse
|
6
|
Tristão-Pereira C, Fuster V, Lopez-Jimenez A, Fernández-Pena A, Semerano A, Fernandez-Nueda I, Garcia-Lunar I, Ayuso C, Sanchez-Gonzalez J, Ibanez B, Gispert JD, Cortes-Canteli M. Subclinical atherosclerosis and brain health in midlife: Rationale and design of the PESA-Brain study. Am Heart J 2024; 278:195-207. [PMID: 39322173 DOI: 10.1016/j.ahj.2024.09.028] [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: 05/29/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
RATIONALE Cognitive decline and dementia have been reportedly linked to atherosclerosis, the main cause of cardiovascular disease. Cohort studies identifying early brain alterations associated with subclinical atherosclerosis are warranted to understand the potential of prevention strategies before cerebral damage becomes symptomatic and irreversible. METHODS & DESIGN The Progression of Early Subclinical Atherosclerosis (PESA) study is a longitudinal observational cohort study that recruited 4,184 asymptomatic middle-aged individuals (40-54 years) in 2010 in Madrid (Spain) to thoroughly characterize subclinical atherosclerosis development over time. In this framework, the PESA-Brain study has been designed to identify early structural, functional and vascular brain changes associated with midlife atherosclerosis and cardiovascular risk factors. The PESA-Brain study targets 1,000 participants at the 10-year follow-up PESA visit and consists of thorough neuropsychological testing, advanced multimodal neuroimaging, and quantification of blood-based neuropathological biomarkers. PRIMARY HYPOTHESIS We hypothesize that, in middle-age, the presence of cardiovascular risk factors and a high burden of subclinical atherosclerosis will be associated with structural, functional and vascular brain alterations, greater amyloid burden and subtle cognitive impairment. We further hypothesize that the link between subclinical atherosclerosis and poor brain health in midlife will be mediated by cerebrovascular pathology and intracranial atherosclerosis. ENROLLMENT DATES The PESA-Brain study started in October 2020 and is estimated to be completed by December 2024. CONCLUSION This study is in a unique position to unveil novel relationships between cardiovascular and brain alterations in the health-to-disease transition, which may have important implications for interventional and therapeutic approaches. CLINICALTRIALS gov identifier: NCT01410318.
Collapse
Affiliation(s)
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, US.
| | | | | | - Aurora Semerano
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Ines Garcia-Lunar
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, University Hospital La Moraleja, Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carmen Ayuso
- Health Research Institute, Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Health Research Institute, Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Juan Domingo Gispert
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Cortes-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Health Research Institute, Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain; Centro Internacional de Neurociencia Cajal - Consejo Superior de Investigaciones Científicas (CINC-CSIC), Madrid, Spain.
| |
Collapse
|
7
|
Li XL, Wang RT, Tan CC, Tan L, Xu W. Systolic blood pressure variability in late-life predicts cognitive trajectory and risk of Alzheimer's disease. Front Aging Neurosci 2024; 16:1448034. [PMID: 39420926 PMCID: PMC11483855 DOI: 10.3389/fnagi.2024.1448034] [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: 06/12/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Background The relationship of systolic blood pressure variability (SBPV) with Alzheimer's disease (AD) remains controversial. We aimed to explore the roles of SBPV in predicting AD incidence and to test the pathways that mediated the relationship of SBPV with cognitive functions. Methods Longitudinal data across 96 months (T0 to T4) were derived from the Alzheimer's disease Neuroimaging Initiative cohort. SBPV for each participant was calculated based on the four measurements of SBP across 24 months (T0 to T3). At T3, logistic regression models were used to test the SBPV difference between 86 new-onset AD and 743 controls. Linear regression models were used to test the associations of SBPV with cognition and AD imaging endophenotypes for 743 non-demented participants (median age = 77.0, female = 42%). Causal mediation analyses were conducted to explore the effects of imaging endophenotypes in mediating the relationships of SBPV with cognitive function. Finally, Cox proportional hazard model was utilized to explore the association of SBPV with incident risk of AD (T3 to T4, mean follow-up = 3.5 years). Results Participants with new-onset AD at T3 had significantly higher SBPV compared to their controls (p = 0.018). Higher SBPV was associated with lower scores of cognitive function (p = 0.005 for general cognition, p = 0.029 for memory, and p = 0.016 for executive function), higher cerebral burden of amyloid deposition by AV45 PET (p = 0.044), lower brain metabolism by FDG PET (p = 0.052), and higher burden of white matter hyperintensities (WMH) (p = 0.012). Amyloid pathology, brain metabolism, and WMH partially (ranging from 17.44% to 36.10%) mediated the associations of SBPV with cognition. Higher SBPV was significantly associated with elevated risk of developing AD (hazard ratio = 1.29, 95% confidence interval = 1.07 to 1.57, p = 0.008). Conclusion These findings supported that maintaining stable SBP in late life helped lower the risk of AD, partially by modulating amyloid pathology, cerebral metabolism, and cerebrovascular health.
Collapse
Affiliation(s)
- Xiao-Lu Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Medical College, Qingdao University, Qingdao, China
| | - Ruo-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Mok VCT, Cai Y, Markus HS. Vascular cognitive impairment and dementia: Mechanisms, treatment, and future directions. Int J Stroke 2024; 19:838-856. [PMID: 39283037 PMCID: PMC11490097 DOI: 10.1177/17474930241279888] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 08/17/2024] [Indexed: 10/21/2024]
Abstract
Worldwide, around 50 million people live with dementia, and this number is projected to triple by 2050. It has been estimated that 20% of all dementia cases have a predominant cerebrovascular pathology, while perhaps another 20% of vascular diseases contribute to a mixed dementia picture. Therefore, the vascular contribution to dementia affects 20 million people currently and will increase markedly in the next few decades, particularly in lower- and middle-income countries.In this review, we discuss the mechanisms of vascular cognitive impairment (VCI) and review management. VCI refers to the spectrum of cerebrovascular pathologies that contribute to any degree of cognitive impairment, ranging from subjective cognitive decline, to mild cognitive impairment, to dementia. While acute cognitive decline occurring soon after a stroke is the most recognized form of VCI, chronic cerebrovascular disease, in particular cerebral small-vessel disease, can cause insidious cognitive decline in the absence of stroke. Moreover, cerebrovascular disease not only commonly co-occurs with Alzheimer's disease (AD) and increases the probability that AD pathology will result in clinical dementia, but may also contribute etiologically to the development of AD pathologies.Despite its enormous health and economic impact, VCI has been a neglected research area, with few adequately powered trials of therapies, resulting in few proven treatments. Current management of VCI emphasizes prevention and treatment of stroke and vascular risk factors, with most evidence for intensive hypertension control. Reperfusion therapies in acute stroke may attenuate the risk of VCI. Associated behavioral symptoms such as apathy and poststroke emotionalism are common. We also highlight novel treatment strategies that will hopefully lead to new disease course-modifying therapies. Finally, we highlight the importance of including cognitive endpoints in large cardiovascular prevention trials and the need for an increased research focus and funding for this important area.
Collapse
Affiliation(s)
- Vincent Chung Tong Mok
- Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Gerald Choa Neuroscience Institute, Li Ka Shing Institute of Health Science, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Yuan Cai
- Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Gerald Choa Neuroscience Institute, Li Ka Shing Institute of Health Science, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Pan X, Lei Z, Chen J, Jia C, Deng J, Liu Y, Luo X, Wang L, Zi D, Wang Z, Li S, Tan J. Blocking α 1 Adrenergic Receptor as a Novel Target for Treating Alzheimer's Disease. ACS Chem Neurosci 2024. [PMID: 39325017 DOI: 10.1021/acschemneuro.4c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
While amyloidopathy and tauopathy have been recognized as hallmarks in Alzheimer's disease (AD) brain, recently, increasing lines of evidence have supported the pathological roles of cerebrovascular changes in the pathogenesis and progression of AD. Restoring or ameliorating the impaired cerebrovascular function during the early phase of the disease may yield benefits against the cognitive decline in AD. In the present study, we evaluated the potential therapeutic effects of nicergoline [NG, a well-known α1 adrenergic receptor (ADR) blocker and vasodilator] against AD through ameliorating vascular abnormalities. Our in vitro data revealed that NG could reverse β-amyloid1-42 (Aβ1-42)-induced PKC/ERK1/2 activation, the downstream pathway of α1-ADR activation, in α1-ADR-overexpressed N2a cells. NG also blocked Aβ1-42- or phenylephrine-induced constrictions in isolated rat arteries. All these in vitro data may suggest ADR-dependent impacts of Aβ on vascular function and the reversal effect of NG. In addition, the ameliorating impacts of NG treatment on cerebral vasoconstriction, vasoremodeling, and cognitive decline were investigated in vivo in a PSAPP transgenic AD mouse model. Consistent with in vitro findings, the chronic treatment of NG significantly ameliorated the cerebrovascular dysfunctions and Aβ plaque depositions in the brain. Moreover, an improved cognitive performance was also observed. Taken together, our findings supported the beneficial effects of NG on AD through adrenergic-related mechanisms and highlighted the therapeutic potential of α1-adrenergic vasomodulators against AD pathologies.
Collapse
Affiliation(s)
- Xidong Pan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Zhifeng Lei
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Jiang Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Congcong Jia
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Institute of Brain Science and Brain-Inspired Research, Jinan 271016, China
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jie Deng
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Ying Liu
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Xingmei Luo
- Comprehensive Ward, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Likun Wang
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Zi
- Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550004, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
- Institute of Translational Medicine; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| |
Collapse
|
10
|
Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Advances in pathogenesis and treatment of vascular endothelial injury-related diseases mediated by mitochondrial abnormality. Front Pharmacol 2024; 15:1422686. [PMID: 39281286 PMCID: PMC11394189 DOI: 10.3389/fphar.2024.1422686] [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: 04/24/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Vascular endothelial cells, serving as a barrier between blood and the arterial wall, play a crucial role in the early stages of the development of atherosclerosis, cardiovascular diseases (CVDs), and Alzheimer's disease (AD). Mitochondria, known as the powerhouses of the cell, are not only involved in energy production but also regulate key biological processes in vascular endothelial cells, including redox signaling, cellular aging, calcium homeostasis, angiogenesis, apoptosis, and inflammatory responses. The mitochondrial quality control (MQC) system is essential for maintaining mitochondrial homeostasis. Current research indicates that mitochondrial dysfunction is a significant driver of endothelial injury and CVDs. This article provides a comprehensive overview of the causes of endothelial injury in CVDs, ischemic stroke in cerebrovascular diseases, and AD, elucidating the roles and mechanisms of mitochondria in these conditions, and aims to develop more effective therapeutic strategies. Additionally, the article offers treatment strategies for cardiovascular and cerebrovascular diseases, including the use of clinical drugs, antioxidants, stem cell therapy, and specific polyphenols, providing new insights and methods for the clinical diagnosis and treatment of related vascular injuries to improve patient prognosis and quality of life. Future research should delve deeper into the molecular and mechanistic links between mitochondrial abnormalities and endothelial injury, and explore how to regulate mitochondrial function to prevent and treat CVDs.
Collapse
Affiliation(s)
- Boxian Pang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Guangtong Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Tieliang Pang
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xinyao Sun
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Liu
- Bioscience Department, University of Nottingham, Nottingham, United Kingdom
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Huang X, Jiang F, Ma Y, Zhu K, Wang Z, Hua Z, Yu J, Zhang L. A bibliometric analysis of endoplasmic reticulum stress and atherosclerosis. Front Physiol 2024; 15:1392454. [PMID: 38938744 PMCID: PMC11210825 DOI: 10.3389/fphys.2024.1392454] [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: 03/25/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
The mechanisms underlying the occurrence and development of atherosclerosis (AS) are diverse, among which endoplasmic reticulum stress (ERS) is an important mechanism that should not be overlooked. However, up to now, there has been no bibliometric study on the relationship between ERS and AS. To understand the research progress in ERS and AS, this paper conducted a statistical analysis of publications in this field using bibliometrics. A total of 1,035 records were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, and the R package "bibliometric" were used to analyze the spatiotemporal distribution, countries, authors, institutions, journals, references, and keywords of the literature, and to present the basic information of this field through visualized maps, as well as determine the collaboration relationships among researchers in this field. This field has gradually developed and stabilized over the past 20 years. The current research hotspots in this field mainly include the relationship between ERS and AS-related cells, the mechanisms by which ERS promotes AS, related diseases, and associated cytokines, etc. Vascular calcification, endothelial dysfunction, NLRP3 inflammasome, and heart failure represent the frontier research in this field and are becoming new research hotspots. It is hoped that this study will provide new insights for research and clinical work in the field of ERS and AS.
Collapse
Affiliation(s)
- Xinyu Huang
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Yongbo Ma
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Kunpeng Zhu
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhenyuan Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhen Hua
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Jie Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| |
Collapse
|
12
|
Ourry V, Binette AP, St-Onge F, Strikwerda-Brown C, Chagnot A, Poirier J, Breitner J, Arenaza-Urquijo EM, Rabin JS, Buckley R, Gonneaud J, Marchant NL, Villeneuve S. How Do Modifiable Risk Factors Affect Alzheimer's Disease Pathology or Mitigate Its Effect on Clinical Symptom Expression? Biol Psychiatry 2024; 95:1006-1019. [PMID: 37689129 DOI: 10.1016/j.biopsych.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/11/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023]
Abstract
Epidemiological studies show that modifiable risk factors account for approximately 40% of the population variability in risk of developing dementia, including sporadic Alzheimer's disease (AD). Recent findings suggest that these factors may also modify disease trajectories of people with autosomal-dominant AD. With positron emission tomography imaging, it is now possible to study the disease many years before its clinical onset. Such studies can provide key knowledge regarding pathways for either the prevention of pathology or the postponement of its clinical expression. The former "resistance pathway" suggests that modifiable risk factors could affect amyloid and tau burden decades before the appearance of cognitive impairment. Alternatively, the resilience pathway suggests that modifiable risk factors may mitigate the symptomatic expression of AD pathology on cognition. These pathways are not mutually exclusive and may appear at different disease stages. Here, in a narrative review, we present neuroimaging evidence that supports both pathways in sporadic AD and autosomal-dominant AD. We then propose mechanisms for their protective effect. Among possible mechanisms, we examine neural and vascular mechanisms for the resistance pathway. We also describe brain maintenance and functional compensation as bases for the resilience pathway. Improved mechanistic understanding of both pathways may suggest new interventions.
Collapse
Affiliation(s)
- Valentin Ourry
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Clinical Memory Research Unit, Department of Clinical Sciences, Lunds Universitet, Malmö, Sweden
| | - Frédéric St-Onge
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cherie Strikwerda-Brown
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; School of Psychological Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Audrey Chagnot
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - John Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Eider M Arenaza-Urquijo
- Environment and Health over the Lifecourse Programme, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Buckley
- Melbourne School of Psychological Sciences University of Melbourne, Parkville, Victoria, Australia; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Julie Gonneaud
- Normandie University, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Natalie L Marchant
- Division of Psychiatry, University College London, London, United Kingdom
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Grenon MB, Papavergi MT, Bathini P, Sadowski M, Lemere CA. Temporal Characterization of the Amyloidogenic APPswe/PS1dE9;hAPOE4 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:5754. [PMID: 38891941 PMCID: PMC11172317 DOI: 10.3390/ijms25115754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating disorder with a global prevalence estimated at 55 million people. In clinical studies administering certain anti-beta-amyloid (Aβ) antibodies, amyloid-related imaging abnormalities (ARIAs) have emerged as major adverse events. The frequency of these events is higher among apolipoprotein ε4 allele carriers (APOE4) compared to non-carriers. To reflect patients most at risk for vascular complications of anti-Aβ immunotherapy, we selected an APPswe/PS1dE9 transgenic mouse model bearing the human APOE4 gene (APPPS1:E4) and compared it with the same APP/PS1 mouse model bearing the human APOE3 gene (APOE ε3 allele; APPPS1:E3). Using histological and biochemical analyses, we characterized mice at three ages: 8, 12, and 16 months. Female and male mice were assayed for general cerebral fibrillar and pyroglutamate (pGlu-3) Aβ deposition, cerebral amyloid angiopathy (CAA), microhemorrhages, apoE and cholesterol composition, astrocytes, microglia, inflammation, lysosomal dysfunction, and neuritic dystrophy. Amyloidosis, lipid deposition, and astrogliosis increased with age in APPPS1:E4 mice, while inflammation did not reveal significant changes with age. In general, APOE4 carriers showed elevated Aβ, apoE, reactive astrocytes, pro-inflammatory cytokines, microglial response, and neuritic dystrophy compared to APOE3 carriers at different ages. These results highlight the potential of the APPPS1:E4 mouse model as a valuable tool in investigating the vascular side effects associated with anti-amyloid immunotherapy.
Collapse
Affiliation(s)
- Martine B. Grenon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Section Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maria-Tzousi Papavergi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Praveen Bathini
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| | - Martin Sadowski
- Departments of Neurology, Psychiatry, and Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| |
Collapse
|
14
|
Lei T, Yang Z, Li H, Qin M, Gao H. Interactions between nanoparticles and pathological changes of vascular in Alzheimer's disease. Adv Drug Deliv Rev 2024; 207:115219. [PMID: 38401847 DOI: 10.1016/j.addr.2024.115219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Emerging evidence suggests that vascular pathological changes play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The dysfunction of the cerebral vasculature occurs in the early course of AD, characterized by alterations in vascular morphology, diminished cerebral blood flow (CBF), impairment of the neurovascular unit (NVU), vasculature inflammation, and cerebral amyloid angiopathy. Vascular dysfunction not only facilitates the influx of neurotoxic substances into the brain, triggering inflammation and immune responses but also hampers the efflux of toxic proteins such as Aβ from the brain, thereby contributing to neurodegenerative changes in AD. Furthermore, these vascular changes significantly impact drug delivery and distribution within the brain. Therefore, developing targeted delivery systems or therapeutic strategies based on vascular alterations may potentially represent a novel breakthrough in AD treatment. This review comprehensively examines various aspects of vascular alterations in AD and outlines the current interactions between nanoparticles and pathological changes of vascular.
Collapse
Affiliation(s)
- Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zixiao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
15
|
Ip BYM, Ko H, Lam BYK, Au LWC, Lau AYL, Huang J, Kwok AJ, Leng X, Cai Y, Leung TWH, Mok VCT. Current and Future Treatments of Vascular Cognitive Impairment. Stroke 2024; 55:822-839. [PMID: 38527144 DOI: 10.1161/strokeaha.123.044174] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Affiliation(s)
- Bonaventure Yiu Ming Ip
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Bonnie Yin Ka Lam
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Lisa Wing Chi Au
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Alexander Yuk Lun Lau
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Andrew John Kwok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Xinyi Leng
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Yuan Cai
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Thomas Wai Hong Leung
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Vincent Chung Tong Mok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| |
Collapse
|
16
|
Nagai M, Dote K, Park S, Turana Y, Buranakitjaroen P, Cheng HM, Soenarta AA, Li Y, Kario K. Obstructive sleep apnea and non-dipper: epiphenomena or risks of Alzheimer's disease?: a review from the HOPE Asia Network. Hypertens Res 2024; 47:271-280. [PMID: 37875673 DOI: 10.1038/s41440-023-01440-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 10/26/2023]
Abstract
Obstructive sleep apnea (OSA) and associated nocturnal blood pressure (BP) surges is associated with non-dipper. On the other hand, the relationship between neurodegenerative diseases and non-dipper hypertension has been reported. To date, few studies have evaluated the relationships of nocturnal BP dipping patterns and OSA in relation to neurodegenerative diseases, particularly Alzheimer's disease (AD). This review examines the etiology of the association between OSA and the non-dipper pattern of hypertension and how both are involved in the development of AD. To set the stage for this review, we first focus on the pathophysiology of AD, which is interrelated with sleep apnea and non-dipper through dysregulation of central autonomic network.
Collapse
Affiliation(s)
- Michiaki Nagai
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan.
| | - Keigo Dote
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University Health System, Seoul, Korea
| | - Yuda Turana
- Department of Neurology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Peera Buranakitjaroen
- Division of Hypertension, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Hao-Min Cheng
- Institute of Public Health and Community Medicine Research Center, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Ph.D. Program of Interdisciplinary Medicine (PIM), National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan
- Department of Medical Education, Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Arieska Ann Soenarta
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, University of Indonesia, National Cardiovascular Center, Harapan Kita, Jakarta, Indonesia
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
17
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
18
|
Starmans NLP, Kappelle LJ, Muller M, Staals J, Teunissen CE, Biessels GJ, van der Flier WM, Wolters FJ. Blood Pressure Variability and Plasma Biomarkers of Neuronal Injury and Alzheimer's Disease: A Clinic-Based Study of Patients with Diseases Along the Heart-Brain Axis. J Alzheimers Dis 2024; 99:1207-1215. [PMID: 38788076 PMCID: PMC11191465 DOI: 10.3233/jad-240119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/26/2024]
Abstract
Higher blood pressure variability (BPV) predisposes to cognitive decline. To investigate underlying mechanisms, we measured 24-h ambulatory BPV, nocturnal dipping and orthostatic hypotension in 518 participants with vascular cognitive impairment, carotid occlusive disease, heart failure, or reference participants. We determined cross-sectional associations between BPV indices and plasma biomarkers of neuronal injury (neurofilament light chain) and Alzheimer's disease (phosphorylated-tau-181 and Aβ42/Aβ40). None of the BPV indices were significantly associated with any of the biomarkers. Hence, in patients with diseases along the heart-brain axis, we found no evidence for an association between BPV and selected markers of neuronal injury or Alzheimer's disease.
Collapse
Affiliation(s)
| | - Laurens Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Majon Muller
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Julie Staals
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Charlotte Elisabeth Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje Maria van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank Johannes Wolters
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - on behalf of the Heart-Brain Connection Consortium
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
19
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
20
|
Frentz I, van Arendonk J, Leeuwis AE, Vernooij MW, van der Flier WM, Bos D, De Deyn PP, Wolters FJ, Ikram MA. Interaction Between Arteriosclerosis and Amyloid-β on Cognitive Function. J Alzheimers Dis 2024; 97:953-961. [PMID: 38217596 PMCID: PMC10836547 DOI: 10.3233/jad-230604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Dementia is a multifactorial disease, with Alzheimer's disease (AD) and vascular pathology often co-occurring in many individuals with dementia. Yet, the interplay between AD and vascular pathology in cognitive decline is largely undetermined. OBJECTIVE The aim of the present study was to examine the joint effect of arteriosclerosis and AD pathology on cognition in the general population without dementia. METHODS We determined the interaction between blood-based AD biomarkers and CT-defined arteriosclerosis on cognition in 2,229 dementia-free participants of the population-based Rotterdam Study (mean age: 68.9 years, 52% women) cross-sectionally. RESULTS Amyloid-β (Aβ)42 and arterial calcification were associated with cognitive performance. After further adjustment for confounders in a model that combined all biomarkers, only arterial calcification remained independently associated with cognition. There was a significant interaction between arterial calcification and Aβ42 and between arterial calcification and the ratio of Aβ42/40. Yet, estimates attenuated, and interactions were no longer statistically significant after adjustment for cardio metabolic risk factors. CONCLUSIONS Arteriosclerosis and AD display additive interaction-effects on cognition in the general population, that are due in part to cardio metabolic risk factors. These findings suggest that joint assessment of arteriosclerosis and AD pathology is important for understanding of disease etiology in individuals with cognitive impairment.
Collapse
Affiliation(s)
- Ingeborg Frentz
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Neurology, UMCG, Groningen, The Netherlands
| | - Joyce van Arendonk
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, GD Rotterdam, The Netherlands
| | - Anna E. Leeuwis
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Meike W. Vernooij
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, GD Rotterdam, The Netherlands
| | - Wiesje M. van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, GD Rotterdam, The Netherlands
| | - Peter Paul De Deyn
- Department of Neurology, UMCG, Groningen, The Netherlands
- Alzheimer Centre Groningen, UMCG, Groningen, The Netherlands
| | - Frank J. Wolters
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, GD Rotterdam, The Netherlands
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Saito S, Suzuki K, Ohtani R, Maki T, Kowa H, Tachibana H, Washida K, Kawabata N, Mizuno T, Kanki R, Sudoh S, Kitaguchi H, Shindo K, Shindo A, Oka N, Yamamoto K, Yasuno F, Kakuta C, Kakuta R, Yamamoto Y, Hattori Y, Takahashi Y, Nakaoku Y, Tonomura S, Oishi N, Aso T, Taguchi A, Kagimura T, Kojima S, Taketsuna M, Tomimoto H, Takahashi R, Fukuyama H, Nagatsuka K, Yamamoto H, Fukushima M, Ihara M. Efficacy and Safety of Cilostazol in Mild Cognitive Impairment: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2344938. [PMID: 38048134 PMCID: PMC10696485 DOI: 10.1001/jamanetworkopen.2023.44938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/15/2023] [Indexed: 12/05/2023] Open
Abstract
Importance Recent evidence indicates the efficacy of β-amyloid immunotherapy for the treatment of Alzheimer disease, highlighting the need to promote β-amyloid removal from the brain. Cilostazol, a selective type 3 phosphodiesterase inhibitor, promotes such clearance by facilitating intramural periarterial drainage. Objective To determine the safety and efficacy of cilostazol in mild cognitive impairment. Design, Setting, and Participants The COMCID trial (A Trial of Cilostazol for Prevention of Conversion from Mild Cognitive Impairment to Dementia) was an investigator-initiated, double-blind, phase 2 randomized clinical trial. Adult participants were registered between May 25, 2015, and March 31, 2018, and received placebo or cilostazol for up to 96 weeks. Participants were treated in the National Cerebral and Cardiovascular Center and 14 other regional core hospitals in Japan. Patients with mild cognitive impairment with Mini-Mental State Examination (MMSE) scores of 22 to 28 points (on a scale of 0 to 30, with lower scores indicating greater cognitive impairment) and Clinical Dementia Rating scores of 0.5 points (on a scale of 0, 0.5, 1, 2, and 3, with higher scores indicating more severe dementia) were enrolled. The data were analyzed from May 1, 2020, to December 1, 2020. Interventions The participants were treated with placebo, 1 tablet twice daily, or cilostazol, 50 mg twice daily, for up to 96 weeks. Main Outcomes and Measures The primary end point was the change in the total MMSE score from baseline to the final observation. Safety analyses included all adverse events. Results The full analysis set included 159 patients (66 [41.5%] male; mean [SD] age, 75.6 [5.2] years) who received placebo or cilostazol at least once. There was no statistically significant difference between the placebo and cilostazol groups for the primary outcome. The least-squares mean (SE) changes in the MMSE scores among patients receiving placebo were -0.1 (0.3) at the 24-week visit, -0.8 (0.3) at 48 weeks, -1.2 (0.4) at 72 weeks, and -1.3 (0.4) at 96 weeks. Among those receiving cilostazol, the least-squares mean (SE) changes in MMSE scores were -0.6 (0.3) at 24 weeks, -1.0 (0.3) at 48 weeks, -1.1 (0.4) at 72 weeks, and -1.8 (0.4) at 96 weeks. Two patients (2.5%) in the placebo group and 3 patients (3.8%) in the cilostazol group withdrew owing to adverse effects. There was 1 case of subdural hematoma in the cilostazol group, which may have been related to the cilostazol treatment; the patient was successfully treated surgically. Conclusions and Relevance In this randomized clinical trial, cilostazol was well tolerated, although it did not prevent cognitive decline. The efficacy of cilostazol should be tested in future trials. Trial Registration ClinicalTrials.gov Identifier: NCT02491268.
Collapse
Affiliation(s)
- Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Keisuke Suzuki
- Innovation Center for Translational Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Ryo Ohtani
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hisatomo Kowa
- Division of Neurology, Kobe University Hospital, Kobe, Japan
| | | | - Kazuo Washida
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rie Kanki
- Department of Neurology, Osaka City General Hospital, Osaka, Japan
| | - Shinji Sudoh
- Department of Neurology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Hiroshi Kitaguchi
- Department of Neurology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Katsuro Shindo
- Department of Neurology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Akihiro Shindo
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Nobuyuki Oka
- Department of Neurology, National Hospital Organization Minami Kyoto Hospital, Joyo, Japan
| | - Keiichi Yamamoto
- Internal Medicine and Neurology, Nara Midori Clinic, Nara, Japan
| | - Fumihiko Yasuno
- Department of Psychiatry, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Chikage Kakuta
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Ryosuke Kakuta
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yumi Yamamoto
- Department of Molecular Innovation in Lipidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yorito Hattori
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yukako Takahashi
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yuriko Nakaoku
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Tonomura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Aso
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Tatsuo Kagimura
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Shinsuke Kojima
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Masanori Taketsuna
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidenao Fukuyama
- Research and Educational Unit of Leaders for Integrated Medical System, Kyoto University, Kyoto, Japan
| | - Kazuyuki Nagatsuka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Haruko Yamamoto
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
22
|
Fresnais D, Ihle-Hansen H, Lundström E, Andersson ÅG, Fure B. Cerebrovascular Hemodynamics in Cognitive Impairment and Dementia: A Systematic Review and Meta-Analysis of Transcranial Doppler Studies. Dement Geriatr Cogn Disord 2023; 52:277-295. [PMID: 38008061 PMCID: PMC10911167 DOI: 10.1159/000535422] [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: 06/28/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023] Open
Abstract
INTRODUCTION Transcranial Doppler (TCD) sonography is a noninvasive tool for measuring cerebrovascular hemodynamics. Studies have reported alterations in cerebrovascular hemodynamics in normal aging, mild cognitive impairment (MCI), and dementia, as well as in different etiologies of dementia. This systematic review and meta-analysis was designed to investigate the relationship between cerebral blood velocity (CBv) and pulsatility index (PI) in the middle cerebral artery (MCA) in persons with MCI and dementia. METHODS A systematic literature search was conducted in Pubmed, Embase, Cochrane Library, Epistemonikos, PsychINFO, and CINAHL. The search was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. After screening of 33,439 articles, 86 were reviewed in full-text, and 35 fulfilled the inclusion criteria. RESULTS CBv was significantly lower and PI significantly higher in MCA in vascular dementia (VaD) and Alzheimer's disease (AD) compared to cognitively normal (CN) older persons. Also, CBv was lower in MCI compared to CN. There were no significant differences in CBv in MCA in AD compared with VaD, although PI was higher in VaD compared to AD. CONCLUSION Alterations in cerebrovascular hemodynamics are seen in AD, VaD, and MCI. While PI was slightly higher in VaD compared to AD, the reduction in CBv appears to be equally pronounced across neurodegenerative and vascular etiologies of dementia.
Collapse
Affiliation(s)
- David Fresnais
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
- Department of Internal Medicine, Central Hospital Karlstad, Karlstad, Sweden
| | | | - Erik Lundström
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
- Department of Neurology, Academic University Hospital Uppsala, Uppsala, Sweden
| | - Åsa G. Andersson
- Department of Geriatrics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
| | - Brynjar Fure
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
- Department of Internal Medicine, Central Hospital Karlstad, Karlstad, Sweden
| |
Collapse
|
23
|
Akyol O, Akyol S, Chou MC, Chen S, Liu CK, Selek S, Soares JC, Chen CH. Lipids and lipoproteins may play a role in the neuropathology of Alzheimer's disease. Front Neurosci 2023; 17:1275932. [PMID: 38033552 PMCID: PMC10687420 DOI: 10.3389/fnins.2023.1275932] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease (AD) and other classes of dementia are important public health problems with overwhelming social, physical, and financial effects for patients, society, and their families and caregivers. The pathophysiology of AD is poorly understood despite the extensive number of clinical and experimental studies. The brain's lipid-rich composition is linked to disturbances in lipid homeostasis, often associated with glucose and lipid abnormalities in various neurodegenerative diseases, including AD. Moreover, elevated low-density lipoprotein (LDL) cholesterol levels may be related to a higher probability of AD. Here, we hypothesize that lipids, and electronegative LDL (L5) in particular, may be involved in the pathophysiology of AD. Although changes in cholesterol, triglyceride, LDL, and glucose levels are seen in AD, the cause remains unknown. We believe that L5-the most electronegative subfraction of LDL-may be a crucial factor in understanding the involvement of lipids in AD pathology. LDL and L5 are internalized by cells through different receptors and mechanisms that trigger separate intracellular pathways. One of the receptors involved in L5 internalization, LOX-1, triggers apoptotic pathways. Aging is associated with dysregulation of lipid homeostasis, and it is believed that alterations in lipid metabolism contribute to the pathogenesis of AD. Proposed mechanisms of lipid dysregulation in AD include mitochondrial dysfunction, blood-brain barrier disease, neuronal signaling, inflammation, and oxidative stress, all of which lead ultimately to memory loss through deficiency of synaptic integration. Several lipid species and their receptors have essential functions in AD pathogenesis and may be potential biomarkers.
Collapse
Affiliation(s)
- Omer Akyol
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| | | | - Mei-Chuan Chou
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shioulan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Kuan Liu
- Institute of Precision Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Salih Selek
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Chu-Huang Chen
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
24
|
Khan F, Qiu H. Amyloid-β: A potential mediator of aging-related vascular pathologies. Vascul Pharmacol 2023; 152:107213. [PMID: 37625763 PMCID: PMC11793904 DOI: 10.1016/j.vph.2023.107213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Aging is one of the most promising risk factors for vascular diseases, however, the precise mechanisms mediating aging-related pathologies are not fully understood. Amyloid beta (Aβ), a peptide produced by the proteolytic processing of amyloid precursor protein (APP), is known as a key mediator of brain damage involved in the pathogenesis of Alzheimer's disease (AD). Recently, it was found that the accumulation of Aβ in the vascular wall is linked to a range of aging-related vascular pathologies, indicating a potential role of Aβ in the pathogenesis of aging-associated vascular diseases. In the present review, we have updated the molecular regulation of Aβ in vascular cells and tissues, summarized the relevance of the Aβ deposition with vascular aging and diseases, and the role of Aβ dysregulation in aging-associated vascular pathologies, including the impaired vascular response, endothelial dysfunction, oxidative stress, and inflammation. This review will provide advanced information in understanding aging-related vascular pathologies and a new avenue to explore therapeutic targets.
Collapse
Affiliation(s)
- Fazlullah Khan
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, The University of Arizona, Phoenix 85004, AZ, USA
| | - Hongyu Qiu
- Translational Cardiovascular Research Center, Department of Internal Medicine, College of Medicine-Phoenix, The University of Arizona, Phoenix 85004, AZ, USA.
| |
Collapse
|
25
|
Xu Z, Han Z, Wang J, Jin R, Li Z, Wu Z, Zhao Z, Lv S, Zhao X, Liu Y, Guo X, Tao L. Association Between Long-Term Exposure to Fine Particulate Matter Constituents and Progression of Cerebral Blood Flow Velocity in Beijing: Modifying Effect of Greenness. GEOHEALTH 2023; 7:e2023GH000796. [PMID: 37449300 PMCID: PMC10337285 DOI: 10.1029/2023gh000796] [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: 02/05/2023] [Revised: 05/23/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Few studies have explored the effects of fine particulate matter (PM2.5) and its constituents on the progression of cerebral blood flow velocity (BFV) and the potential modifying role of greenness. In this study, we investigated the association of PM2.5 and its constituents, including sulfate (SO4 2-), nitrate (NO3 -), ammonium (NH4 +), organic matter (OM), and black carbon (BC), with the progression of BFV in the middle cerebral artery. Participants from the Beijing Health Management Cohort who underwent at least two transcranial Doppler sonography examinations during 2015-2020 were recruited. BFV change and BFV change rate were used to define the progression of cerebral BFV. Linear mixed effects models were employed to analyze the data, and the weighted quantile sum regression assessed the contribution of PM2.5 constituents. Additionally, greenness was examined as a modifier. Among the examined constituents, OM exhibited the strongest association with BFV progression. An interquartile range increase in PM2.5 and OM exposure concentrations was associated with a decrease of -16.519 cm/s (95% CI: -17.837, -15.201) and -15.403 cm/s (95% CI: -16.681, -14.126) in BFV change, and -10.369 cm/s/year (95% CI: -11.387, -9.352) and -9.615 cm/s/year (95% CI: -10.599, -8.632) in BFV change rate, respectively. Furthermore, stronger associations between PM2.5 and BFV progression were observed in individuals working in areas with lower greenness, those aged under 45 years, and females. In conclusion, reducing PM2.5 levels in the air, particularly the OM constituent, and enhancing greenness could potentially contribute to the protection of cerebrovascular health.
Collapse
Affiliation(s)
- Zongkai Xu
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Ze Han
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Jinqi Wang
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Rui Jin
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Zhiwei Li
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Zhiyuan Wu
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
- Center of Precision HealthSchool of Medical and Health SciencesEdith Cowan UniversityJoondalupWAAustralia
| | - Zemeng Zhao
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Shiyun Lv
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Xiaoyu Zhao
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Yueruijing Liu
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Xiuhua Guo
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| | - Lixin Tao
- Beijing Municipal Key Laboratory of Clinical EpidemiologyDepartment of Epidemiology and Health StatisticsSchool of Public HealthCapital Medical UniversityBeijingChina
| |
Collapse
|
26
|
Sun F. The impact of blood pressure variability on cognition: current limitations and new advances. J Hypertens 2023; 41:888-905. [PMID: 37016905 PMCID: PMC10158606 DOI: 10.1097/hjh.0000000000003422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/18/2023] [Accepted: 02/23/2023] [Indexed: 04/06/2023]
Abstract
Dementia is the most common neurodegenerative disease in the aging population. Emerging evidence indicates that blood pressure (BP) variability is correlated with cognitive impairment and dementia independent of mean BP levels. The state-of-the-art review summarizes the latest evidence regarding the impact of BP variability on cognition in cognitively intact populations, patients with mild cognitive impairment, and different dementia types, focusing on the important confounding factors and new advances. This review also summarizes the potential mechanisms underlying the relationship between BP variability and cognitive impairment, and dementia, briefly discussing sex differences in the relationship. At last, current limitations and future perspectives are discussed to optimize BP management in preventing cognitive impairment and dementia.
Collapse
Affiliation(s)
- Fen Sun
- Department of Anatomy, College of Basic Medicine
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Su M, Nizamutdinov D, Liu H, Huang JH. Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24119272. [PMID: 37298224 DOI: 10.3390/ijms24119272] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the world's primary cause of dementia, a condition characterized by significant progressive declines in memory and intellectual capacities. While dementia is the main symptom of Alzheimer's, the disease presents with many other debilitating symptoms, and currently, there is no known treatment exists to stop its irreversible progression or cure the disease. Photobiomodulation has emerged as a very promising treatment for improving brain function, using light in the range from red to the near-infrared spectrum depending on the application, tissue penetration, and density of the target area. The goal of this comprehensive review is to discuss the most recent achievements in and mechanisms of AD pathogenesis with respect to neurodegeneration. It also provides an overview of the mechanisms of photobiomodulation associated with AD pathology and the benefits of transcranial near-infrared light treatment as a potential therapeutic solution. This review also discusses the older reports and hypotheses associated with the development of AD, as well as some other approved AD drugs.
Collapse
Affiliation(s)
- Matthew Su
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Damir Nizamutdinov
- Department of Neurosurgery, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott and White Health, Temple, TX 76508, USA
| | - Hanli Liu
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Jason H Huang
- Department of Neurosurgery, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott and White Health, Temple, TX 76508, USA
| |
Collapse
|
28
|
Yahagi-Estevam M, Farias-Itao DS, Leite REP, Rodriguez RD, Pasqualucci CA, Nitrini R, Jacob-Filho W, Power MC, Suemoto CK. The Potential Role of Selection Bias in the Association Between Coronary Atherosclerosis and Cognitive Impairment. J Alzheimers Dis 2023:JAD220820. [PMID: 37182864 DOI: 10.3233/jad-220820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Coronary atherosclerosis assessed in vivo was associated with cognitive impairment; however, conflicting findings have been reported in autopsy samples. OBJECTIVE Our aims were to assess the association between atherosclerotic stenosis in the coronary arteries and cognitive impairment and to investigate the possibility of selection bias in an autopsy study. METHODS Coronary arteries were collected, and the largest luminal stenosis was measured. Sociodemographic, clinical, and cognitive information were reported by a reliable next-of-kin. The association was tested using logistic and linear regressions adjusted for sociodemographic and clinical variables. We restricted the sample to individuals that were born in 1935 or earlier and stratified the analysis by cause of death to investigate the role of selection bias. RESULTS In 253 participants (mean age = 78.0±8.5 years old, 48% male), stenosis was not associated with cognitive impairment (OR = 0.85, 95% CI = 0.69; 1.06, p = 0.15). In individuals who were born before 1936 in the absence of cardiovascular disease as the cause of death, greater stenosis was associated with cognitive impairment (OR = 4.02, 95% CI = 1.39; 11.6, p = 0.01). On the other hand, this association was not present among those born in 1935 or earlier who died of cardiovascular diseases (OR = 0.83, 95% CI = 0.60; 1.16, p = 0.28). CONCLUSION We found that higher coronary stenosis was associated with cognitive impairment only in individuals born in 1935 or earlier and who had not died from cardiovascular diseases. Selection bias may be an important issue when investigating risk factors for chronic degenerative diseases in older individuals using autopsy samples.
Collapse
Affiliation(s)
| | | | - Renata Elaine Paraizo Leite
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group - LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Discipline of Geriatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | - Carlos Augusto Pasqualucci
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group - LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Ricardo Nitrini
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group - LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Department of Neurology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Wilson Jacob-Filho
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group - LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Discipline of Geriatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Melinda C Power
- Department of Epidemiology, George Washington University, Washington, DC, USA
| | - Claudia Kimie Suemoto
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group - LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Discipline of Geriatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
| |
Collapse
|
29
|
Sapkota S, Erickson K, Fletcher E, Tomaszewski Farias SE, Jin LW, DeCarli C. Vascular Risk Predicts Plasma Amyloid β 42/40 Through Cerebral Amyloid Burden in Apolipoprotein E ε4 Carriers. Stroke 2023; 54:1227-1235. [PMID: 37021572 PMCID: PMC10121244 DOI: 10.1161/strokeaha.122.041854] [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: 11/02/2022] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Understanding the neurobiological underpinnings between established multimodal dementia risk factors and noninvasive blood-based biomarkers may lead to greater precision and earlier identification of older adults at risk of accelerated decline and dementia. We examined whether key vascular and genetic risk impact the association between cerebral amyloid burden and plasma aβ (amyloid β) 42/40 in nondemented older adults. METHODS We used nondemented older adults from the UCD-ADRC (University of California, Davis-Alzheimer's Disease Research Center) study (n=96) and Alzheimer's Disease Neuroimaging Initiative (n=104). Alzheimer's Disease Neuroimaging Initiative was examined as confirmatory study cohort. We followed a cross-sectional design and examined linear regression followed by mediation analyses. Vascular risk score was obtained as the sum of hypertension, diabetes, hyperlipidemia, coronary artery disease, and cerebrovascular disease. Apolipoprotein E (APOE) ε4+ risk was genotyped, and plasma aβ42 and aβ40 were assayed. Cerebral amyloid burden was quantified using Florbetapir-PET scans. Baseline age was included as a covariate in all models. RESULTS Vascular risk significantly predicted cerebral amyloid burden in Alzheimer's Disease Neuroimaging Initiative but not in the UCD-ADRC cohort. Cerebral amyloid burden was associated with plasma aβ 42/40 in both cohorts. Higher vascular risk increased cerebral amyloid burden was indirectly associated with reduced plasma aβ 42/40 in Alzheimer's Disease Neuroimaging Initiative but not in UCD-ADRC cohort. However, when stratified by APOE ε4+ risk, we consistently observed this indirect relationship only in APOE ε4+ carriers across both cohorts. CONCLUSIONS Vascular risk is indirectly associated with the level of plasma aβ 42/40 via cerebral amyloid burden only in APOE ε4+ carriers. Nondemented older adults with genetic vulnerability to dementia and accelerated decline may benefit from careful monitoring of vascular risk factors directly associated with cerebral amyloid burden and indirectly with plasma aβ 42/40.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| | - Kelsey Erickson
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| | - Evan Fletcher
- University of California, and Department of Pathology and Laboratory Medicine (K.E., L.-W.J.), University of California, Davis
| | | | - Lee-Way Jin
- University of California, and Department of Pathology and Laboratory Medicine (K.E., L.-W.J.), University of California, Davis
| | - Charles DeCarli
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| |
Collapse
|
30
|
Cerebral amyloid-β deposition in patients with heart disease or carotid occlusive disease: A systematic review and meta-analysis. J Neurol Sci 2023; 445:120551. [PMID: 36669349 DOI: 10.1016/j.jns.2023.120551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Cardiovascular disease is an important contributor to cognitive impairment. This likely involves prototypical vascular disease mechanisms like ischemia, but cardiovascular disease might also impact the brain by accelerating cerebral amyloid-β accumulation. We aimed to determine whether there is an association between heart disease or carotid occlusive disease (COD) and cerebral amyloid-β burden. METHODS We conducted a systematic review of studies investigating cerebral amyloid-β burden, measured with positron emission tomography, in adults with and without heart disease or COD. Where possible, we obtained standardized mean differences (SMD) of amyloid-β standardized uptake volume ratios (SUVr) for meta-analysis. RESULTS Eight cross-sectional studies were identified (1478 participants, aged 60-81 years, 51% female). Three studies on heart disease (two on atrial fibrillation (AF) only, one on AF, coronary artery disease and heart failure) did not find a difference in amyloid-β burden between patients and controls. The pooled difference for 746 participants with and without AF did not reach significance (SMD SUVr 0.14, 95%CI -0.06-0.34). Of the five studies on COD (one on differences between participants with and without COD, four on differences between hemispheres in unilateral COD), four did not find a difference in amyloid-β between participants or hemispheres. The pooled difference in amyloid-β load between hemispheres in 24 patients with unilateral COD was not significant (SMD SUVr -0.13, 95%CI -0.70-0.43). CONCLUSION Based on current studies, although limited and heterogeneous, there is insufficient evidence to support the hypothesis that heart disease or COD are associated with increased cerebral amyloid-β burden.
Collapse
|
31
|
Lehmann DJ, Elshorbagy A, Hurley MJ. Many Paths to Alzheimer's Disease: A Unifying Hypothesis Integrating Biological, Chemical, and Physical Risk Factors. J Alzheimers Dis 2023; 95:1371-1382. [PMID: 37694367 DOI: 10.3233/jad-230295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Sporadic Alzheimer's disease (AD) is a complex, multifactorial disease. We should therefore expect to find many factors involved in its causation. The known neuropathology seen at autopsy in patients dying with AD is not consistently seen in all patients with AD and is sometimes seen in patients without dementia. This suggests that patients follow different paths to AD, with different people having slightly different combinations of predisposing physical, chemical and biologic risk factors, and varying neuropathology. This review summarizes what is known of the biologic and chemical predisposing factors and features in AD. We postulate that, underlying the neuropathology of AD is a progressive failure of neurons, with advancing age or other morbidity, to rid themselves of entropy, i.e., the disordered state resulting from brain metabolism. Understanding the diverse causes of AD may allow the development of new therapies targeted at blocking the paths that lead to dementia in each subset of patients.
Collapse
Affiliation(s)
- Donald J Lehmann
- Oxford Project to Investigate Memory and Ageing (OPTIMA), Department of Pharmacology, University of Oxford, Oxford, UK
| | - Amany Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Michael J Hurley
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
32
|
Abstract
Although the cause(s) of Alzheimer's disease in the majority of cases remains elusive, it has long been associated with hypertension. In animal models of the disease, hypertension has been shown to exacerbate Alzheimer-like pathology and behavior, while in humans, hypertension during mid-life increases the risk of developing the disease later in life. Unfortunately, once individuals are diagnosed with the disease, there are few therapeutic options available. There is neither an effective symptomatic treatment, one that treats the debilitating cognitive and memory deficits, nor, more importantly, a neuroprotective treatment, one that stops the relentless progression of the pathology. Further, there is no specific preventative treatment that offsets the onset of the disease. A key factor or clue in this quest for an effective preventative and therapeutic treatment may lie in the contribution of hypertension to the disease. In this review, we explore the idea that photobiomodulation, the application of specific wavelengths of light onto body tissues, can reduce the neuropathology and behavioral deficits in Alzheimer's disease by controlling hypertension. We suggest that treatment with photobiomodulation can be an effective preventative and therapeutic option for this neurodegenerative disease.
Collapse
Affiliation(s)
- Audrey Valverde
- Université Grenoble Alpes, Fonds de dotation Clinatec, Grenoble, France
| | - John Mitrofanis
- Université Grenoble Alpes, Fonds de dotation Clinatec, Grenoble, France,
Institute of Ophthalmology, University College London, London, United Kingdom,Correspondence to: John Mitrofanis, E-mail:
| |
Collapse
|
33
|
Yu X, Lophatananon A, Mekli K, Burns A, Muir KR, Guo H. A suggested shared aetiology of dementia - a colocalization study. Neurobiol Aging 2022; 117:71-82. [PMID: 35675752 DOI: 10.1016/j.neurobiolaging.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/11/2022]
Abstract
Identification of shared causal genes between dementia and its related clinical outcomes can help understand shared aetiology and multimorbidity surrounding dementia. We performed the HyPrColoc colocalization analysis to detect possible shared causal genes between dementia or Alzheimer's disease (AD) and 5 selected traits: stroke, diabetes, atherosclerosis, cholesterol level, and alcohol consumption within 601 dementia or AD associated genetic regions using summary results of the UK Biobank genome-wide association studies. Functional analysis was performed on the candidate causal genes to explore potential biological pathways. Rs150562240 in the LPIN3 gene was identified as a candidate shared causal variant across dementia, AD and atherosclerosis. Evidence for pairwise colocalization between dementia and stroke, dementia (or AD) and atherosclerosis, and dementia (or AD) and diabetes was found in 2, 6 and 2 genetic regions respectively. Colocalization signals between diabetes and the other 3 non-dementia/AD traits were detected in 5 regions. The colocalization evidence shown in our study suggested shared aetiology between dementia and related diseases such as stroke, atherosclerosis, and diabetes.
Collapse
Affiliation(s)
- Xinzhu Yu
- Centre for Biostatistics, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK
| | - Artitaya Lophatananon
- Centre for Integrated Genomic Medicine, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK
| | - Krisztina Mekli
- Centre for Integrated Genomic Medicine, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK
| | - Alistair Burns
- Division of Neuroscience and Experimental Psychology, School of Social Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK
| | - Kenneth R Muir
- Centre for Integrated Genomic Medicine, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK
| | - Hui Guo
- Centre for Biostatistics, Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester UK.
| |
Collapse
|
34
|
Liao J, Chen G, Liu X, Wei ZZ, Yu SP, Chen Q, Ye K. C/EBPβ/AEP signaling couples atherosclerosis to the pathogenesis of Alzheimer's disease. Mol Psychiatry 2022; 27:3034-3046. [PMID: 35422468 PMCID: PMC9912845 DOI: 10.1038/s41380-022-01556-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/08/2022]
Abstract
Atherosclerosis (ATH) and Alzheimer's disease (AD) are both age-dependent inflammatory diseases, associated with infiltrated macrophages and vascular pathology and overlapping molecules. C/EBPβ, an Aβ or inflammatory cytokine-activated transcription factor, and AEP (asparagine endopeptidase) are intimately implicated in both ATH and AD; however, whether C/EBPβ/AEP signaling couples ATH to AD pathogenesis remains incompletely understood. Here we show that C/EBPβ/AEP pathway mediates ATH pathology and couples ATH to AD. Deletion of C/EBPβ or AEP from primary macrophages diminishes cholesterol load, and inactivation of this pathway reduces foam cell formation and lesions in aorta in ApoE-/- mice, fed with HFD (high-fat-diet). Knockout of ApoE from 3xTg AD mouse model augments serum LDL and increases lesion areas in the aorta. Depletion of C/EBPβ or AEP from 3xTg/ApoE-/- mice substantially attenuates these effects and elevates cerebral blood flow and vessel length, improving cognitive functions. Strikingly, knockdown of ApoE from the hippocampus of 3xTg mice decreases the cerebral blood flow and vessel length and aggravates AD pathologies, leading to cognitive deficits. Inactivation of C/EBPβ/AEP pathway alleviates these events and restores cognitive functions. Hence, our findings demonstrate that C/EBPβ/AEP signaling couples ATH to AD via mediating vascular pathology.
Collapse
Affiliation(s)
- Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Guiqin Chen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
35
|
Tian Z, Ji X, Liu J. Neuroinflammation in Vascular Cognitive Impairment and Dementia: Current Evidence, Advances, and Prospects. Int J Mol Sci 2022; 23:ijms23116224. [PMID: 35682903 PMCID: PMC9181710 DOI: 10.3390/ijms23116224] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is a major heterogeneous brain disease caused by multiple factors, and it is the second most common type of dementia in the world. It is caused by long-term chronic low perfusion in the whole brain or local brain area, and it eventually develops into severe cognitive dysfunction syndrome. Because of the disease’s ambiguous classification and diagnostic criteria, there is no clear treatment strategy for VCID, and the association between cerebrovascular pathology and cognitive impairment is controversial. Neuroinflammation is an immunological cascade reaction mediated by glial cells in the central nervous system where innate immunity resides. Inflammatory reactions could be triggered by various damaging events, including hypoxia, ischemia, and infection. Long-term chronic hypoperfusion-induced ischemia and hypoxia can overactivate neuroinflammation, causing apoptosis, blood–brain barrier damage and other pathological changes, triggering or aggravating the occurrence and development of VCID. In this review, we will explore the mechanisms of neuroinflammation induced by ischemia and hypoxia caused by chronic hypoperfusion and emphasize the important role of neuroinflammation in the development of VCID from the perspective of immune cells, immune mediators and immune signaling pathways, so as to provide valuable ideas for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Zhengming Tian
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
| | - Xunming Ji
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100069, China
- Correspondence: (X.J.); (J.L.); Tel.: +86-13520729063 (J.L.)
| | - Jia Liu
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
- Correspondence: (X.J.); (J.L.); Tel.: +86-13520729063 (J.L.)
| |
Collapse
|
36
|
Reagan AM, Onos KD, Heuer SE, Sasner M, Howell GR. Improving mouse models for the study of Alzheimer's disease. Curr Top Dev Biol 2022; 148:79-113. [PMID: 35461569 DOI: 10.1016/bs.ctdb.2021.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease whose risk is influenced by genetic and environmental factors. Although a number of pathological hallmarks have been extensively studied over the last several decades, a complete picture of disease initiation and progression remains unclear. We now understand that numerous cell types and systems are involved in AD pathogenesis, and that this cellular profile may present differently for each individual, making the creation of relevant mouse models challenging. However, with increasingly diverse data made available by genome-wide association studies, we can identify and examine new genes and pathways involved in genetic risk for AD, many of which involve vascular health and inflammation. When developing mouse models, it is critical to assess (1) an aging timeline that represents onset and progression in humans, (2) genetic variants and context, (3) environmental factors present in human populations that result in both neuropathological and functional changes-themes that we address in this chapter.
Collapse
Affiliation(s)
| | | | - Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States.
| |
Collapse
|
37
|
Baradaran H, Gupta A. Carotid Artery Stiffness: Imaging Techniques and Impact on Cerebrovascular Disease. Front Cardiovasc Med 2022; 9:852173. [PMID: 35369341 PMCID: PMC8964780 DOI: 10.3389/fcvm.2022.852173] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/16/2022] [Indexed: 11/24/2022] Open
Abstract
Arterial stiffness is an important measure of vascular aging and atherosclerosis. Though it is measured in many well-known epidemiologic cohort studies, arterial stiffness is often overlooked in routine clinical practice for a number of reasons including difficulties in measurement, variations in definition, and uncertainties surrounding treatment. Central arterial stiffness, a surrogate for aortic stiffness, is the most commonly measured marker of arterial stiffness. In addition to central stiffness, there are also a number of ultrasound based techniques to measure local vascular stiffness, including carotid stiffness. There is evidence that both local carotid stiffness and central arterial stiffness measures are associated with multiple cerebrovascular processes, including stroke and cognitive dysfunction. Mechanistic explanations supporting this association include increased flow load experienced by the cerebral microvasculature leading to cerebral parenchymal damage. In this article, we review definitions of carotid artery stiffness measures and pathophysiologic mechanisms underpinning its association with plaque development and downstream cerebral pathology. We will review the evidence surrounding the association of carotid stiffness measures with downstream manifestations including stroke, cerebral small vessel disease detected on brain MR such as white matter hyperintensities and covert brain infarctions, brain atrophy, and cognitive dysfunction. With consistent definitions, measurement methods, and further scientific support, carotid stiffness may have potential as an imaging-based risk factor for stroke and cognitive decline.
Collapse
Affiliation(s)
- Hediyeh Baradaran
- Department of Radiology, University of Utah, Salt Lake City, UT, United States
- *Correspondence: Hediyeh Baradaran
| | - Ajay Gupta
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
38
|
Imbimbo C, Spallazzi M, Ferrari-Pellegrini F, Villa A, Zilioli A, Mutti C, Parrino L, Lazzeroni D. Heart rate variability and cognitive performance in adults with cardiovascular risk. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100136. [PMID: 36324405 PMCID: PMC9616279 DOI: 10.1016/j.cccb.2022.100136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/16/2022] [Accepted: 03/11/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Heart rate variability (HRV), a measure of autonomic function, has been associated with both cardiovascular disease and cognitive dysfunction. In turn, cardiovascular risk has been linked to an increased risk of dementia onset. However, whether autonomic dysfunction may represent an early marker of cognitive decline in individuals with high cardiovascular risk is still an open issue. METHODS We performed a complete 24-hour HRV analysis in 50 middle-aged and elderly subjects with cardiovascular risk as assessed with the European Society of Cardiology Systematic Coronary Risk Evaluation (ESC SCORE). Cognitive performance was evaluated by Montreal Cognitive Assessment (MoCA), Free and Cued Selective Reminding Test (FCSRT) and Stroop Color and Word Test. Stepwise regression was used to identify significant associations between 24-hour ambulatory ECGs parameters and cognitive performances. RESULTS There were 30 women and 20 men with mean age of 64.9 years (range 51-77) and the mean ESC SCORE was 6%. Four subjects were diagnosed with mild cognitive impairment. Associations were found between measures of HRV and measures of cognition. Ultra-low frequency (ULF) band power of HRV significantly correlated with MoCA (r = 0.424, p = 0.003), also after adjustment for demographics and education. A significant association was also found between the ESC SCORE and ULF band power (r = -0.470, p = 0.0009). CONCLUSIONS Ultra-low frequency band power of HRV is associated with cognitive performance of middle-aged and elderly subjects with cardiovascular risk. This finding may indicate that autonomic nervous system dysregulation plays a role in developing cardiovascular risk and cognitive decline.
Collapse
Affiliation(s)
- Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Marco Spallazzi
- Department of Specialistic and General Medicine, Unit of Neurology, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Francesca Ferrari-Pellegrini
- Cognitive and Motoric Center, Medicine and Geriatric-Rehabilitation Department of Parma, Azienda Ospedaliero-Universitaria, Parma, Italy
| | | | - Alessandro Zilioli
- Department of Specialistic and General Medicine, Unit of Neurology, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Carlotta Mutti
- Department of Specialistic and General Medicine, Unit of Neurology, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Liborio Parrino
- Department of Specialistic and General Medicine, Unit of Neurology, Azienda Ospedaliero-Universitaria, Parma, Italy
| | | |
Collapse
|
39
|
Salehpour F, Khademi M, Bragin DE, DiDuro JO. Photobiomodulation Therapy and the Glymphatic System: Promising Applications for Augmenting the Brain Lymphatic Drainage System. Int J Mol Sci 2022; 23:ijms23062975. [PMID: 35328396 PMCID: PMC8950470 DOI: 10.3390/ijms23062975] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
The glymphatic system is a glial-dependent waste clearance pathway in the central nervous system, devoted to drain away waste metabolic products and soluble proteins such as amyloid-beta. An impaired brain glymphatic system can increase the incidence of neurovascular, neuroinflammatory, and neurodegenerative diseases. Photobiomodulation (PBM) therapy can serve as a non-invasive neuroprotective strategy for maintaining and optimizing effective brain waste clearance. In this review, we discuss the crucial role of the glymphatic drainage system in removing toxins and waste metabolites from the brain. We review recent animal research on the neurotherapeutic benefits of PBM therapy on glymphatic drainage and clearance. We also highlight cellular mechanisms of PBM on the cerebral glymphatic system. Animal research has shed light on the beneficial effects of PBM on the cerebral drainage system through the clearance of amyloid-beta via meningeal lymphatic vessels. Finally, PBM-mediated increase in the blood–brain barrier permeability with a subsequent rise in Aβ clearance from PBM-induced relaxation of lymphatic vessels via a vasodilation process will be discussed. We conclude that PBM promotion of cranial and extracranial lymphatic system function might be a promising strategy for the treatment of brain diseases associated with cerebrospinal fluid outflow abnormality.
Collapse
Affiliation(s)
- Farzad Salehpour
- College for Light Medicine and Photobiomodulation, D-82319 Starnberg, Germany;
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
| | - Mahsa Khademi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51666, Iran;
| | - Denis E. Bragin
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| | - Joseph O. DiDuro
- ProNeuroLIGHT LLC, Phoenix, AZ 85041, USA
- Correspondence: ; Tel.: +1-(845)-203-9204
| |
Collapse
|
40
|
Keeter WC, Ma S, Stahr N, Moriarty AK, Galkina EV. Atherosclerosis and multi-organ-associated pathologies. Semin Immunopathol 2022; 44:363-374. [PMID: 35238952 PMCID: PMC9069968 DOI: 10.1007/s00281-022-00914-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the vascular system that is characterized by the deposition of modified lipoproteins, accumulation of immune cells, and formation of fibrous tissue within the vessel wall. The disease occurs in vessels throughout the body and affects the functions of almost all organs including the lymphoid system, bone marrow, heart, brain, pancreas, adipose tissue, liver, kidneys, and gastrointestinal tract. Atherosclerosis and associated factors influence these tissues via the modulation of local vascular functions, induction of cholesterol-associated pathologies, and regulation of local immune responses. In this review, we discuss how atherosclerosis interferers with functions of different organs via several common pathways and how the disturbance of immunity in atherosclerosis can result in disease-provoking dysfunctions in multiple tissues. Our growing appreciation of the implication of atherosclerosis and associated microenvironmental conditions in the multi-organ pathology promises to influence our understanding of CVD-associated disease pathologies and to provide new therapeutic opportunities.
Collapse
Affiliation(s)
- W Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Shelby Ma
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Natalie Stahr
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Alina K Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA.
| |
Collapse
|
41
|
Li M, Kitamura A, Beverley J, Koudelka J, Duncombe J, Lennen R, Jansen MA, Marshall I, Platt B, Wiegand UK, Carare RO, Kalaria RN, Iliff JJ, Horsburgh K. Impaired Glymphatic Function and Pulsation Alterations in a Mouse Model of Vascular Cognitive Impairment. Front Aging Neurosci 2022; 13:788519. [PMID: 35095472 PMCID: PMC8793139 DOI: 10.3389/fnagi.2021.788519] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Large vessel disease and carotid stenosis are key mechanisms contributing to vascular cognitive impairment (VCI) and dementia. Our previous work, and that of others, using rodent models, demonstrated that bilateral common carotid stenosis (BCAS) leads to cognitive impairment via gradual deterioration of the neuro-glial-vascular unit and accumulation of amyloid-β (Aβ) protein. Since brain-wide drainage pathways (glymphatic) for waste clearance, including Aβ removal, have been implicated in the pathophysiology of VCI via glial mechanisms, we hypothesized that glymphatic function would be impaired in a BCAS model and exacerbated in the presence of Aβ. Male wild-type and Tg-SwDI (model of microvascular amyloid) mice were subjected to BCAS or sham surgery which led to a reduction in cerebral perfusion and impaired spatial learning acquisition and cognitive flexibility. After 3 months survival, glymphatic function was evaluated by cerebrospinal fluid (CSF) fluorescent tracer influx. We demonstrated that BCAS caused a marked regional reduction of CSF tracer influx in the dorsolateral cortex and CA1-DG molecular layer. In parallel to these changes increased reactive astrogliosis was observed post-BCAS. To further investigate the mechanisms that may lead to these changes, we measured the pulsation of cortical vessels. BCAS impaired vascular pulsation in pial arteries in WT and Tg-SwDI mice. Our findings show that BCAS influences VCI and that this is paralleled by impaired glymphatic drainage and reduced vascular pulsation. We propose that these additional targets need to be considered when treating VCI.
Collapse
Affiliation(s)
- Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Akihiro Kitamura
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Department of Neurology, Shiga University of Medical Science, Otsu, Japan
| | - Joshua Beverley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Lennen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Maurits A Jansen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Ian Marshall
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bettina Platt
- School of Medicine, Medical Sciences and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Ulrich K Wiegand
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roxana O Carare
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, United States
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
42
|
Levin O, Bogolepova A, Lobzin V. General mechanisms of the pathogenesis of neurodenerative and cerebrovascular diseases and the possibilities of their correction. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:11-16. [DOI: 10.17116/jnevro202212205111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
43
|
Bougea A, Anagnostouli M, Angelopoulou E, Spanou I, Chrousos G. Psychosocial and Trauma-Related Stress and Risk of Dementia: A Meta-Analytic Systematic Review of Longitudinal Studies. J Geriatr Psychiatry Neurol 2022; 35:24-37. [PMID: 33205677 DOI: 10.1177/0891988720973759] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Stress has deleterious effects on brain health and yet, the prognostic value of psychosocial stress regarding the most common types of dementias, including Alzheimer disease, is still unclear. The primary aim of this systematic review was to explore the association between psychosocial stress and late onset dementia. We classified 24articles from Medline, PsycINFO, CINAHL, and Web of Science, as pertaining toxic categories of psychosocial and trauma-related stress (low socio-economic status [SES] related inequalities, marital status, posttraumatic stress disorder, work stress, "vital exhaustion" [VE], and, combined stressors). Using the Quality of Prognosis Studies in Systematic Reviews tool, we judged the quality of evidence to be low. This systematic review provided some non-robust, yet suggestive evidence that the above psychosocial types of stress are associated with increased risk of dementia in later life. Future robust, longitudinal studies with repeated validated measures of psychosocial stress and dementiaare required to strengthen or refute these findings.
Collapse
Affiliation(s)
- Anastasia Bougea
- Memory & Movement Disorders Clinic, 1st Department of Neurology, 69078Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Anagnostouli
- Memory & Movement Disorders Clinic, 1st Department of Neurology, 69078Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Demyelinating Diseases Clinic, 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece
| | - Efthalia Angelopoulou
- Memory & Movement Disorders Clinic, 1st Department of Neurology, 69078Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Spanou
- Memory & Movement Disorders Clinic, 1st Department of Neurology, 69078Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
44
|
Wang W, Gottesman RF, Meyer ML, Hughes TM, Sullivan KJ, Wong DF, Lakshminarayan K, Lutsey PL. Carotid Intima-Media Thickness and Amyloid-β Deposition: The ARIC-PET Study. J Alzheimers Dis 2022; 88:17-22. [PMID: 35527548 PMCID: PMC10167843 DOI: 10.3233/jad-215671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We assessed whether carotid intima-media thickness (cIMT) is prospectively associated with amyloid-β (Aβ). 332 nondemented Atherosclerosis Risk in Communities Study participants with carotid ultrasounds (1990-1992) and PET scans (2012-2014) were studied. Participants in the highest (versus lowest) cIMT tertile had 2.17 times the odds of elevated Aβ (95% CI: 1.15-4.11), after demographic and APOE ɛ4 adjustments. An interaction with APOE ɛ4 was observed (p = 0.02). Greater cIMT was associated with elevated Aβ independent of vascular risk factors among those with ≥1 APOE ɛ4 allele, but not in noncarriers. In this cohort, higher cIMT was associated with Aβ deposition 22 years later, particularly among APOE ɛ4 carriers.
Collapse
Affiliation(s)
- Wendy Wang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Rebecca F. Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, Maryland
| | - Michelle L. Meyer
- Department of Emergency Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy M. Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kevin J. Sullivan
- Department of Medicine: The MIND Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dean F. Wong
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
- Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Kamakshi Lakshminarayan
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
45
|
Nagai M, Kato M, Dote K. Visit-to-visit blood pressure variability in mild cognitive impairment: A possible marker of Alzheimer's disease in the SPRINT study? J Clin Hypertens (Greenwich) 2021; 23:2129-2132. [PMID: 34806836 PMCID: PMC8696211 DOI: 10.1111/jch.14388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Michiaki Nagai
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Masaya Kato
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Keigo Dote
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| |
Collapse
|
46
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
47
|
Li S, Wang C, Wang Z, Tan J. Involvement of cerebrovascular abnormalities in the pathogenesis and progression of Alzheimer's disease: an adrenergic approach. Aging (Albany NY) 2021; 13:21791-21806. [PMID: 34479211 PMCID: PMC8457611 DOI: 10.18632/aging.203482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD), as the most common neurodegenerative disease in elder population, is pathologically characterized by β-amyloid (Aβ) plaques, neurofibrillary tangles composed of highly-phosphorylated tau protein and consequently progressive neurodegeneration. However, both Aβ and tau fails to cover the whole pathological process of AD, and most of the Aβ- or tau-based therapeutic strategies are all failed. Increasing lines of evidence from both clinical and preclinical studies have indicated that age-related cerebrovascular dysfunctions, including the changes in cerebrovascular microstructure, blood-brain barrier integrity, cerebrovascular reactivity and cerebral blood flow, accompany or even precede the development of AD-like pathologies. These findings may raise the possibility that cerebrovascular changes are likely pathogenic contributors to the onset and progression of AD. In this review, we provide an appraisal of the cerebrovascular alterations in AD and the relationship to cognitive impairment and AD pathologies. Moreover, the adrenergic mechanisms leading to cerebrovascular and AD pathologies were further discussed. The contributions of early cerebrovascular factors, especially through adrenergic mechanisms, should be considered and treasured in the diagnostic, preventative, and therapeutic approaches to address AD.
Collapse
Affiliation(s)
- Song Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Che Wang
- Department of Pharmaceutical Chemistry, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
48
|
Romagnoli T, Ortolani B, Sanz JM, Trentini A, Seripa D, Nora ED, Capatti E, Cervellati C, Passaro A, Zuliani G, Brombo G. Serum Apo J as a potential marker of conversion from mild cognitive impairment to dementia. J Neurol Sci 2021; 427:117537. [PMID: 34147956 DOI: 10.1016/j.jns.2021.117537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Apolipoprotein J (ApoJ) is present in both plasma and tissues, including brain. Growing evidence suggest that this protein may play an early role on the development of the two most common forms of dementia, Alzheimer's disease (AD) and vascular dementia (VD). OBJECTIVE To evaluate whether serum ApoJ levels might be able to predict the progression to AD, VD, or mixed dementia (AD&VD) in individuals with mild cognitive impairment (MCI). METHODS Serum ApoJ was measured in 196 MCI subjects (aged ≥60 years) with a median follow up of 2.9 years. RESULTS One hundred thirty-two of the enrolled MCI subjects converted to dementia. Among these, 45% developed AD, 33% mixed dementia, 13% VD (VD), and 9% other forms of dementia. A significant trend toward a progressive reduction in the incidence of dementia, regardless of the type, from tertile I (83.1%), to tertile II (63.1%), to tertile III (56.1%) was observed (p = 0.003). After adjustment for potential confounders, a twofold increase in the risk of conversion to dementia was found in subjects belonging to tertile I of Apo J compared with tertile III; the risk increased after two years of follow up, while no differences emerged within the first 2 years. CONCLUSIONS Our results suggest that in MCI subjects, low APOJ levels may be associated with increased risk of developing dementia.
Collapse
Affiliation(s)
- Tommaso Romagnoli
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Beatrice Ortolani
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Juana Maria Sanz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; Department of ROMAGNA, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Trentini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; Department of ROMAGNA, University of Ferrara, 44121 Ferrara, Italy.
| | - Davide Seripa
- Research Laboratory, Complex Structure of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Italy
| | - Edoardo Dalla Nora
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Eleonora Capatti
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Angelina Passaro
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Gloria Brombo
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
49
|
Bandyopadhyay S. Role of Neuron and Glia in Alzheimer's Disease and Associated Vascular Dysfunction. Front Aging Neurosci 2021; 13:653334. [PMID: 34211387 PMCID: PMC8239194 DOI: 10.3389/fnagi.2021.653334] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloidogenicity and vascular dysfunction are the key players in the pathogenesis of Alzheimer’s disease (AD), involving dysregulated cellular interactions. An intricate balance between neurons, astrocytes, microglia, oligodendrocytes and vascular cells sustains the normal neuronal circuits. Conversely, cerebrovascular diseases overlap neuropathologically with AD, and glial dyshomeostasis promotes AD-associated neurodegenerative cascade. While pathological hallmarks of AD primarily include amyloid-β (Aβ) plaques and neurofibrillary tangles, microvascular disorders, altered cerebral blood flow (CBF), and blood-brain barrier (BBB) permeability induce neuronal loss and synaptic atrophy. Accordingly, microglia-mediated inflammation and astrogliosis disrupt the homeostasis of the neuro-vascular unit and stimulate infiltration of circulating leukocytes into the brain. Large-scale genetic and epidemiological studies demonstrate a critical role of cellular crosstalk for altered immune response, metabolism, and vasculature in AD. The glia associated genetic risk factors include APOE, TREM2, CD33, PGRN, CR1, and NLRP3, which correlate with the deposition and altered phagocytosis of Aβ. Moreover, aging-dependent downregulation of astrocyte and microglial Aβ-degrading enzymes limits the neurotrophic and neurogenic role of glial cells and inhibits lysosomal degradation and clearance of Aβ. Microglial cells secrete IGF-1, and neurons show a reduced responsiveness to the neurotrophic IGF-1R/IRS-2/PI3K signaling pathway, generating amyloidogenic and vascular dyshomeostasis in AD. Glial signals connect to neural stem cells, and a shift in glial phenotype over the AD trajectory even affects adult neurogenesis and the neurovascular niche. Overall, the current review informs about the interaction of neuronal and glial cell types in AD pathogenesis and its critical association with cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
50
|
Sriranjan RS, Tarkin JM, Evans NR, Le EPV, Chowdhury MM, Rudd JHF. Atherosclerosis imaging using PET: Insights and applications. Br J Pharmacol 2021; 178:2186-2203. [PMID: 31517992 DOI: 10.1111/bph.14868] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/02/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
PET imaging is able to harness biological processes to characterise high-risk features of atherosclerotic plaque prone to rupture. Current radiotracers are able to track inflammation, microcalcification, hypoxia, and neoangiogenesis within vulnerable plaque. 18 F-fluorodeoxyglucose (18 F-FDG) is the most commonly used radiotracer in vascular studies and is employed as a surrogate marker of plaque inflammation. Increasingly, 18 F-FDG and other PET tracers are also being used to provide imaging endpoints in cardiovascular interventional trials. The evolution of novel PET radiotracers, imaging protocols, and hybrid scanners are likely to enable more efficient and accurate characterisation of high-risk plaque. This review explores the role of PET imaging in atherosclerosis with a focus on PET tracers utilised in clinical research and the applications of PET imaging to cardiovascular drug development.
Collapse
Affiliation(s)
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Nicholas R Evans
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Elizabeth P V Le
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | | | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|