1
|
Bucci T, Choi SE, Tsang CT, Yiu KH, Buckley BJ, Pignatelli P, Scheitz JF, Lip GY, Abdul-Rahim AH. Incident dementia in ischaemic stroke patients with early cardiac complications: A propensity-score matched cohort study. Eur Stroke J 2024:23969873241293573. [PMID: 39487764 DOI: 10.1177/23969873241293573] [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: 11/04/2024] Open
Abstract
INTRODUCTION The risk of dementia in patients with stroke-heart syndrome (SHS) remains unexplored. PATIENTS AND METHODS Retrospective analysis using the TriNetX network, including patients with ischaemic stroke from 2010 to 2020. These patients were categorised into two groups: those with SHS (heart failure, myocardial infarction, ventricular fibrillation, or Takotsubo cardiomyopathy within 30 days post-stroke) and those without SHS. The primary outcome was the 1-year risk of dementia (vascular dementia, dementia in other disease, unspecified dementia, or Alzheimer's disease). The secondary outcome was the 1-year risk of all-cause death. Cox regression analysis after 1:1 propensity score matching (PSM) was performed to calculate the hazard ratios (HRs) and 95% confidence intervals (CIs) for the outcomes. RESULTS We included 52,971 patients with SHS (66.6 ± 14.6 years, 42.2% females) and 854,232 patients without SHS (64.7 ± 15.4 years, 48.2% females). Following PSM, 52,970 well-balanced patients were considered in each group. Patients with SHS had a higher risk of incident dementia compared to those without SHS (HR 1.28, 95%CI 1.20-1.36). The risk was the highest during the first 31 days of follow-up (HR 1.51, 95%CI 1.31-1.74) and was mainly driven by vascular and mixed forms. The increased risk of dementia in patients with SHS, was independent of oral anticoagulant use, sex and age but it was the highest in those aged <75 years compared to ⩾75 years. DISCUSSION AND CONCLUSION SHS is associated with increased risk of dementia. Future studies are needed to develop innovative strategies for preventing complications associated with stroke-heart syndrome and improving the long-term prognosis of these patients.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Sylvia E Choi
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Christopher Tw Tsang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kai-Hang Yiu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Benjamin Jr Buckley
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Cardiovascular Health Sciences, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Jan F Scheitz
- Department of Neurology and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gregory Yh Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Azmil H Abdul-Rahim
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Stroke Division, Department Medicine for Older People, Mersey and West Lancashire Teaching Hospitals NHS Trust, Prescot, UK
| |
Collapse
|
2
|
Zhang Y, Zhang R, Hong H, Wang S, Xie L, Cui L, Li J, Hong L, Li K, Zeng Q, Zhou Y, Zhang M, Sun J, Huang P. An Investigation of Cerebral Vascular Functional Properties in Middle-to-Old Age Community People With High Vascular Risk Profiles. J Magn Reson Imaging 2024; 60:2020-2029. [PMID: 38329184 DOI: 10.1002/jmri.29278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Vascular degeneration is an important cause of brain damage in aging. Assessing the functional properties of the cerebral vascular system may aid early diagnosis and prevention. PURPOSE To investigate the relationships between potential vascular functional markers and vascular risks, brain parenchymal damage, and cognition. STUDY TYPE Retrospective. SUBJECTS Two hundred two general community subjects (42-80 years, males/females: 127/75). FIELD STRENGTH/SEQUENCE 3 T, spin echo T1W/T2W/FLAIR, resting-state functional MRI with an echo-planar sequence (rsfMRI), pseudo-continuous arterial spin labeling (pCASL) with a three-dimensional gradient-spin echo sequence. ASSESSMENT Cerebral blood flow (CBF) in gray matter calculated using pCASL, blood transit times calculated using rsfMRI, and the SD of internal carotid arteries signal (ICAstd) calculated using rsfMRI; visual assessment for lacunes; quantification of white matter hyperintensity volume; permutation test for quality control; collection of demographic and clinical data, Montreal Cognitive Assessment, Mini-Mental State Examination. STATISTICAL TESTS Kolmogorov-Smirnov test; Spearman rank correlation analysis; Multivariable linear regression analysis controlling for covariates; The level of statistical significance was set at P < 0.05. RESULTS Age was negatively associated with ICAstd (β = -0.180). Diabetes was associated with longer blood transit time from large arteries to capillary bed (β = 0.185, adjusted for age, sex, and intracranial volume). Larger ICAstd was associated with less presence of lacunes (odds ratio: 0.418, adjusted for age and sex). Higher gray matter CBF (β = 0.154) and larger ICAstd (β = 0.136) were associated with better MoCA scores (adjusted for age, sex, and education). DATA CONCLUSION Prolonged blood transit time, decreased ICAstd, and diminished CBF were associated with vascular dysfunction and cognitive impairment. They may serve as vascular functional markers in future studies. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiting Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linyun Xie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Cui
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jixuan Li
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Luwei Hong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaicheng Li
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Zhou
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianzhong Sun
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Hiorth YH, Schulz J, Pedersen KF, Tysnes O, Alves G. Orthostatic Hypotension and Risk of Mild Cognitive Impairment and Dementia in Parkinson's Disease. Mov Disord Clin Pract 2024; 11:1365-1372. [PMID: 39108067 PMCID: PMC11542402 DOI: 10.1002/mdc3.14179] [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: 02/21/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Orthostatic hypotension (OH) is a common condition in Parkinson's disease (PD) with a possible link to cognitive decline. OBJECTIVE The aim was to explore the association between OH and PD-associated mild cognitive impairment (PD-MCI) and dementia (PDD) over 9 years in a population-based incident PD cohort. METHODS We prospectively followed up patients from PD diagnosis with serial blood pressure measurements, clinical examinations, and neuropsychological assessments. We defined OH using (1) consensus-based criteria and (2) clinically significant OH by mean arterial pressure (MAP) in standing position ≤75 mmHg. PD-MCI and PDD were diagnosed according to acknowledged criteria. We applied generalized estimating equations models to investigate associations between OH measurements and cognitive impairment over time. Weibull accelerated failure time regression models were used to study if early OH (≤3 years of PD diagnosis) accelerates the time to incident PD-MCI and PDD. RESULTS Of 186 enrolled patients, consensus-based OH affected 68.8%, clinically significant OH 33.9%, PD-MCI 60.8%, and PDD 31.2%. Consensus-based OH was associated with PD-MCI (odds ratio [OR]: 2.04, 95% confidence interval: 1.44-2.90, P < 0.001), whereas clinically significant OH was associated with both PD-MCI (OR: 1.95, 1.11-3.43, P = 0.020) and PDD (OR: 3.66, 1.95-6.86, P < 0.001). Early clinically significant OH, but not early consensus-based OH, reduced time to incident PD-MCI by 54% (P = 0.021) and time to PDD by 44% (P = 0.003) independently of potential confounders, including supine hypertension and cardiovascular disease. CONCLUSIONS MAP in standing position emerged as a stronger predictor of cognitive decline than OH determined using consensus-based criteria. These findings have implications for both research and clinical practice.
Collapse
Affiliation(s)
- Ylva Hivand Hiorth
- Department of Physical Medicine and RehabilitationStavanger University HospitalStavangerNorway
- Centre for Movement Disorders, Stavanger University HospitalStavangerNorway
| | - Jörn Schulz
- Department of Mathematics and PhysicsUniversity of StavangerStavangerNorway
| | - Kenn Freddy Pedersen
- Centre for Movement Disorders, Stavanger University HospitalStavangerNorway
- Department of NeurologyStavanger University HospitalStavangerNorway
| | - Ole‐Bjørn Tysnes
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of NeurologyHaukeland University HospitalBergenNorway
| | - Guido Alves
- Centre for Movement Disorders, Stavanger University HospitalStavangerNorway
- Department of NeurologyStavanger University HospitalStavangerNorway
- Department of Chemistry, Bioscience and Environmental EngineeringUniversity of StavangerStavangerNorway
| |
Collapse
|
4
|
Fan LL, Fang H, Zheng JY, Qiu YH, Wu GL, Cai YF, Chen YB, Zhang SJ. Taohong Siwu decoction alleviates cognitive impairment by suppressing endoplasmic reticulum stress and apoptosis signaling pathway in vascular dementia rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118407. [PMID: 38824979 DOI: 10.1016/j.jep.2024.118407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu Decoction (TSD), a classic traditional Chinese medicine formula, is used for the treatment of vascular diseases, including vascular dementia (VD). However, the mechanisms remain unclear. AIM OF STUDY This study aimed to investigate whether TSD has a positive effect on cognitive impairment in VD rats and to confirm that the mechanism of action is related to the Endoplasmic Reticulum stress (ERs) and cell apoptosis signaling pathway. MATERIALS AND METHODS A total of 40 male adult Sprague-Dawley rats were divided into four groups: sham-operated group (Sham), the two-vessel occlusion group (2VO), the 2VO treated with 4.5 g/kg/d TSD group (2VO + TSD-L), the 2VO treated with 13.5 g/kg/d TSD group (2VO + TSD-H). The rats underwent either 2VO surgery or sham surgery. Postoperative TSD treatment was given for 4 consecutive weeks. Behavioral tests were initiated at the end of gastrulation. Open-field test (OFT) was used to detect the activity level. The New Object Recognition test (NOR) was used to test long-term memory. The Morris water maze (MWM) test was used to examine the foundation of spatial learning and memory. As a final step, the hippocampus was taken for molecular testing. The protein levels of GRP78 (Bip), p-PERK, PERK, IRE1α, p-IRE1α, ATF6, eIF2α, p-eIF2α, ATF4, XBP1, Bcl-2 and Bax were determined by Western blot. Immunofluorescence visualizes molecular expression. RESULTS In the OFT, residence time in the central area was significantly longer in both TSD treatment groups compared to the 2VO group. In the NOR, the recognition index was obviously elevated in both TSD treatment groups. The 2VO group had a significantly longer escape latency and fewer times in crossing the location of the platform compared with the Sham group in MWM. TSD treatment reversed this notion. Pathologically, staining observations confirmed that TSD inhibited hippocampal neuronal loss and alleviated the abnormal reduction of the Nissl body. In parallel, TUNEL staining illustrated that TSD decelerated neuronal apoptosis. Western Blot demonstrated that TSD reduces the expression of ERs and apoptotic proteins. CONCLUSION In this study, the significant ameliorative effect on cognitive impairment of TSD has been determined by comparing the behavioral data of the 4 groups of rats. Furthermore, it was confirmed that this effect of TSD was achieved by suppressing the ERs-mediated apoptosis signaling pathway.
Collapse
Affiliation(s)
- Ling-Ling Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Hao Fang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jia-Yi Zheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yu-Hui Qiu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Guang-Liang Wu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China.
| | - Yun-Bo Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM Guangzhou, 510000, China.
| |
Collapse
|
5
|
Aimagambetova B, Ariko T, Merritt S, Rundek T. Arterial stiffness measured by pulse wave velocity correlated with cognitive decline in hypertensive individuals: a systematic review. BMC Neurol 2024; 24:393. [PMID: 39415095 PMCID: PMC11481605 DOI: 10.1186/s12883-024-03905-8] [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: 05/27/2023] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Arterial stiffness is a degenerative modification in the arterial wall that significantly affects normal aging. Arterial hypertension is a major risk factor for cerebrovascular impairment. Pulse wave velocity (PWV) is an established gold standard for measuring arterial stiffness. Studies demonstrated that individuals with elevated blood pressure (BP) and PWV are more likely to experience worse cognitive decline compared to those with either condition alone. The aim of this review is to explore the clinical importance of arterial stiffness for cognitive function in older adults with hypertension. METHODS The systematic review was reported following the PRISMA 2020 guidelines and Cochrane protocol and was registered in NIHR PROSPERO. PubMed, Embase, Web of Science, CINAHL, and Cochrane databases were searched for relevant publications up to December 2022. Articles were filtered by age and type of study and only those including a sample size of at least 500 individuals were selected. Screening of abstracts and full-text review of selected articles were carried out through Covidence. RESULTS The full-text review included a total of 434 articles. Twenty-eight prospective studies have met the inclusion criteria. Selected studies used PWV as the main measurement of stiffness: 24 used carotid-femoral, 2 used brachial-ankle, 1 used aortic PWV, and 11 compared different measures. Studies demonstrated a strong association between increased BP and PWV with brain damage and cognitive deterioration among older adults. One study did not find an interaction with hypertension, while another study found that PWV but not BP was associated with cognitive decline. Few studies showed that the association between stiffness and cognitive outcomes was not significant after adjustment for BP. Several authors suggested that cognitive decline induced by stiff vasculature and hypertension benefited from antihypertensive therapy. CONCLUSION The results of this review demonstrated that arterial hypertension is an important factor linking arterial stiffness to cognitive health in older individuals. BP plays a crucial role in brain integrity, whereas PWV was shown to be a strong measure associated with cognitive decline. Together, they can lead to disabling cognitive outcomes. Early screening of stiffness, BP control, and compliance with treatment are essential for cerebrovascular disease prevention. TRIAL REGISTRATION NIHR PROSPERO registry ID: CRD42022379887 .
Collapse
Affiliation(s)
- Botagoz Aimagambetova
- Department: University of Miami Miller School of Medicine, Evelyn F. McKnight Brain Institute, 1120 NW 14th St, Miami, Fl, 33136, USA.
| | - Taylor Ariko
- Department: University of Miami Miller School of Medicine, Evelyn F. McKnight Brain Institute, 1120 NW 14th St, Miami, Fl, 33136, USA
| | - Stacy Merritt
- Department: University of Miami Miller School of Medicine, Evelyn F. McKnight Brain Institute, 1120 NW 14th St, Miami, Fl, 33136, USA
| | - Tatjana Rundek
- Department: University of Miami Miller School of Medicine, Evelyn F. McKnight Brain Institute, 1120 NW 14th St, Miami, Fl, 33136, USA
| |
Collapse
|
6
|
Palmer JA, Kaufman CS, Whitaker-Hilbig AA, Billinger SA. APOE4 carriers display loss of anticipatory cerebral vascular regulation over AD progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315344. [PMID: 39417136 PMCID: PMC11482999 DOI: 10.1101/2024.10.11.24315344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Maintenance of cerebral blood flow during orthostasis is impaired with aging and associated with cognitive decline, but the effect of Apolipoprotein 4-allele (APOE4) is unknown. METHODS Older adults (n=108) (APOE4 carriers, n=47; noncarriers, n=61) diagnosed as cognitively-normal (NC), MCI, or AD participated. Middle cerebral artery blood velocity (MCAv), assessed using Transcranial Doppler ultrasound, and beat-to-beat mean arterial blood pressure (MAP) were continuously recorded during a sit-to-stand transition. Anticipatory and orthostatic-induced MCAv and MAP responses were compared between genotypes and across disease progression. RESULTS Cognitively-normal APOE4 carriers showed greater anticipatory MCAv increase, greater MCAv decrease with orthostasis, and shorter latency of peripheral MAP responses to orthostasis compared to noncarriers. MCAv and MAP responses were delayed and attenuated across the APOE4 disease progression, with no differences between genotypes in MCI and AD. DISCUSSION APOE4 carriers and noncarriers present with distinct phenotypes of cerebral vascular dysfunction during hemodynamic orthostatic challenge. Unique cerebral and peripheral vascular compensation observed in APOE4 carriers may be lost as AD progresses.
Collapse
Affiliation(s)
- Jacqueline A. Palmer
- Division of Physical Therapy and Rehabilitation Science, University of Minnesota Medical School, , Minneapolis, MN, United States of America
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Carolyn S. Kaufman
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States of America
- Department of Internal Medicine, Stanford Health Care, Stanford University, Palo Alto, CA, United States of America
| | - Alicen A. Whitaker-Hilbig
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, 123 Milwaukee Way, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Sandra A. Billinger
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| |
Collapse
|
7
|
Combes BF, Kalva SK, Benveniste PL, Tournant A, Law MH, Newton J, Krüger M, Weber RZ, Dias I, Noain D, Dean-Ben XL, Konietzko U, Baumann CR, Gillberg PG, Hock C, Nitsch RM, Cohen-Adad J, Razansky D, Ni R. Spiral volumetric optoacoustic tomography of reduced oxygen saturation in the spinal cord of M83 mouse model of Parkinson's disease. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06938-w. [PMID: 39382580 DOI: 10.1007/s00259-024-06938-w] [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: 06/25/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE Metabolism and bioenergetics in the central nervous system play important roles in the pathophysiology of Parkinson's disease (PD). Here, we employed a multimodal imaging approach to assess oxygenation changes in the spinal cord of the transgenic M83 murine model of PD overexpressing the mutated A53T alpha-synuclein form in comparison with non-transgenic littermates. METHODS In vivo spiral volumetric optoacoustic tomography (SVOT) was performed to assess oxygen saturation (sO2) in the spinal cords of M83 mice and non-transgenic littermates. Ex vivo high-field T1-weighted (T1w) magnetic resonance imaging (MRI) at 9.4T was used to assess volumetric alterations in the spinal cord. 3D SVOT analysis and deep learning-based automatic segmentation of T1w MRI data for the mouse spinal cord were developed for quantification. Immunostaining for phosphorylated alpha-synuclein (pS129 α-syn), as well as vascular organization (CD31 and GLUT1), was performed after MRI scan. RESULTS In vivo SVOT imaging revealed a lower sO2SVOT in the spinal cord of M83 mice compared to non-transgenic littermates at sub-100 μm spatial resolution. Ex vivo MRI-assisted by in-house developed deep learning-based automatic segmentation (validated by manual analysis) revealed no volumetric atrophy in the spinal cord of M83 mice compared to non-transgenic littermates at 50 μm spatial resolution. The vascular network was not impaired in the spinal cord of M83 mice in the presence of pS129 α-syn accumulation. CONCLUSION We developed tools for deep-learning-based analysis for the segmentation of mouse spinal cord structural MRI data, and volumetric analysis of sO2SVOT data. We demonstrated non-invasive high-resolution imaging of reduced sO2SVOT in the absence of volumetric structural changes in the spinal cord of PD M83 mouse model.
Collapse
Affiliation(s)
- Benjamin F Combes
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Sandeep Kumar Kalva
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Pierre-Louis Benveniste
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| | - Agathe Tournant
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Man Hoi Law
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Joshua Newton
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Maik Krüger
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Inês Dias
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| | - Xose Luis Dean-Ben
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| | - Per-Göran Gillberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Decker KP, Sanjana F, Rizzi N, Kramer MK, Cerjanic AM, Johnson CL, Martens CR. Comparing single- and multi-post labeling delays for the measurements of resting cerebral and hippocampal blood flow for cerebrovascular testing in midlife adults. Front Physiol 2024; 15:1437973. [PMID: 39416381 PMCID: PMC11480070 DOI: 10.3389/fphys.2024.1437973] [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: 05/24/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives To assess the reliability and validity of measuring resting cerebral blood flow (CBF) and hippocampal CBF using a single-post-labeling delay (PLD) and a multi-PLD pseudo-continuous arterial spin labeling (pCASL) protocol for cerebrovascular reactivity (CVR) testing. Methods 25 healthy, midlife adults (57 ± 4 years old) were imaged in a Siemens Prisma 3T magnetic resonance imaging (MRI) scanner. Resting CBF and hippocampal CBF were assessed using two pCASL protocols, our modified single-PLD protocol (pCASL-MOD) to accommodate the needs for CVR testing and the multi-PLD Human Connectome Project (HCP) Lifespan protocol to serve as the reference control (pCASL-HCP). During pCASL-MOD, CVR was calculated as the change in CBF from rest to hypercapnia (+9 mmHg increase in end-tidal partial pressure of carbon dioxide [PETCO2]) and then normalized for PETCO2. The reliability and validity in resting gray matter (GM) CBF, white matter (WM) CBF, and hippocampal CBF between pCASL-MOD and pCASL-HCP protocols were examined using correlation analyses, paired t-tests, and Bland Altman plots. Results The pCASL-MOD and pCASL-HCP protocols were significantly correlated for resting GM CBF [r = 0.72; F (1, 23) = 25.24, p < 0.0001], WM CBF [r = 0.57; F (1, 23) = 10.83, p = 0.003], and hippocampal CBF [r = 0.77; F (1, 23) = 32.65, p < 0.0001]. However, pCASL-MOD underestimated resting GM CBF (pCASL-MOD: 53.7 ± 11.1 v. pCASL-HCP: 69.1 ± 13.1 mL/100 g/min; p < 0.0001), WM CBF (pCASL-MOD: 32.4 ± 4.8 v. pCASL-HCP: 35.5 ± 6.9 mL/100 g/min; p = 0.01), and hippocampal CBF (pCASL-MOD: 50.5 ± 9.0 v. pCASL-HCP: 68.1 ± 12.5 mL/100 g/min; p < 0.0001). PETCO2 increased by 8.0 ± 0.7 mmHg to induce CVR (GM CBF: 4.8% ± 2.6%; WM CBF 2.9% ± 2.5%; and hippocampal CBF: 3.4% ± 3.8%). Conclusion Our single-PLD pCASL-MOD protocol reliably measured CBF and hippocampal CBF at rest given the significant correlation with the multi-PLD pCASL-HCP protocol. Despite the lower magnitude relative to pCASL-HCP, we recommend using our pCASL-MOD protocol for CVR testing in which an exact estimate of CBF is not required such as the assessment of relative change in CBF to hypercapnia.
Collapse
Affiliation(s)
- Kevin P. Decker
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States
| | - Faria Sanjana
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States
| | - Nick Rizzi
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States
| | - Mary K. Kramer
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Alexander M. Cerjanic
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Curtis L. Johnson
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Christopher R. Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States
| |
Collapse
|
9
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
10
|
Vitali F, Torrandell-Haro G, Branigan G, Arias Aristizabal J, Reiman E, Bedrick EJ, Brinton RD, Weinkauf C. Asymptomatic carotid artery stenosis is associated with increased Alzheimer's disease and non-Alzheimer's disease dementia risk. Stroke Vasc Neurol 2024:svn-2024-003164. [PMID: 39266210 DOI: 10.1136/svn-2024-003164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND In the absence of a cerebrovascular accident, whether asymptomatic extracranial carotid atherosclerotic disease (aECAD) affects Alzheimer's disease (AD) and non-AD dementia risk is not clear. Understanding whether aECAD is associated with an increased risk for AD is important as it is present in roughly 10% of the population over 60 and could represent a modifiable risk factor for AD and non-AD dementia. METHODS This retrospective cohort study analysed Mariner insurance claims. Enrolment criteria included patients aged 55 years or older with at least 5 years of data and no initial dementia diagnosis. Subjects with and without aECAD were evaluated for subsequent AD and non-AD dementia diagnoses. Propensity score matching was performed using confounding factors identified by logistic regression. χ2 tests and Kaplan-Meier survival curves were used to evaluate the impact of aECAD diagnosis on AD and non-AD dementia risk over time. RESULTS 767 354 patients met enrolment criteria. After propensity score matching, 62 963 subjects with aECAD and 62 963 subjects without ECAD were followed through data records. The aECAD cohort exhibited an increased relative risk of 1.22 (95% CI 1.15 to 1.29, p<0.001) for AD and 1.48 (95% CI 1.38 to 1.59, p<0.001) for non-AD dementias compared with the propensity score-matched cohort without aECAD. The increased AD risk associated with aECAD was evident in patients younger than 75 years old and was less apparent in patients over 75 years of age. CONCLUSIONS aECAD is associated with an increased risk of developing AD and non-AD dementias. These findings underscore the need for further prospective evaluation of interactions between aECAD and dementia, with potential implications for change of clinical care in both of these large patient populations.
Collapse
Affiliation(s)
- Francesca Vitali
- Neurology, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Georgina Torrandell-Haro
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Gregory Branigan
- The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Juan Arias Aristizabal
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Eric Reiman
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - Edward J Bedrick
- Center for Biomedical Informatics and Biostatistics, University of Arizona Medical Center - University Campus, Tucson, Arizona, USA
| | - Roberta Diaz Brinton
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Craig Weinkauf
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
11
|
Bhattacharjee A, Purohit P, Roy PK. Neuroimaging-based drug discovery for amyloid clearance therapy in Alzheimer's disease using validated causation analysis. Psychiatry Res Neuroimaging 2024; 345:111890. [PMID: 39489926 DOI: 10.1016/j.pscychresns.2024.111890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/25/2024] [Accepted: 09/02/2024] [Indexed: 11/05/2024]
Abstract
Aging-induced hepatic dysfunction can impair cholesterol metabolism, reducing the availability of cholic acid (CA, bile-acid) in brain. CA is reported to have neuroprotective characteristics in preclinical investigations of Alzheimer's disease (AD). Our aim is to probe the causal-connectivity between the players: amyloid, cholic acid and cerebral-blood-flow, and thereby explore therapeutic applicability in AD. From AD neuroimaging initiative biospecimen platform, we evaluated serum cholic-acid (182 healthy/136 AD individuals). We also assessed 50 healthy/50 Alzheimer's subjects containing MRI-ASL scanning (cerebral blood-flow, CBF) and PET-AV45 scanning (amyloid-load). We performed computational causal connectivity to determine the cause-effect relationship among the parameters. Serum cholic acid in AD subjects substantially decreased to half of controls. Causal-connectivity revealed two novel causative pathways: (i) Decreasing serum CA markedly increased amyloid-load; (ii) Increasing amyloid-load distinctly decreased CBF. We substantiated these two causation pathways respectively with collateral available preclinical observations: (a) increased cholic acid reduces amyloid formation by diminishing gamma-secretase; (b) this decreased amyloid induces capillary-flow enhancement by relaxing vascular pericytes. Indeed, cholic acid can increase amyloid-clearance factor. Neuroimaging-based causal connectivity analysis showed that repositioned pharmacological modulation by cholate derivatives may have appreciable potential as novel window for therapeutic approach to AD. Indicative clinical validation is furnished from available therapeutic trial leads.
Collapse
Affiliation(s)
- Anindita Bhattacharjee
- School of Bio-Medical Engineering, Indian Institute of Technology (B.H.U.), Varanasi 221005, India
| | - Pratik Purohit
- School of Bio-Medical Engineering, Indian Institute of Technology (B.H.U.), Varanasi 221005, India
| | - Prasun K Roy
- Department of Life Sciences, Shiv Nadar University (SNU), Delhi NCR 201314, India; SNU-Dassault Systemes Centre of Excellence, Shiv Nadar University, Delhi NCR 201314, India.
| |
Collapse
|
12
|
Carmine D, Aeschbacher S, Coslovsky M, Hennings E, Paladini RE, Peter R, Burger M, Reichlin T, Rodondi N, Müller AS, Ammann P, Conte G, Auricchio A, Moschovitis G, Bardoczi JB, Stauber A, De Perna ML, Zuern CS, Sinnecker T, Badertscher P, Sticherling C, Bonati LH, Conen D, Krisai P, Osswald S, Kühne M. Blood pressure, brain lesions and cognitive decline in patients with atrial fibrillation. Front Cardiovasc Med 2024; 11:1449506. [PMID: 39314770 PMCID: PMC11417621 DOI: 10.3389/fcvm.2024.1449506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background The influence of atrial fibrillation (AF) and blood pressure (BP) on brain lesions and cognitive function is unclear. We aimed to investigate the association of BP with different types of brain lesions and cognitive decline in patients with AF. Methods Overall, 1,213 AF patients underwent standardized brain magnetic resonance imaging at baseline and after 2 years, as well as yearly neurocognitive testing. BP was measured at baseline and categorized according to guidelines. New lesions were defined as new or enlarged brain lesions after 2 years. We defined cognitive decline using three different neurocognitive tests. Logistic and Cox regression analyses were performed to examine the associations of BP with new brain lesions and cognitive decline. Results The mean age was 71 ± 8.4 years, 74% were male and mean BP was 135 ± 18/79 ± 12 mmHg. New ischemic lesions and white matter lesions were found in 5.4% and 18.4%, respectively. After multivariable adjustment, BP was not associated with the presence of new brain lesions after 2 years. There was no association between BP and cognitive decline over a median follow-up of 6 years when using the Montreal Cognitive Assessment or Digit Symbol Substitution Test. However, BP categories were inversely associated with cognitive decline using the Semantic Fluency Test, with the strongest association in patients with hypertension grade 1 [Hazard Ratio (95% Confidence Interval) 0.57(0.42 to 0.77)], compared to patients with optimal BP (p for linear trend: 0.025). Conclusions In a large cohort of AF patients, there was no association between BP and incidence of brain lesions after 2 years. Also, there was no consistent association between BP and cognitive decline over a follow-up of 6 years. Clinical Trial Registration https://clinicaltrials.gov/study/NCT02105844, Identifier (NCT02105844).
Collapse
Affiliation(s)
- Désirée Carmine
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Coslovsky
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Clinical Trial Unit Basel, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Elisa Hennings
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Rebecca E. Paladini
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Raffaele Peter
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Melanie Burger
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tobias Reichlin
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolas Rodondi
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andreas S. Müller
- Department of Cardiology, Triemli Hospital Zürich, Zürich, Switzerland
| | - Peter Ammann
- Department of Cardiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Giulio Conte
- Division of Cardiology, Ente Ospedaliero Cantonale (EOC), Cardiocentro Ticino Institute, Regional Hospital of Lugano, Lugano, Switzerland
| | - Angelo Auricchio
- Division of Cardiology, Ente Ospedaliero Cantonale (EOC), Cardiocentro Ticino Institute, Regional Hospital of Lugano, Lugano, Switzerland
| | - Giorgio Moschovitis
- Division of Cardiology, Ente Ospedaliero Cantonale (EOC), Cardiocentro Ticino Institute, Regional Hospital of Lugano, Lugano, Switzerland
| | - Julia B. Bardoczi
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Annina Stauber
- Department of Cardiology, Triemli Hospital Zürich, Zürich, Switzerland
| | - Maria Luisa De Perna
- Division of Cardiology, Ente Ospedaliero Cantonale (EOC), Cardiocentro Ticino Institute, Regional Hospital of Lugano, Lugano, Switzerland
| | - Christine S. Zuern
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tim Sinnecker
- Department of Neurology and Stroke Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Patrick Badertscher
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christian Sticherling
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Leo H. Bonati
- Research Department, Reha Rheinfelden, Rheinfelden, Switzerland
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Philipp Krisai
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Osswald
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Kühne
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Cardiology/Electrophysiology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
13
|
Mohammadzadeh M, Khoshakhlagh AH, Grafman J. Air pollution: a latent key driving force of dementia. BMC Public Health 2024; 24:2370. [PMID: 39223534 PMCID: PMC11367863 DOI: 10.1186/s12889-024-19918-4] [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: 06/05/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Many researchers have studied the role of air pollutants on cognitive function, changes in brain structure, and occurrence of dementia. Due to the wide range of studies and often contradictory results, the present systematic review was conducted to try and clarify the relationship between air pollutants and dementia. To identify studies for this review, a systematic search was conducted in Scopus, PubMed, and Web of Science databases (without historical restrictions) until May 22, 2023. The PECO statement was created to clarify the research question, and articles that did not meet the criteria of this statement were excluded. In this review, animal studies, laboratory studies, books, review articles, conference papers and letters to the editors were avoided. Also, studies focused on the effect of air pollutants on cellular and biochemical changes (without investigating dementia) were also excluded. A quality assessment was done according to the type of design of each article, using the checklist developed by the Joanna Briggs Institute (JBI). Finally, selected studies were reviewed and discussed in terms of Alzheimer's dementia and non-Alzheimer's dementia. We identified 14,924 articles through a systematic search in databases, and after comprehensive reviews, 53 articles were found to be eligible for inclusion in the current systematic review. The results showed that chronic exposure to higher levels of air pollutants was associated with adverse effects on cognitive abilities and the presence of dementia. Studies strongly supported the negative effects of PM2.5 and then NO2 on the brain and the development of neurodegenerative disorders in old age. Because the onset of brain structural changes due to dementia begins decades before the onset of disease symptoms, and that exposure to air pollution is considered a modifiable risk factor, taking preventive measures to reduce air pollution and introducing behavioral interventions to reduce people's exposure to pollutants is advisable.
Collapse
Affiliation(s)
- Mahdiyeh Mohammadzadeh
- Department of Health in Emergencies and Disasters, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Climate Change and Health Research Center (CCHRC), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Khoshakhlagh
- Department of Occupational Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran.
| | - Jordan Grafman
- Department of Physical Medicine & Rehabilitation, Neurology, Cognitive Neurology and Alzheimer's Center, Department of Psychiatry, Feinberg School of Medicine & Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| |
Collapse
|
14
|
Desikan SK, Brahmbhatt B, Patel J, Kankaria AA, Anagnostakos J, Dux M, Beach K, Gray VL, McDonald T, Crone C, Sikdar S, Sorkin JD, Lal BK. Cognitive impairment in asymptomatic carotid artery stenosis is associated with abnormal segments in the Circle of Willis. J Vasc Surg 2024; 80:746-755.e2. [PMID: 38710420 PMCID: PMC11343677 DOI: 10.1016/j.jvs.2024.04.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/11/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
OBJECTIVE Our group has previously demonstrated that patients with asymptomatic carotid artery stenosis (ACAS) demonstrate cognitive impairment. One proposed mechanism for cognitive impairment in patients with ACAS is cerebral hypoperfusion due to flow-restriction. We tested whether the combination of a high-grade carotid stenosis and inadequate cross-collateralization in the Circle of Willis (CoW) resulted in worsened cognitive impairment. METHODS Twenty-four patients with high-grade (≥70% diameter-reducing) ACAS underwent carotid duplex ultrasound, cognitive assessment, and 3D time-of-flight magnetic resonance angiography. The cognitive battery consisted of nine neuropsychological tests assessing four cognitive domains: learning and recall, attention and working memory, motor and processing speed, and executive function. Raw cognitive scores were converted into standardized T-scores. A structured interpretation of the magnetic resonance angiography images was performed with each segment of the CoW categorized as being either normal or abnormal. Abnormal segments of the CoW were defined as segments characterized as narrowed or occluded due to congenital aplasia or hypoplasia, or acquired atherosclerotic stenosis or occlusion. Linear regression was used to estimate the association between the number of abnormal segments in the CoW, and individual cognitive domain scores. Significance was set to P < .05. RESULTS The mean age of the patients was 66.1 ± 9.6 years, and 79.2% (n = 19) were male. A significant negative association was found between the number of abnormal segments in the CoW and cognitive scores in the learning and recall (β = -6.5; P = .01), and attention and working memory (β = -7.0; P = .02) domains. There was a trend suggesting a negative association in the motor and processing speed (β = -2.4; P = .35) and executive function (β = -4.5; P = .06) domains that did not reach significance. CONCLUSIONS In patients with high-grade ACAS, the concomitant presence of increasing occlusive disease in the CoW correlates with worse cognitive function. This association was significant in the learning and recall and attention and working memory domains. Although motor and processing speed and executive function also declined numerically with increasing abnormal segments in the CoW, the relationship was not significant. Since flow restriction at a carotid stenosis compounded by inadequate collateral compensation across a diseased CoW worsens cerebral perfusion, our findings support the hypothesis that cerebral hypoperfusion underlies the observed cognitive impairment in patients with ACAS.
Collapse
Affiliation(s)
- Sarasijhaa K Desikan
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD.
| | - Binal Brahmbhatt
- Department of Bioengineering, George Mason University, Fairfax, VA
| | - Jigar Patel
- Radiology Service, Veterans Affairs Medical Center, Baltimore, MD
| | - Aman A Kankaria
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD
| | - John Anagnostakos
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD
| | - Moira Dux
- Neuropsychology Section, Veterans Affairs Medical Center, Baltimore, MD
| | - Kirk Beach
- D. Eugene Strandness Vascular Laboratory, Department of Surgery, University of Washington, Seattle, WA
| | - Vicki L Gray
- Department of Physical Therapy and Rehabilitation Science, University of Maryland, Baltimore, MD
| | - Tara McDonald
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD
| | - Caroline Crone
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD
| | | | - John D Sorkin
- Baltimore VA Geriatric Research, Education, and Clinical Center, Baltimore, MD; Department of Medicine, Division of Gerontology and Palliative Care, University of Maryland School of Medicine, Baltimore, MD
| | - Brajesh K Lal
- Division of Vascular Surgery, University of Maryland School of Medicine, Baltimore, MD; Vascular Service, Veterans Affairs Medical Center, Baltimore, MD.
| |
Collapse
|
15
|
Ou YN, Kuo K, Yang L, Zhang YR, Huang SY, Chen SD, Deng YT, Guo Y, Zhang RQ, Wu BS, Tan L, Dong Q, Feng JF, Cheng W, Yu JT. Longitudinal associations of cardiovascular health and vascular events with incident dementia. Stroke Vasc Neurol 2024; 9:418-428. [PMID: 37827852 PMCID: PMC11420916 DOI: 10.1136/svn-2023-002665] [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: 06/26/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
INTRODUCTION Evidence supporting cardiovascular diseases could increase the risk of dementia remains fragmented. A comprehensive study to illuminate the distinctive associations across different dementia types is still lacking. This study is sought to: (1) determine the clinical validity of Framingham General Cardiovascular Risk Score (FGCRS) for dementia assessment and (2) examine the associations between cardiovascular diseases and the risk of dementia. METHODS A total of 432 079 dementia-free individuals at baseline from UK Biobank were included. Multivariable Cox proportional hazard models were used to investigate the prospective associations for FGCRS and a series of cardiovascular diseases with all-cause dementia (ACD) and its major components, Alzheimer's disease (AD) and vascular dementia (VaD). RESULTS During a median follow-up of 110.1 months, 4711 individuals were diagnosed with dementia. FGCRS was associated with increased risks across the dementia spectrum. In stratification analysis, high-risk groups have demonstrated the greatest dementia burdens, particularly to VaD. Over 74 traits, 9 adverse associations, such as chronic ischaemic heart disease (ACD: HR=1.354; AD: HR=1.269; VaD: HR=1.768), atrioventricular block (ACD: HR=1.562; AD: HR=1.556; VaD: HR=2.069), heart failure (ACD: HR=1.639; AD: HR=1.543; VaD: HR=2.141) and hypotension (ACD: HR=2.912; AD: HR=2.361; VaD: HR=3.315) were observed. Several distinctions were also found, with atrial fibrillation, cerebral infarction, and haemorrhage only associated with greater risks of ACD and VaD. DISCUSSION By identifying distinctive associations between cardiovascular diseases and dementia, this study has established a comprehensive 'mapping' that may untangle the long-standing discrepancy. FGCRS has demonstrated its predictivity beyond cardiovascular diseases burdens, suggesting potential opportunities for implantation.
Collapse
Affiliation(s)
- Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Kevin Kuo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Rui-Qi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, People's Republic of China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, People's Republic of China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
16
|
Owens CD, Pinto CB, Mukli P, Gulej R, Velez FS, Detwiler S, Olay L, Hoffmeister JR, Szarvas Z, Muranyi M, Peterfi A, Pinaffi‐Langley ACDC, Adams C, Sharps J, Kaposzta Z, Prodan CI, Kirkpatrick AC, Tarantini S, Csiszar A, Ungvari Z, Olson AL, Li G, Balasubramanian P, Galvan V, Bauer A, Smith ZA, Dasari TW, Whitehead S, Medapti MR, Elahi FM, Thanou A, Yabluchanskiy A. Neurovascular coupling, functional connectivity, and cerebrovascular endothelial extracellular vesicles as biomarkers of mild cognitive impairment. Alzheimers Dement 2024; 20:5590-5606. [PMID: 38958537 PMCID: PMC11350141 DOI: 10.1002/alz.14072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Mild cognitive impairment (MCI) is a prodromal stage of dementia. Understanding the mechanistic changes from healthy aging to MCI is critical for comprehending disease progression and enabling preventative intervention. METHODS Patients with MCI and age-matched controls (CN) were administered cognitive tasks during functional near-infrared spectroscopy (fNIRS) recording, and changes in plasma levels of extracellular vesicles (EVs) were assessed using small-particle flow cytometry. RESULTS Neurovascular coupling (NVC) and functional connectivity (FC) were decreased in MCI compared to CN, prominently in the left-dorsolateral prefrontal cortex (LDLPFC). We observed an increased ratio of cerebrovascular endothelial EVs (CEEVs) to total endothelial EVs in patients with MCI compared to CN, correlating with structural MRI small vessel ischemic damage in MCI. LDLPFC NVC, CEEV ratio, and LDLPFC FC had the highest feature importance in the random Forest group classification. DISCUSSION NVC, CEEVs, and FC predict MCI diagnosis, indicating their potential as markers for MCI cerebrovascular pathology. HIGHLIGHTS Neurovascular coupling (NVC) is impaired in mild cognitive impairment (MCI). Functional connectivity (FC) compensation mechanism is lost in MCI. Cerebrovascular endothelial extracellular vesicles (CEEVs) are increased in MCI. CEEV load strongly associates with cerebral small vessel ischemic lesions in MCI. NVC, CEEVs, and FC predict MCI diagnosis over demographic and comorbidity factors.
Collapse
|
17
|
Prasun P, Vermeire K, Alali A. Cerebral venous sinus thrombosis and SCN1A, a novel association? Mol Biol Rep 2024; 51:883. [PMID: 39088020 DOI: 10.1007/s11033-024-09820-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Cerebral venous sinus thrombosis (CVST) is a rare cause of stroke. Acquired and inherited prothrombotic conditions are the most common risk factors for CVST. Sometimes, an etiology is not found. Wide utilization of next generation sequencing technologies in clinical practice may lead to identification of risk factors other than those classically associated with CVST. METHOD AND RESULTS This retrospective clinical-laboratory observational study has a reference patient who presented with CVST as an adolescent. Work up for prothrombotic conditions showed high homocysteine level secondary to homozygosity for a common polymorphism, c.677 C > T in the methylenetetrahydrofolate reductase (MTHFR) gene. His older unaffected brother has a similar MTHFR genotype and high homocysteine. The whole exome sequencing revealed a likely pathogenic variant in the sodium voltage gated channel, alpha subunit 1(SCN1A) gene. CONCLUSION CVST is a multifactorial disease. Prothrombotic conditions are the most common risk factors for CVST. High homocysteine due to the common MTHFR polymorphisms was previously attributed to various thrombotic conditions including CVST. Although high homocysteine due to MTHFR polymorphism may be a contributing factor, additional risk factors such as blood flow abnormalities during SCN1A related seizures may be needed for thrombosis.
Collapse
Affiliation(s)
- Pankaj Prasun
- Division of Genetics, Department of Pediatrics, West Virginia University Medicine, One Medical Center Drive, Morgantown, WV, 26506, USA.
| | - Kylie Vermeire
- Division of Genetics, Department of Pediatrics, West Virginia University Medicine, One Medical Center Drive, Morgantown, WV, 26506, USA
| | - Abdulrazak Alali
- Division of Genetics, Department of Pediatrics, West Virginia University Medicine, One Medical Center Drive, Morgantown, WV, 26506, USA
| |
Collapse
|
18
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2024; 0:revneuro-2024-0046. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [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: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
19
|
Hergert DC, Gaasedelen O, Ryman SG, Prestopnik J, Caprihan A, Rosenberg GA. Blood-Brain Barrier Permeability Is Associated With Cognitive Functioning in Normal Aging and Neurodegenerative Diseases. J Am Heart Assoc 2024; 13:e034225. [PMID: 38979810 PMCID: PMC11292768 DOI: 10.1161/jaha.124.034225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The purpose of this study was to investigate the relationship between blood-brain barrier (BBB) permeability and cognitive functioning in healthy older adults and individuals with neurodegenerative diseases. METHODS AND RESULTS A total of 124 participants with Alzheimer disease, cerebrovascular disease, or a mix Alzheimer's and cerebrovascular diseases and 55 controlparticipants underwent magnetic resonance imaging and neuropsychological testing. BBB permeability was measured with dynamic contrast-enhanced magnetic resonance imaging and white matter injury was measured using a quantitative diffusion-tensor imaging marker of white matter injury. Structural equation modeling was used to examine the relationships between BBB permeability, vascular risk burden, white matter injury, and cognitive functioning. Vascular risk burden predicted BBB permeability (r=0.24, P<0.05) and white matter injury (r=0.38, P<0.001). BBB permeability predicted increased white matter injury (r=0.34, P<0.001) and increased white matter injury predicted lower cognitive functioning (r=-0.51, P<0.001). CONCLUSIONS The study provides empirical support for a vascular contribution to white matter injury and cognitive impairment, directly or indirectly via BBB permeability. This highlights the importance of targeting modifiable vascular risk factors to help mitigate future cognitive decline.
Collapse
Affiliation(s)
- Danielle C. Hergert
- US Department of Energy (Contractor), Kirtland Air Force BaseAlbuquerqueNMUSA
| | | | - Sephira G. Ryman
- The Mind Research Network/Lovelace Biomedical Research InstituteAlbuquerqueNMUSA
- Department of NeurologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Jillian Prestopnik
- Center for Memory & AgingUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Arvind Caprihan
- The Mind Research Network/Lovelace Biomedical Research InstituteAlbuquerqueNMUSA
| | - Gary A. Rosenberg
- Center for Memory & AgingUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of NeurologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| |
Collapse
|
20
|
Tan CH, Tan JJX. Associations of cardiac function and arterial stiffness with cerebrovascular disease. Int J Cardiol 2024; 407:132037. [PMID: 38604451 DOI: 10.1016/j.ijcard.2024.132037] [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: 01/09/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) represent diffuse small vessel disease implicating the cardiac, systemic, and cerebral vasculatures. As the brain may be the end-organ of cumulative vascular disease, and higher education is protective of both cardiovascular and brain health, we aim to clarify their intertwining relationships. METHODS We evaluated participants (mean age = 64) from the UK Biobank with neuroimaging measures of WMHs, left ventricular ejection fraction (LVEF) quantified using cardiovascular MRI, and arterial stiffness index (ASI) quantified using finger photoplethysmography. We used multiple regression to evaluate the basic, independent, and interactive relationships of LVEF status (n = 27,512) and ASI (n = 33,584) with WMHs. Moderated mediation analysis was used to determine whether the relationship between LVEF status and WMH was mediated by ASI and moderated by education. RESULTS Abnormal LVEF (β = -0.082, p < 0.001) and higher ASI (β = 0.02, p < 0.001) were associated with greater WMHs separately and independently, but not interactively. Moderated mediation analyses revealed that the relationship between abnormal LVEF and WMH was mediated by ASI, for individuals with lower education (β = -0.004, p < 0.001). Abnormal LVEF was associated with lower cortical thickness in 16 predominantly frontotemporal and select parietal regions (FDR, q < 0.05). CONCLUSIONS Cardiovascular dysfunction is associated with regional cerebral atrophy and may precipitate cerebrovascular disease via stiffening of systemic vasculatures, particularly for individuals with lower education. Integrative approaches to study biophysiological vascular systems can elucidate the complex interplay between biological and social determinants of brain and cerebrovascular health.
Collapse
Affiliation(s)
- Chin Hong Tan
- Department of Psychology, Nanyang Technological University, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Jacinth J X Tan
- School of Social Sciences, Singapore Management University, Singapore, Singapore
| |
Collapse
|
21
|
Li Y, Huang Y, Wei F, Li T, Wang Y. Development and validation of a risk prediction model for motoric cognitive risk syndrome in older adults. Aging Clin Exp Res 2024; 36:143. [PMID: 39002102 PMCID: PMC11246282 DOI: 10.1007/s40520-024-02797-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/22/2024] [Indexed: 07/15/2024]
Abstract
OBJECTIVE The objective of this study was to develop a risk prediction model for motoric cognitive risk syndrome (MCR) in older adults. METHODS Participants were selected from the 2015 China Health and Retirement Longitudinal Study database and randomly assigned to the training group and the validation group, with proportions of 70% and 30%, respectively. LASSO regression analysis was used to screen the predictors. Then, identified predictors were included in multivariate logistic regression analysis and used to construct model nomogram. The performance of the model was evaluated by area under the receiver operating characteristic (ROC) curve (AUC), calibration curves and decision curve analysis (DCA). RESULTS 528 out of 3962 participants (13.3%) developed MCR. Multivariate logistic regression analysis showed that weakness, chronic pain, limb dysfunction score, visual acuity score and Five-Times-Sit-To-Stand test were predictors of MCR in older adults. Using these factors, a nomogram model was constructed. The AUC values for the training and validation sets of the predictive model were 0.735 (95% CI = 0.708-0.763) and 0.745 (95% CI = 0.705-0.785), respectively. CONCLUSION The nomogram constructed in this study is a useful tool for assessing the risk of MCR in older adults, which can help clinicians identify individuals at high risk.
Collapse
Affiliation(s)
- Yaqin Li
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Yuting Huang
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Fangxin Wei
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Tanjian Li
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Yu Wang
- The Community Service Center of Jinan University, The First Affiliated Hospital of Jinan University, Tianhe District, Guangzhou, Guangzhou Province, China.
| |
Collapse
|
22
|
Yamamoto N, Nakano M, Nochioka K, Yasuda M, Kunikata H, Nakazawa T, Yasuda S. Ocular blood flow dynamics following sinus rhythm restoration through catheter ablation: laser speckle flowgraphy in patients with persistent atrial fibrillation. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae071. [PMID: 39224622 PMCID: PMC11367941 DOI: 10.1093/ehjimp/qyae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/05/2024] [Indexed: 09/04/2024]
Abstract
Aims Laser speckle flowgraphy (LSFG) is a well-established tool renowned for its non-invasive and reproducible assessment of ocular blood flow. While rhythm control therapies, such as catheter ablation (CA), have shown promise in enhancing cognitive function in atrial fibrillation (AF) patients, the acute impact of CA on microcirculatory changes, particularly in ocular blood flow, remains a topic of limited understanding. The present study aims to delve into the potential of LSFG in detecting microcirculatory alterations following the restoration of sinus rhythm (SR) through CA in patients with AF. Methods and results We studied 8 paroxysmal AF (Paf) and 20 persistent AF (PeAF) patients (mean age 67 ± 6 years, 26% female) undergoing CA. Ocular blood flow was assessed using LSFG by measuring the mean blur rate (MBR) pre- and post-CA. Post-CA, all PeAF patients achieved SR restoration, resulting in a significant increase in tissue MBR (10.0 ± 2.2 to 10.8 ± 2.9, P = 0.021). In contrast, Paf patients showed no significant difference between pre- and post-MBR (12.0 ± 2.7 vs. 11.8 ± 2.6, P = 0.76). Conclusion LSFG analysis effectively identified microcirculatory changes in patients undergoing CA for PeAF, suggesting that therapeutic interventions targeting the heart may have broader implications for ocular and cerebral health, establishing a novel 'cardio-oculo-cerebral relationship'.
Collapse
Affiliation(s)
- Nobuhiko Yamamoto
- Departments of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Makoto Nakano
- Departments of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Kotaro Nochioka
- Departments of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Kunikata
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Yasuda
- Departments of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
23
|
Wang J, Liu D, Guo C, Duan Y, Hu Z, Tian M, Xu Q, Niu Y, Yan G. Association between garden work and risk of incident dementia in an older population in China: a national cohort study. Public Health 2024; 232:74-81. [PMID: 38749151 DOI: 10.1016/j.puhe.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/07/2024] [Accepted: 04/09/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVES Evidence on the association between garden work and risk of incident dementia in the older Chinese population is limited. This study aimed to explore the association between the frequency of garden work and risk of incident dementia in an older population in China. STUDY DESIGN This was a national cohort study. METHODS This study analysed data from 8676 participants (median age: 86 years) from the Chinese Longitudinal Healthy Longevity Survey. Cox proportional hazard models were used to assess the association between the frequency of garden work and risk of incident dementia using hazard ratios (HRs) and 95% confidence intervals (CIs). Multiplicative and additive interaction effects were calculated between the frequency of garden work and age, sex or residence on incident dementia; subgroup analyses of the association were also conducted by age, sex and residence. In addition, sensitivity analyses were performed to assess the robustness of the results. RESULTS During 4.31 years (median) of follow-up, 633 participants developed dementia. Compared with participants who did not engage in garden work, the adjusted risk of incident dementia for those who regularly or almost daily engaged in garden work decreased by 28% (HR = 0.72, 95% CI: 0.57-0.93). An additive interaction effect between frequency of garden work and age on incident dementia was observed, with subgroup analyses demonstrating similar statistically significant associations among participants aged ≥85 years, women and city or town residents. Sensitivity analyses were consistent with the primary analysis in the present study. CONCLUSIONS Frequent engagement in garden work may be associated with a reduced risk of dementia and may be an effective measure to prevent incident dementia in the older population in China.
Collapse
Affiliation(s)
- J Wang
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - D Liu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - C Guo
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Y Duan
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Z Hu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - M Tian
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Q Xu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Y Niu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - G Yan
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
24
|
Rajeev V, Tabassum NI, Fann DY, Chen CP, Lai MK, Arumugam TV. Intermittent Metabolic Switching and Vascular Cognitive Impairment. J Obes Metab Syndr 2024; 33:92-107. [PMID: 38736362 PMCID: PMC11224924 DOI: 10.7570/jomes24010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/25/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024] Open
Abstract
Intermittent fasting (IF), a dietary pattern alternating between eating and fasting periods within a 24-hour cycle, has garnered recognition for its potential to enhance both healthspan and lifespan in animal models and humans. It also shows promise in alleviating age-related diseases, including neurodegeneration. Vascular cognitive impairment (VCI) spans a severity range from mild cognitive deficits to severe cognitive deficits and loss of function in vascular dementia. Chronic cerebral hypoperfusion has emerged as a significant contributor to VCI, instigating vascular pathologies such as microbleeds, blood-brain barrier dysfunction, neuronal loss, and white matter lesions. Preclinical studies in rodents strongly suggest that IF has the potential to attenuate pathological mechanisms, including excitotoxicity, oxidative stress, inflammation, and cell death pathways in VCI models. Hence, this supports evaluating IF in clinical trials for both existing and at-risk VCI patients. This review compiles existing data supporting IF's potential in treating VCI-related vascular and neuronal pathologies, emphasizing the mechanisms by which IF may mitigate these issues. Hence providing a comprehensive overview of the available data supporting IF's potential in treating VCI by emphasizing the underlying mechanisms that make IF a promising intervention for VCI.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nishat I. Tabassum
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - David Y. Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Mitchell K.P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Thiruma V. Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
25
|
Engstrom AC, Alitin JP, Kapoor A, Dutt S, Lohman T, Sible IJ, Marshall AJ, Shenasa F, Gaubert A, Ferrer F, Nguyen A, Bradford DR, Rodgers K, Sordo L, Head E, Shao X, Wang DJ, Nation DA. Spontaneous cerebrovascular reactivity at rest in older adults with and without mild cognitive impairment and memory deficits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.18.24309109. [PMID: 38946941 PMCID: PMC11213117 DOI: 10.1101/2024.06.18.24309109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Older adults with mild cognitive impairment (MCI) exhibit deficits in cerebrovascular reactivity (CVR), suggesting CVR is a biomarker for vascular contributions to MCI. This study examined if spontaneous CVR is associated with MCI and memory impairment. Methods 161 older adults free of dementia or major neurological/psychiatric disorders were recruited. Participants underwent clinical interviews, cognitive testing, venipuncture for Alzheimer's biomarkers, and brain MRI. Spontaneous CVR was quantified during 5 minutes of rest. Results Whole brain CVR was negatively associated with age, but not MCI. Lower CVR in the parahippocampal gyrus (PHG) was found in participants with MCI and was linked to worse memory performance on memory tests. Results remained significant after adjusting for Alzheimer's biomarkers and vascular risk factors. Conclusion Spontaneous CVR deficits in the PHG are observed in older adults with MCI and memory impairment, indicating medial temporal microvascular dysfunction's role in cognitive decline.
Collapse
Affiliation(s)
- Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - John Paul Alitin
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Shubir Dutt
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Trevor Lohman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa J Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Aimée Gaubert
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Farrah Ferrer
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Nguyen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - David Robert Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Lorena Sordo
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Xingfeng Shao
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Danny Jj Wang
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
26
|
Rasti SD, Sugiarto AAR, Nuryandi APA, Arvianti MZ, Yomara RT, Nagasastra J, Julario R, Fagi RA, Windrati DMH. Revitalizing brain perfusion: Unveiling advancements through rhythm control strategies in atrial fibrillation-A systematic review. J Arrhythm 2024; 40:411-422. [PMID: 38939785 PMCID: PMC11199798 DOI: 10.1002/joa3.13056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 06/29/2024] Open
Abstract
Background Recent evidence suggests an elevated risk of cognitive impairment and dementia in individuals with atrial fibrillation (AF), irrespective of stroke occurrence. AF, known to reduce brain perfusion, particularly through silent cerebral ischemia, underscores the intricate relationship between cardiac and cerebral health. The heart plays a crucial role in supporting normal brain function, and rhythm control, a standard AF treatment, has demonstrated enhancements in brain perfusion. This systematic review aimed to examine published data concerning the influence of rhythm control on brain perfusion in patients with atrial fibrillation. Methods A systematic search for relevant studies was carried out in Scopus, PubMed, Cochrane Reviews, ProQuest, and EBSCOhost, spanning from their inception until April 30, 2023. Studies that specifically examined brain perfusion following any form of rhythm control in atrial fibrillation were included in the review. Results The review encompassed 10 studies involving 436 participants. Among these, six utilized electrical cardioversion for rhythm control. The majority (8 out of 10) demonstrated that restoring sinus rhythm markedly enhances brain perfusion. In one of the two remaining studies, notable improvement was observed specifically in a region closely linked to cognition. Additionally, both studies reporting data on the Mini-Mental State Examination (MMSE) showed a consistent and significant increase in scores following rhythm control. Conclusion Successful rhythm control in AF emerges as a significant contributor to enhanced brain perfusion, suggesting a potential therapeutic avenue for reducing cognitive impairment incidence. However, further validation through larger prospective studies and randomized trials is warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rerdin Julario
- Department of Cardiology and Vascular MedicineDr. Soetomo General Hospital, Faculty of Medicine, University of AirlanggaSurabayaIndonesia
| | - Rosi Amrilla Fagi
- Department of Cardiology and Vascular MedicineDr. Soetomo General Hospital, Faculty of Medicine, University of AirlanggaSurabayaIndonesia
| | | |
Collapse
|
27
|
Cheng Y, Lin L, Huang P, Zhang J, Wang Y, Pan X. Hypotension with neurovascular changes and cognitive dysfunction: An epidemiological, pathobiological, and treatment review. Chin Med J (Engl) 2024:00029330-990000000-01080. [PMID: 38785189 DOI: 10.1097/cm9.0000000000003103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Indexed: 05/25/2024] Open
Abstract
ABSTRACT Hypotension is a leading cause of age-related cognitive impairment. The available literature evidences that vascular factors are associated with dementia and that hypotension alters cerebral perfusion flow and can aggravate the neurodegeneration of Alzheimer's disease (AD). Despite the discovery of biomarkers and the recent progress made in neurovascular biology, epidemiology, and brain imaging, some key issues remain largely unresolved: the potential mechanisms underlying the neural deterioration observed in AD, the effect of cerebrovascular alterations on cognitive deficits, and the positive effects of hypotension treatment on cognition. Therefore, further well-designed studies are needed to unravel the potential association between hypotension and cognitive dysfunction and reveal the potential benefits of hypotension treatment for AD patients. Here, we review the current epidemiological, pathobiological, and treatment-related literature on neurovascular changes and hypotension-related cognitive dysfunction and highlight the unsettled but imminent issues that warrant future research endeavors.
Collapse
Affiliation(s)
- Yingzhe Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Lin Lin
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Peilin Huang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Jiejun Zhang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Center for Geriatrics, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yanping Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
28
|
Huang Z, Hamblin MR, Zhang Q. Photobiomodulation in experimental models of Alzheimer's disease: state-of-the-art and translational perspectives. Alzheimers Res Ther 2024; 16:114. [PMID: 38773642 PMCID: PMC11106984 DOI: 10.1186/s13195-024-01484-x] [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: 02/07/2024] [Accepted: 05/15/2024] [Indexed: 05/24/2024]
Abstract
Alzheimer's disease (AD) poses a significant public health problem, affecting millions of people across the world. Despite decades of research into therapeutic strategies for AD, effective prevention or treatment for this devastating disorder remains elusive. In this review, we discuss the potential of photobiomodulation (PBM) for preventing and alleviating AD-associated pathologies, with a focus on the biological mechanisms underlying this therapy. Future research directions and guidance for clinical practice for this non-invasive and non-pharmacological therapy are also highlighted. The available evidence indicates that different treatment paradigms, including transcranial and systemic PBM, along with the recently proposed remote PBM, all could be promising for AD. PBM exerts diverse biological effects, such as enhancing mitochondrial function, mitigating the neuroinflammation caused by activated glial cells, increasing cerebral perfusion, improving glymphatic drainage, regulating the gut microbiome, boosting myokine production, and modulating the immune system. We suggest that PBM may serve as a powerful therapeutic intervention for AD.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
29
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [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: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
30
|
Weijs RWJ, Oudegeest-Sander MH, Hopman MTE, Thijssen DHJ, Claassen JAHR. Cerebrovascular CO 2 reactivity and dynamic cerebral autoregulation through the eighth decade of life and their implications for cognitive decline. J Cereb Blood Flow Metab 2024; 44:712-725. [PMID: 38064286 PMCID: PMC11197147 DOI: 10.1177/0271678x231219568] [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: 06/03/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 04/26/2024]
Abstract
Aging is accompanied by a decrease in cerebral blood flow (CBF), especially in the presence of preclinical cognitive decline. The role of cerebrovascular physiology including regulatory mechanisms of CBF in processes underlying aging and subclinical cognitive decline is, however, not fully understood. We explored changes in cerebrovascular CO2 reactivity and dynamic cerebral autoregulation (dCA) through the eighth decade of life, and their relation with early cognitive decline. After 10.9 years, twenty-eight (age, 80.0 ± 3.5 years; 46% female) out of forty-eight healthy older adults who had participated in a previous study (age at baseline, 70 ± 4 years; 42% female), underwent repeated transcranial Doppler assessments. Linear mixed-model analyses revealed small reductions in cerebrovascular CO2 reactivity with aging (-0.37%/mmHg, P = 0.041), whereas dCA was modestly enhanced (gain: -0.009 cm/s/mmHg, P = 0.038; phase: +8.9 degrees, P = 0.004). These changes were more pronounced in participants who had developed subjective memory complaints at follow-up. Our observations confirm that dCA is not impaired in aging, despite lower cerebral perfusion and cerebrovascular reactivity. Altogether, this unique longitudinal study highlights the involvement of cerebrovascular health in preclinical cognitive decline, which is of clinical relevance in the development of dementia management strategies.
Collapse
Affiliation(s)
- Ralf WJ Weijs
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Madelijn H Oudegeest-Sander
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria TE Hopman
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dick HJ Thijssen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Jurgen AHR Claassen
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
31
|
Pinheiro FI, Araújo-Filho I, do Rego ACM, de Azevedo EP, Cobucci RN, Guzen FP. Hepatopancreatic metabolic disorders and their implications in the development of Alzheimer's disease and vascular dementia. Ageing Res Rev 2024; 96:102250. [PMID: 38417711 DOI: 10.1016/j.arr.2024.102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Dementia has been faced with significant public health challenges and economic burdens that urges the need to develop safe and effective interventions. In recent years, an increasing number of studies have focused on the relationship between dementia and liver and pancreatic metabolic disorders that result in diseases such as diabetes, obesity, hypertension and dyslipidemia. Previous reports have shown that there is a plausible correlation between pathologies caused by hepatopancreatic dysfunctions and dementia. Glucose, insulin and IGF-1 metabolized in the liver and pancreas probably have an important influence on the pathophysiology of the most common dementias: Alzheimer's and vascular dementia. This current review highlights recent studies aimed at identifying convergent mechanisms, such as insulin resistance and other diseases, linked to altered hepatic and pancreatic metabolism, which are capable of causing brain changes that ultimately lead to dementia.
Collapse
Affiliation(s)
- Francisco I Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Irami Araújo-Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Amália C M do Rego
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Eduardo P de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil
| | - Ricardo N Cobucci
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil; Postgraduate Program in Science Applied to Women`s Health, Medical School, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Fausto P Guzen
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil; Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil.
| |
Collapse
|
32
|
Faraci FM, Scheer FA. Hypertension: Causes and Consequences of Circadian Rhythms in Blood Pressure. Circ Res 2024; 134:810-832. [PMID: 38484034 PMCID: PMC10947115 DOI: 10.1161/circresaha.124.323515] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024]
Abstract
Hypertension is extremely common, affecting approximately 1 in every 2 adults globally. Chronic hypertension is the leading modifiable risk factor for cardiovascular disease and premature mortality worldwide. Despite considerable efforts to define mechanisms that underlie hypertension, a potentially major component of the disease, the role of circadian biology has been relatively overlooked in both preclinical models and humans. Although the presence of daily and circadian patterns has been observed from the level of the genome to the whole organism, the functional and structural impact of biological rhythms, including mechanisms such as circadian misalignment, remains relatively poorly defined. Here, we review the impact of daily rhythms and circadian systems in regulating blood pressure and the onset, progression, and consequences of hypertension. There is an emphasis on the impact of circadian biology in relation to vascular disease and end-organ effects that, individually or in combination, contribute to complex phenotypes such as cognitive decline and the loss of cardiac and brain health. Despite effective treatment options for some individuals, control of blood pressure remains inadequate in a substantial portion of the hypertensive population. Greater insight into circadian biology may form a foundation for novel and more widely effective molecular therapies or interventions to help in the prevention, treatment, and management of hypertension and its related pathophysiology.
Collapse
Affiliation(s)
- Frank M. Faraci
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
- Department of Neuroscience and Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
| | - Frank A.J.L. Scheer
- Division of Sleep Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, 02115
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, Massachusetts, 02115
| |
Collapse
|
33
|
Allison EY, Al-Khazraji BK. Cerebrovascular adaptations to habitual resistance exercise with aging. Am J Physiol Heart Circ Physiol 2024; 326:H772-H785. [PMID: 38214906 PMCID: PMC11221804 DOI: 10.1152/ajpheart.00625.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/13/2024]
Abstract
Resistance training (RT) is associated with improved metabolism, bone density, muscular strength, and lower risk of osteoporosis, sarcopenia, and cardiovascular disease. Although RT imparts many physiological benefits, cerebrovascular adaptations to chronic RT are not well defined. Participation in RT is associated with greater resting peripheral arterial diameters, improved endothelial function, and general cardiovascular health, whereas simultaneously linked to reductions in central arterial compliance. Rapid blood pressure fluctuations during resistance exercise, combined with reduced arterial compliance, could lead to cerebral microvasculature damage and subsequent cerebral hypoperfusion. Reductions in cerebral blood flow (CBF) accompany normal aging, where chronic reductions in CBF are associated with changes in brain structure and function, and increased risk of neurodegeneration. It remains unclear whether reductions in arterial compliance with RT relate to subclinical cerebrovascular pathology, or if such adaptations require interpretation in the context of RT specifically. The purpose of this narrative review is to synthesize literature pertaining to cerebrovascular adaptations to RT at different stages of the life span. This review also aims to identify gaps in the current understanding of the long-term impacts of RT on cerebral hemodynamics and provide a mechanistic rationale for these adaptations as they relate to aging, cerebral vasculature, and overall brain health.
Collapse
Affiliation(s)
- Elric Y Allison
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Baraa K Al-Khazraji
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
A. Shirsath M, O'Connor JD, Boyle R, Newman L, Knight SP, Hernandez B, Whelan R, Meaney JF, Kenny RA. Slower speed of blood pressure recovery after standing is associated with accelerated brain aging: Evidence from The Irish Longitudinal Study on Ageing (TILDA). CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100212. [PMID: 38445293 PMCID: PMC10912350 DOI: 10.1016/j.cccb.2024.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
Background Impaired recovery of blood pressure (BP) in response to standing up is a prevalent condition in older individuals. We evaluated the relationship between the early recovery of hemodynamic responses to standing and brain health in adults over 50. Methods Participants from The Irish Longitudinal Study on Ageing (TILDA) (n=411; age 67.6 ± 7.3 years; 53.4 % women) performed an active stand challenge while blood pressure and heart rate were continuously monitored. The recovery of these parameters was determined as the slope of the BP and HR response, following the initial drop/rise after standing. We have previously reported a novel and validated measure of brain ageing using MRI data, which measures the difference between biological brain age and chronological age, providing a brain-predicted age difference (brainPAD) score. Results Slower recovery of systolic and diastolic BP was found to be significantly associated with higher brainPAD scores (i.e., biologically older brains), where a one-year increase in brainPAD was associated with a decrease of 0.02 mmHg/s and 0.01 mmHg/s in systolic and diastolic BP recovery, respectively, after standing. Heart rate (HR) recovery was not significantly associated with brainPAD score. Conclusion These results demonstrate that slower systolic and diastolic BP recovery in the early phase after standing is associated with accelerated brain aging in older individuals. This suggests that the BP response to standing, measured using beat-to-beat monitoring, has the potential to be used as a marker of accelerated brain aging, relying on a simple procedure and devices that are easily accessible.
Collapse
Affiliation(s)
- Morgana A. Shirsath
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - John D. O'Connor
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
- School of Engineering, Ulster University, Northern Ireland, UK
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Louise Newman
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Silvin P. Knight
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Belinda Hernandez
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Robert Whelan
- Trinity College Institute of Neuroscience, Trinity College, University of Dublin, Ireland
- Global Brain Health Institute, Trinity College, Trinity College Dublin, Ireland
| | - James F. Meaney
- National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| |
Collapse
|
35
|
Vipin A, Lee BTK, Kumar D, Soo SA, Leow YJ, Ghildiyal S, Lee FPHE, Hilal S, Kandiah N. The role of perfusion, grey matter volume and behavioural phenotypes in the data-driven classification of cognitive syndromes. Alzheimers Res Ther 2024; 16:40. [PMID: 38368378 PMCID: PMC10874041 DOI: 10.1186/s13195-024-01410-1] [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: 07/23/2023] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND The use of structural and perfusion brain imaging in combination with behavioural information in the prediction of cognitive syndromes using a data-driven approach remains to be explored. Here, we thus examined the contribution of brain structural and perfusion imaging and behavioural features to the existing classification of cognitive syndromes using a data-driven approach. METHODS Study participants belonged to the community-based Biomarker and Cognition Cohort Study in Singapore who underwent neuropsychological assessments, structural-functional MRI and blood biomarkers. Participants had a diagnosis of cognitively normal (CN), subjective cognitive impairment (SCI), mild cognitive impairment (MCI) and dementia. Cross-sectional structural and cerebral perfusion imaging, behavioural scale data including mild behaviour impairment checklist, Pittsburgh Sleep Quality Index and Depression, Anxiety and Stress scale data were obtained. RESULTS Three hundred seventy-three participants (mean age 60.7 years; 56% female sex) with complete data were included. Principal component analyses demonstrated that no single modality was informative for the classification of cognitive syndromes. However, multivariate glmnet analyses revealed a specific combination of frontal perfusion and temporo-frontal grey matter volume were key protective factors while the severity of mild behaviour impairment interest sub-domain and poor sleep quality were key at-risk factors contributing to the classification of CN, SCI, MCI and dementia (p < 0.0001). Moreover, the glmnet model showed best classification accuracy in differentiating between CN and MCI cognitive syndromes (AUC = 0.704; sensitivity = 0.698; specificity = 0.637). CONCLUSIONS Brain structure, perfusion and behavioural features are important in the classification of cognitive syndromes and should be incorporated by clinicians and researchers. These findings illustrate the value of using multimodal data when examining syndrome severity and provide new insights into how cerebral perfusion and behavioural impairment influence classification of cognitive syndromes.
Collapse
Affiliation(s)
- Ashwati Vipin
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - Bernett Teck Kwong Lee
- Centre for Biomedical Informatics, 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Dilip Kumar
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - See Ann Soo
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - Yi Jin Leow
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - Smriti Ghildiyal
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - Faith Phemie Hui En Lee
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore
| | - Saima Hilal
- Saw Swee Hock School of Public Health, Tahir Foundation Building, 12 Science Drive 2, National University of Singapore and National University Health System, Singapore, 117549, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Nagaendran Kandiah
- Dementia Research Centre (Singapore), 11 Mandalay Road, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, 308232, Singapore.
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
36
|
Frenzel S, Bülow R, Dörr M, Felix SB, Friedrich N, Völzke H, Wittfeld K, Grabe HJ, Bahls M. Left ventricular hypertrophy as a risk factor for accelerated brain aging: Results from the Study of Health in Pomerania. Hum Brain Mapp 2024; 45:e26567. [PMID: 38391110 PMCID: PMC10885183 DOI: 10.1002/hbm.26567] [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: 08/28/2023] [Revised: 11/16/2023] [Accepted: 11/30/2023] [Indexed: 02/24/2024] Open
Abstract
Previous studies provided evidence for the importance of cardiac structure abnormalities, in particular greater left ventricular (LV) mass, for brain aging, but longitudinal studies are lacking to date. We included 926 individuals (median age 48 years; 53% women) from the TREND cohort of the Study of Health in Pomerania (SHIP) without reduced ejection fraction or a history of myocardial infarction. LV mass index (LVMI) was determined by echocardiography at baseline. Brain morphometric measurements were derived from magnetic resonance images at baseline and 7-year follow-up. Direct effects of baseline LVMI on brain morphometry at follow-up were estimated using linear regression models with adjustment for baseline brain morphometry. At baseline, median LVMI was 40 g/m2.7 and 241 individuals (26%) met the criterion of LV hypertrophy. After correction for multiple testing, baseline LVMI was directly associated with reduced global cortical thickness and increased cortical brain age at follow-up independent from hypertension and blood pressure. Exposure-outcome relations were nonlinear and significantly stronger in the upper half of the exposure distribution. Specifically, an increase in baseline LVMI from the 50% quantile to the 95% quantile was associated additional 2.7 years (95% confidence interval = [1.5 years, 3.8 years]) of cortical brain age at follow-up. Additional regional analyses yielded bilateral effects on multiple frontal cortical regions. Our findings highlight the role of cardiac structure in brain aging. LVMI constitutes an easily measurable marker that might help to identify persons at risk for cognitive impairment and dementia.
Collapse
Affiliation(s)
- Stefan Frenzel
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Robin Bülow
- Institute of Diagnostic Radiology and NeuroradiologyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Marcus Dörr
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Stephan B. Felix
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- Institute for Community MedicineUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Katharina Wittfeld
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Center for Neurodegenerative Disease (DZNE), Partner Site Rostock/GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Hans J. Grabe
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Center for Neurodegenerative Disease (DZNE), Partner Site Rostock/GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| | - Martin Bahls
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GreifswaldGreifswaldMecklenburg‐Western PomeraniaGermany
| |
Collapse
|
37
|
van den Beukel TC, Wolters FJ, Siebert U, Spiering W, Ikram MA, Vernooij MW, de Jong PA, Bos D. Intracranial arteriosclerosis and the risk of dementia: A population-based cohort study. Alzheimers Dement 2024; 20:869-879. [PMID: 37814499 PMCID: PMC10916985 DOI: 10.1002/alz.13496] [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: 06/17/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND The impact of intracranial arteriosclerosis on dementia remains largely unclear. METHODS In 2339 stroke-free and dementia-free participants (52.2% women, mean age 69.5 years) from the general population, we assessed intracranial carotid artery calcification (ICAC) and vertebrobasilar artery calcification (VBAC) as proxy for arteriosclerosis. Associations with dementia were assessed using Cox models. In addition, indirect effects through cerebral small vessel disease (cSVD) and subcortical brain structure volumes were assessed using causal mediation analyses. RESULTS During a median of 13.4 years (25th-75th percentiles 9.9-14.5) of follow-up, 282 participants developed dementia. Both ICAC presence (hazard ratio [HR]: 1.53, 95% confidence interval [CI]: 1.00-2.32]) and volume (HR per standard deviation: 1.19, 95% CI: 1.01-1.40) increased dementia risk. For VBAC, severe calcifications increased dementia risk (HR for third vs first volume tertile: 1.89, 95% CI: 1.00-3.59). These effects were mediated partly through increased cSVD (percentage mediated for ICAC: 13% and VBAC: 24%). DISCUSSION Intracranial arteriosclerosis increases the risk of dementia.
Collapse
Affiliation(s)
- Tim C. van den Beukel
- Department of EpidemiologyErasmus Medical CenterRotterdamCAThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamCAThe Netherlands
- Department of Radiology and Nuclear MedicineUniversity Medical Center UtrechtUtrechtGAThe Netherlands
| | - Frank J. Wolters
- Department of EpidemiologyErasmus Medical CenterRotterdamCAThe Netherlands
- Department of NeurologyErasmus Medical CenterRotterdamCAThe Netherlands
- Alzheimer CenterErasmus Medical CenterRotterdamCAThe Netherlands
| | - Uwe Siebert
- Center for Health Decision Science, Departments of Epidemiology and Health Policy & ManagementHarvard T.H. Chan School of Public Health, BostonBostonMassachusettsUSA
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research and Health Technology AssessmentUMIT TIROL ‐ University for Health Sciences and TechnologyAustria
- Program on Cardiovascular Research, Institute for Technology Assessment and Department of Radiology, Massachusetts General HospitalHarvard Medical School, BostonBostonMassachusettsUSA
| | - Wilko Spiering
- Department of Vascular MedicineUniversity Medical Center UtrechtUtrechtGAThe Netherlands
| | - M. Arfan Ikram
- Department of EpidemiologyErasmus Medical CenterRotterdamCAThe Netherlands
| | - Meike W. Vernooij
- Department of EpidemiologyErasmus Medical CenterRotterdamCAThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamCAThe Netherlands
| | - Pim A. de Jong
- Department of Radiology and Nuclear MedicineUniversity Medical Center UtrechtUtrechtGAThe Netherlands
| | - Daniel Bos
- Department of EpidemiologyErasmus Medical CenterRotterdamCAThe Netherlands
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamCAThe Netherlands
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
38
|
Roh DJ, Murguia-Fuentes R, Gurel K, Khasiyev F, Rahman S, Bueno PP, Kozii K, Spagnolo-Allende AJ, Cottarelli A, Simonetto M, Ji R, Guo J, Spektor V, Hod EA, Burke DJ, Konofagou E, Rundek T, Wright CB, Marshall RS, Elkind MSV, Gutierrez J. Relationships of Hematocrit With Chronic Covert and Acute Symptomatic Lacunar Ischemic Lesions. Neurology 2024; 102:e207961. [PMID: 38165319 PMCID: PMC10870744 DOI: 10.1212/wnl.0000000000207961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/11/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Red blood cell (RBC) concentrations are known to associate with ischemic stroke. It is unclear whether RBC concentrations associate specifically with small vessel disease lacunar infarcts. We investigated the hypothesis that RBC concentrations associate with both chronic covert and acute symptomatic brain MRI lacunar infarcts. METHODS A cross-sectional observational analysis was performed across 2 cohorts with available hematocrit (as the assessment of RBC concentration exposure) and MRI outcome data. The primary setting was a population-based cohort of stroke-free, older adult (>50 years) participants from the Northern Manhattan Study (NOMAS) enrolled between 2003 and 2009. A second replication sample consisted of patients admitted with acute stroke and enrolled into the Columbia Stroke Registry (CSR) between 2005 and 2020. Associations of hematocrit with (1) chronic, covert lacunar infarcts and (2) symptomatic (i.e., acute) lacunar strokes were separately assessed from the NOMAS and CSR cohorts, respectively, using general additive models after adjusting for relevant covariates. RESULTS Of 1,218 NOMAS participants analyzed, 6% had chronic, covert lacunar infarcts. The association between hematocrit and these covert lacunar infarcts was U-shaped (χ2 = 9.21 for nonlinear associations; p = 0.03), with people with hematocrit extremes being more likely to have covert lacunar infarcts. Of the 1,489 CSR patients analyzed, 23% had acute lacunar strokes. In this sample, only the relationships of increased hematocrit concentrations and lacunar strokes were replicated (adjusted coefficient β = 0.020; SE = 0.009; p = 0.03). DISCUSSION We identified relationships of hematocrit with MRI lacunar infarcts in both stroke-free and ischemic stroke cohorts, respectively. The relationship between increased hematocrit concentrations with lacunar infarcts was replicated in both cohorts. Further studies are required to clarify the mechanisms behind the relationships of hematocrit with ischemic cerebral small vessel disease.
Collapse
Affiliation(s)
- David J Roh
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Ricardo Murguia-Fuentes
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Kursat Gurel
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Farid Khasiyev
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Salwa Rahman
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Pedro Paiva Bueno
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Khrystyna Kozii
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Antonio J Spagnolo-Allende
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Azzurra Cottarelli
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Marialaura Simonetto
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Robin Ji
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jia Guo
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Vadim Spektor
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Eldad A Hod
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Devin J Burke
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Elisa Konofagou
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Tatjana Rundek
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Clinton B Wright
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Randolph S Marshall
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Mitchell S V Elkind
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jose Gutierrez
- From the Departments of Neurology (D.J.R., K.G., S.R., P.P.B., K.K., A.J.S.-A., D.J.B., R.S.M., J. Gutierrez), Pathology and Cell Biology (A.C., E.A.H.), Biomedical Engineering (R.J., E.K.), Psychiatry (J. Guo), and Department of Radiology (V.S.), Vagelos College of Physicians and Surgeons, Columbia University, New York, NY; Department of Neurology (R.M.-F.), Louisiana State University Health Shreveport; Department of Neurology (F.K.), St. Louis University, MO; Department of Neurology (M.S.), Weill Cornell Medical Center, New York, NY; Department of Neurology (T.R.), University of Miami/Jackson Memorial Hospital, FL; National Institute of Neurological Disorders and Stroke (C.B.W.),, Bethesda, MD; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| |
Collapse
|
39
|
Öksüz N, Ghouri R, Taşdelen B, Uludüz D, Özge A. Mild Cognitive Impairment Progression and Alzheimer's Disease Risk: A Comprehensive Analysis of 3553 Cases over 203 Months. J Clin Med 2024; 13:518. [PMID: 38256652 PMCID: PMC10817043 DOI: 10.3390/jcm13020518] [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: 12/11/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
This study aimed to elucidate the long-term progression of mild cognitive impairment (MCI) within a comprehensive longitudinal dataset, distinguish it from healthy aging, explore the influence of a dementia subtype on this progression, and identify potential contributing factors. Patients with prodromal and preclinical cases underwent regular neuropsychological assessments utilizing various tools. The study included a total of 140 participants with MCI, categorized into Alzheimer's disease (AD) and non-AD subtypes. Our dataset revealed an overall progression rate of 92.8% from MCI to the clinical stage of dementia during the follow-up period, with an annual rate of 15.7%. Notably, all prodromal cases of Lewy body dementia/Parkinson's disease (LBD/PDD) and frontotemporal dementia (FTD) advanced to clinical stages, whereas 7% of vascular dementia (VaD) cases and 8.4% of AD cases remained in the prodromal stage throughout follow-up. Furthermore, we observed a faster progression rate in MCI-AD cases compared to non-AD sufferers (53.9% vs. 35.5%, Entropy: 0.850). This study revealed significant cognitive changes in individuals with MCI over time. The mini-mental state examination (MMSE), global deterioration scale (GDS), and calculation tests were the most effective tests for evaluation of MCI. These findings may offer valuable insights for the development of personalized interventions and management strategies for individuals with MCI.
Collapse
Affiliation(s)
- Nevra Öksüz
- Department of Neurology, School of Medicine, Mersin University, Mersin 33110, Turkey; (N.Ö.); (R.G.)
| | - Reza Ghouri
- Department of Neurology, School of Medicine, Mersin University, Mersin 33110, Turkey; (N.Ö.); (R.G.)
| | - Bahar Taşdelen
- Department of Biostatistics, School of Medicine, Mersin University, Mersin 33110, Turkey;
| | - Derya Uludüz
- Department of Neurology, Brain 360 Holistic Approach Center, İstanbul 34353, Turkey;
| | - Aynur Özge
- Department of Neurology, School of Medicine, Mersin University, Mersin 33110, Turkey; (N.Ö.); (R.G.)
| |
Collapse
|
40
|
Fitzgibbon-Collins LK, Coombs GB, Noguchi M, Parihar S, Hughson RL, Borrie M, Peters S, Shoemaker JK, Bhangu J. Standing middle cerebral artery velocity predicts cognitive function and gait speed in older adults with cognitive impairment, and is impacted by sex differences. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100198. [PMID: 38298456 PMCID: PMC10827680 DOI: 10.1016/j.cccb.2023.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 02/02/2024]
Abstract
Upright posture challenges the cerebrovascular system, leading to changes in middle cerebral artery velocity (MCAv) dynamics which are less evident at supine rest. Chronic alterations in MCAv have been linked to hypoperfusion states and the effect that this may have on cognition remains unclear. This study aimed to determine if MCAv and oscillatory metrics of MCAv (ex. pulsatility index, PI) during upright posture are i) associated with cognitive function and gait speed (GS) to a greater extent than during supine rest, and ii) are different between sexes. Beat-by-beat MCAv (transcranial Doppler ultrasound) and mean arterial pressure (MAP, plethysmography) were averaged for 30-seconds during supine-rest through a transition to standing for 53 participants (73±6yrs, 17 females). While controlling for age, multiple linear regressions predicting MoCA scores and GS from age, supine MCAv metrics, and standing MCAv metrics, were completed. Simple linear regressions predicting Montreal Cognitive Assessment (MoCA) score and GS from MCAv metrics were performed separately for females and males. Significance was set to p<0.05. Lower standing diastolic MCAv was a significant (p = 0.017) predictor of lower MoCA scores in participants with mild cognitive impairment, and this relationship only remained significant for males. Lower standing PI was associated with slower GS (p = 0.027, r=-0.306) in both sexes. Our results indicate a relationship between blunted MCAv and altered oscillatory flow profiles during standing, with lower MoCA scores and GS. These relationships were not observed in the supine position, indicating a unique relationship between standing measures of MCAv with cognitive and physical functions.
Collapse
Affiliation(s)
- Laura K Fitzgibbon-Collins
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5C1, Canada
- Department of Kinesiology, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 3K7, Canada
| | - Geoff B Coombs
- Department of Kinesiology, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 3K7, Canada
| | - Mamiko Noguchi
- Department of Kinesiology, University of Waterloo, 200 University Ave W., Waterloo, Ontario N2L 3G1, Canada
| | - Shashankdhwaj Parihar
- Cognitive Clinical Research Group, Parkwood Institute, 550 Wellington Rd., London, Ontario N6C 0A7, Canada
| | - Richard L Hughson
- Schlegel-University of Waterloo Research Institute for Aging, University of Waterloo, 250 Laurelwood Dr., Waterloo, Ontario N2J 0E2, Canada
| | - Michael Borrie
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5C1, Canada
| | - Sue Peters
- School of Physical Therapy, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 3K7, Canada
| | - J Kevin Shoemaker
- Department of Kinesiology, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 3K7, Canada
| | - Jaspreet Bhangu
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5C1, Canada
- Department of Kinesiology, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 3K7, Canada
| |
Collapse
|
41
|
Zhang M, Zhang Z, Li H, Xia Y, Xing M, Xiao C, Cai W, Bu L, Li Y, Park TE, Tang Y, Ye X, Lin WJ. Blockage of VEGF function by bevacizumab alleviates early-stage cerebrovascular dysfunction and improves cognitive function in a mouse model of Alzheimer's disease. Transl Neurodegener 2024; 13:1. [PMID: 38173017 PMCID: PMC10763201 DOI: 10.1186/s40035-023-00388-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the predominant type of dementia worldwide. It is characterized by the progressive and irreversible decline of cognitive functions. In addition to the pathological beta-amyloid (Aβ) deposition, glial activation, and neuronal injury in the postmortem brains of AD patients, increasing evidence suggests that the often overlooked vascular dysfunction is an important early event in AD pathophysiology. Vascular endothelial growth factor (VEGF) plays a critical role in regulating physiological functions and pathological changes in blood vessels, but whether VEGF is involved in the early stage of vascular pathology in AD remains unclear. METHODS We used an antiangiogenic agent for clinical cancer treatment, the humanized monoclonal anti-VEGF antibody bevacizumab, to block VEGF binding to its receptors in the 5×FAD mouse model at an early age. After treatment, memory performance was evaluated by a novel object recognition test, and cerebral vascular permeability and perfusion were examined by an Evans blue assay and blood flow scanning imaging analysis. Immunofluorescence staining was used to measure glial activation and Aβ deposits. VEGF and its receptors were analyzed by enzyme-linked immunosorbent assay and immunoblotting. RNA sequencing was performed to elucidate bevacizumab-associated transcriptional signatures in the hippocampus of 5×FAD mice. RESULTS Bevacizumab treatment administered from 4 months of age dramatically improved cerebrovascular functions, reduced glial activation, and restored long-term memory in both sexes of 5×FAD mice. Notably, a sex-specific change in different VEGF receptors was identified in the cortex and hippocampus of 5×FAD mice. Soluble VEGFR1 was decreased in female mice, while full-length VEGFR2 was increased in male mice. Bevacizumab treatment reversed the altered expression of receptors to be comparable to the level in the wild-type mice. Gene Set Enrichment Analysis of transcriptomic changes revealed that bevacizumab effectively reversed the changes in the gene sets associated with blood-brain barrier integrity and vascular smooth muscle contraction in 5×FAD mice. CONCLUSIONS Our study demonstrated the mechanistic roles of VEGF at the early stage of amyloidopathy and the protective effects of bevacizumab on cerebrovascular function and memory performance in 5×FAD mice. These findings also suggest the therapeutic potential of bevacizumab for the early intervention of AD.
Collapse
Affiliation(s)
- Min Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhan Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Honghong Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Mengdan Xing
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Chuan Xiao
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Wenbao Cai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Lulu Bu
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yi Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yamei Tang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
42
|
Rajan RK, Kumar RP, Ramanathan M. Piceatannol improved cerebral blood flow and attenuated JNK3 and mitochondrial apoptotic pathway in a global ischemic model to produce neuroprotection. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:479-496. [PMID: 37470802 DOI: 10.1007/s00210-023-02616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Cerebral ischemia is one of the leading causes of death and disability worldwide. The only FDA-approved treatment is recanalization with systemic tissue plasminogen activators like alteplase, although reperfusion caused by recanalization can result in neuroinflammation, which can cause brain cell apoptosis. Therefore, after an ischemic/reperfusion injury, interventions are needed to minimize the neuroinflammatory cascade. In the present study, piceatannol (PCT) was studied for its neuroprotective efficacy in a rat model of global ischemic injury by attenuating c-Jun N-terminal kinase 3 (JNK3) downstream signaling. PCT is a resveratrol analog and a polyphenolic stilbenoid naturally occurring in passion fruit and grapes. The neuroprotective efficacy of PCT (1, 5, 10 mg/kg) in ischemic conditions was assessed through pre- and post-treatment. Cerebral blood flow (CBF) and tests for functional recovery were assessed. Protein and gene expression were done for JNK3 and other inflammatory markers. A docking study was performed to identify the amino acid interaction. The results showed that PCT improved motor and memory function as measured by a functional recovery test believed to be due to an increase in cerebral blood flow. Also, the caspase signaling which promotes apoptosis was found to be down-regulated; however, nitric oxide synthase expression was up-regulated, which could explain the enhanced cerebral blood flow (CBF). According to our findings, PCT impeded c-Jun N-terminal kinase 3 (JNK3) signaling by suppressing phosphorylation and disrupting the mitochondrial apoptotic pathway, which resulted in the neuroprotective effect. Molecular docking analysis was performed to investigate the atomic-level interaction of JNK3 and PCT, which reveals that Met149, Leu206, and Lys93 amino acid residues are critical for the interaction of PCT and JNK3. According to our current research, JNK3 downstream signaling and the mitochondrial apoptosis pathway are both inhibited by PCT, which results in neuroprotection under conditions of global brain ischemia. Piceatannol attenuated JNK3 phosphorylation during the ischemic condition and prevented neuronal apoptosis.
Collapse
Affiliation(s)
- Ravi Kumar Rajan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India.
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Girijananda Chowdhury University, Dekargaon, Tezpur, 784501, Assam, India.
| | - Ram Pravin Kumar
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| | - M Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| |
Collapse
|
43
|
Madsen LS, Kjeldsen PL, Ismail R, Parbo P, Østergaard L, Brooks DJ, Eskildsen SF. Capillary dysfunction in healthy elderly APOE ε4 carriers with raised brain Aβ deposition. Alzheimers Dement 2024; 20:459-471. [PMID: 37679610 PMCID: PMC10917038 DOI: 10.1002/alz.13461] [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: 06/09/2023] [Revised: 07/26/2023] [Accepted: 08/13/2023] [Indexed: 09/09/2023]
Abstract
INTRODUCTION Capillary dysfunction, characterized by disturbances in capillary blood flow distribution, might be an overlooked factor in the development of Alzheimer's disease (AD). This study investigated microvascular blood flow in preclinical and prodromal AD individuals. METHODS Using dynamic susceptibility contrast magnetic resonance imaging and positron emission tomography, we examined alterations in microvascular circulation and levels of Aβ deposition in two independent cohorts of APOE ε4 carriers. RESULTS Capillary dysfunction was elevated in both prodromal and preclinical AD individuals compared to age-matched controls. Additionally, the prodromal group exhibited higher levels of capillary dysfunction compared to the preclinical group. DISCUSSION These findings suggest that capillary dysfunction can be detected at the preclinical stage of AD and indicates a worsening of capillary dysfunction throughout the AD continuum. Understanding the interaction between capillary dysfunction and Aβ could provide insights into the relationship between cardiovascular risk factors and the development of AD. HIGHLIGHTS Alzheimer's disease (AD) is associated with disturbances in microvascular circulation. Capillary dysfunction can be detected in preclinical AD. As cognitive symptoms progress in prodromal AD, capillary dysfunction worsens. Capillary dysfunction may impede the clearance of beta-amyloid (Aβ). Capillary dysfunction might contribute to the development of AD.
Collapse
Affiliation(s)
- Lasse S. Madsen
- Center of Functionally Integrative NeuroscienceDepartment of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Nuclear Medicine and PET‐CentreAarhus University HospitalAarhusDenmark
| | - Pernille L. Kjeldsen
- Department of Nuclear Medicine and PET‐CentreAarhus University HospitalAarhusDenmark
| | - Rola Ismail
- Department of Nuclear MedicineSygehus LillebaeltVejleDenmark
| | - Peter Parbo
- Department of Nuclear MedicineOdense University HospitalOdenseDenmark
| | - Leif Østergaard
- Center of Functionally Integrative NeuroscienceDepartment of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of NeuroradiologyAarhus University HospitalAarhusDenmark
| | - David J. Brooks
- Department of Nuclear Medicine and PET‐CentreAarhus University HospitalAarhusDenmark
- Institute of Translational and Clinical ResearchUniversity of Newcastle upon TyneNewcastle upon TyneUK
| | - Simon F. Eskildsen
- Center of Functionally Integrative NeuroscienceDepartment of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
44
|
Mu S, Lu W, Yu G, Zheng L, Qiu J. Deep learning-based grading of white matter hyperintensities enables identification of potential markers in multi-sequence MRI data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 243:107904. [PMID: 37924768 DOI: 10.1016/j.cmpb.2023.107904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 10/06/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are widely-seen in the aging population, which are associated with cerebrovascular risk factors and age-related cognitive decline. At present, structural atrophy and functional alterations coexisted with WMHs lacks comprehensive investigation. This study developed a WMHs risk prediction model to evaluate WHMs according to Fazekas scales, and to locate potential regions with high risks across the entire brain. METHODS We developed a WMHs risk prediction model, which consisted of the following steps: T2 fluid attenuated inversion recovery (T2-FLAIR) image of each participant was firstly segmented into 1000 tiles with the size of 32 × 32 × 1, features from the tiles were extracted using the ResNet18-based feature extractor, and then a 1D convolutional neural network (CNN) was used to score all tiles based on the extracted features. Finally, a multi-layer perceptron (MLP) was constructed to predict the Fazekas scales based on the tile scores. The proposed model was trained using T2-FLAIR images, we selected tiles with abnormal scores in the test set after prediction, and evaluated their corresponding gray matter (GM) volume, white matter (WM) volume, fractional anisotropy (FA), mean diffusivity (MD), and cerebral blood flow (CBF) via longitudinal and multi-sequence Magnetic Resonance Imaging (MRI) data analysis. RESULTS The proposed WMHs risk prediction model could accurately predict the Fazekas ratings based on the tile scores from T2-FLAIR MRI images with accuracy of 0.656, 0.621 in training data set and test set, respectively. The longitudinal MRI validation revealed that most of the high-risk tiles predicted by the WMHs risk prediction model in the baseline images had WMHs in the corresponding positions in the longitudinal images. The validation on multi-sequence MRI demonstrated that WMHs were associated with GM and WM atrophies, WM micro-structural and perfusion alterations in high-risk tiles, and multi-modal MRI measures of most high-risk tiles showed significant associations with Mini Mental State Examination (MMSE) score. CONCLUSION Our proposed WMHs risk prediction model can not only accurately evaluate WMH severities according to Fazekas scales, but can also uncover potential markers of WMHs across modalities. The WMHs risk prediction model has the potential to be used for the early detection of WMH-related alterations in the entire brain and WMH-induced cognitive decline.
Collapse
Affiliation(s)
- Si Mu
- College of Mechanical and Electronic Engineering, Shandong Agricultural University, Tai'an, Shandong, 271000, China
| | - Weizhao Lu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Guanghui Yu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Lei Zheng
- Department of Radiology, Rushan Hospital of Chinese Medicine, Rushan, Shandong, 264500, China.
| | - Jianfeng Qiu
- School of Radiology, Shandong First Medical University & Shandong Academy of Medicine Sciences, Tai'an, Shandong, 271000, China; Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
45
|
Zhang Z, Lim MJR. Incident Dementia After Spontaneous Intracerebral Hemorrhage. J Alzheimers Dis 2024; 99:41-51. [PMID: 38640161 DOI: 10.3233/jad-240111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Post-stroke cognitive impairment and dementia (PSCID) is a complication that affects long-term functional outcomes after stroke. Studies on dementia after long-term follow-up in stroke have focused predominantly on ischemic stroke, which may be different from the development of dementia after spontaneous intracerebral hemorrhage (ICH). In this review, we summarize the existing data and hypotheses on the development of dementia after spontaneous ICH, review the management of post-ICH dementia, and suggest areas for future research. Dementia after spontaneous ICH has a cumulative incidence of up to 32.0-37.4% at 5 years post-ICH. Although the pathophysiology of post-ICH dementia has not been fully understood, two main theoretical frameworks can be considered: 1) the triggering role of ICH (both primary and secondary brain injury) in precipitating cognitive decline and dementia; and 2) the contributory role of pre-existing brain pathology (including small vessel disease and neurodegenerative pathology), reduced cognitive reserve, and genetic factors predisposing to cognitive dysfunction. These pathophysiological pathways may have synergistic effects that converge on dysfunction of the neurovascular unit and disruptions in functional connectivity leading to dementia post-ICH. Management of post-ICH dementia may include screening and monitoring, cognitive therapy, and pharmacotherapy. Non-invasive brain stimulation is an emerging therapeutic modality under investigation for safety and efficacy. Our review highlights that there remains a paucity of data and standardized reporting on incident dementia after spontaneous ICH. Further research is imperative for determining the incidence, risk factors, and pathophysiology of post-ICH dementia, in order to identify new therapies for the treatment of this debilitating condition.
Collapse
Affiliation(s)
- Zheting Zhang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | |
Collapse
|
46
|
Feldman DR, Zeitler EP. Neurologic impact of atrial fibrillation. Curr Opin Cardiol 2024; 39:33-38. [PMID: 37678332 DOI: 10.1097/hco.0000000000001093] [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] [Indexed: 09/09/2023]
Abstract
PURPOSE OF REVIEW Cognitive dysfunction is a complex condition that is becoming increasingly more prevalent. There has been growing acknowledgement that individuals with atrial fibrillation are at an increased risk of cognitive dysfunction beyond the association of age with both disorders. The purpose of this review is to explore the potential underlying mechanisms connecting atrial fibrillation and cognitive dysfunction and to examine the existing evidence for potential treatment options. RECENT FINDINGS Many mechanisms have been proposed for the association between cognitive dysfunction and atrial fibrillation. These include cerebral infarction (both micro and macro embolic events), cerebral microbleeds including those secondary to therapeutic anticoagulation, an increased inflammatory state, cerebral hypoperfusion, and a genetic predisposition to both diseases. Treatments designed to target each of these mechanisms have led to mixed results and there are no specific interventions that have definitively led to a reduction in the incidence of cognitive dysfunction. SUMMARY The relationship between cognitive dysfunction and atrial fibrillation remains poorly understood. Standard of care currently focuses on reducing risk factors, managing stroke risk, and maintaining sinus rhythm in appropriately selected patients. Further work needs to be conducted in this area to limit the progression of cognitive dysfunction in patients with atrial fibrillation.
Collapse
Affiliation(s)
| | - Emily P Zeitler
- Dartmouth Health and Dartmouth-Hitchcock Medical Center
- The Dartmouth Institute, Lebanon, New Hampshire, USA
| |
Collapse
|
47
|
Amin G, Booz GW, Zouein FA. Proteinopathy: Shared Feature Between the Heart and Brain in Alzheimer's Disease. J Cardiovasc Pharmacol 2024; 83:4-7. [PMID: 37890458 PMCID: PMC10842240 DOI: 10.1097/fjc.0000000000001501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ghadir Amin
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Pharmacology and Toxicology, American University of Beirut Medical Center, Faculty of Medicine, Beirut, Lebanon
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| |
Collapse
|
48
|
Ding H, Liu C, Li Y, Ang TFA, Devine S, Liu Y, Au R, Doraiswamy PM. Sex-specific blood biomarkers linked to memory changes in middle-aged adults: The Framingham Heart Study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12569. [PMID: 38545543 PMCID: PMC10966919 DOI: 10.1002/dad2.12569] [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: 08/25/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 06/06/2024]
Abstract
The relationship between sex-specific blood biomarkers and memory changes in middle-aged adults remains unclear. We aimed to investigate this relationship using the data from the Framingham Heart Study (FHS). We conducted association analysis, partial correlation analysis, and causal dose-response curves using blood biomarkers and other data from 793 middle-aged participants (≤ 60 years) from the FHS Offspring Cohort. The results revealed associations of adiponectin and fasting blood glucose with midlife memory change, along with a U-shaped relationship of high-density lipoprotein cholesterol with memory change. No significant associations were found for the other blood biomarkers (e.g., amyloid beta protein 42) with memory change. To our knowledge, this is the first sex-specific network analysis of blood biomarkers related to midlife memory change in a prospective cohort study. Our findings highlight the importance of targeting cardiometabolic risks and the need to validate midlife-specific biomarkers that can accelerate the development of primary preventive strategies.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Chunyu Liu
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Yi Li
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Ting Fang Alvin Ang
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Sherral Devine
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Yulin Liu
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Rhoda Au
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - P. Murali Doraiswamy
- Neurocognitive Disorders ProgramDepartments of Psychiatry and Medicineand the Duke Institute for Brain SciencesDuke University School of MedicineDurhamNorth CarolinaUSA
| |
Collapse
|
49
|
Rowsthorn E, Pham W, Nazem-Zadeh MR, Law M, Pase MP, Harding IH. Imaging the neurovascular unit in health and neurodegeneration: a scoping review of interdependencies between MRI measures. Fluids Barriers CNS 2023; 20:97. [PMID: 38129925 PMCID: PMC10734164 DOI: 10.1186/s12987-023-00499-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The neurovascular unit (NVU) is a complex structure that facilitates nutrient delivery and metabolic waste clearance, forms the blood-brain barrier (BBB), and supports fluid homeostasis in the brain. The integrity of NVU subcomponents can be measured in vivo using magnetic resonance imaging (MRI), including quantification of enlarged perivascular spaces (ePVS), BBB permeability, cerebral perfusion and extracellular free water. The breakdown of NVU subparts is individually associated with aging, pathology, and cognition. However, how these subcomponents interact as a system, and how interdependencies are impacted by pathology remains unclear. This systematic scoping review identified 26 studies that investigated the inter-relationships between multiple subcomponents of the NVU in nonclinical and neurodegenerative populations using MRI. A further 112 studies investigated associations between the NVU and white matter hyperintensities (WMH). We identify two putative clusters of NVU interdependencies: a 'vascular' cluster comprising BBB permeability, perfusion and basal ganglia ePVS; and a 'fluid' cluster comprising ePVS, free water and WMH. Emerging evidence suggests that subcomponent coupling within these clusters may be differentially related to aging, neurovascular injury or neurodegenerative pathology.
Collapse
Affiliation(s)
- Ella Rowsthorn
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Turner Institute for Brain and Mental Health & School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton, VIC, 3168, Australia
| | - William Pham
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mohammad-Reza Nazem-Zadeh
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Radiology, Alfred Health, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Electrical and Computer Systems Engineering, Monash University, 14 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health & School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton, VIC, 3168, Australia
- Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Ian H Harding
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Monash Biomedical Imaging, Monash University, 762-772 Blackburn Road, Clayton, VIC, 3168, Australia.
| |
Collapse
|
50
|
You TY, Dong Q, Cui M. Emerging Links between Cerebral Blood Flow Regulation and Cognitive Decline: A Role for Brain Microvascular Pericytes. Aging Dis 2023:AD.2022.1204. [PMID: 37163446 PMCID: PMC10389833 DOI: 10.14336/ad.2022.1204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/04/2022] [Indexed: 05/12/2023] Open
Abstract
Cognitive impairment associated with vascular etiology has been of considerable interest in the development of dementia. Recent studies have started to uncover cerebral blood flow deficits in initiating cognitive deterioration. Brain microvascular pericytes, the only type of contractile cells in capillaries, are involved in the precise modulation of vascular hemodynamics due to their ability to regulate resistance in the capillaries. They exhibit potential in maintaining the capillary network geometry and basal vascular tone. In addition, pericytes can facilitate better blood flow supply in response to neurovascular coupling. Their dysfunction is thought to disturb cerebral blood flow causing metabolic imbalances or structural injuries, leading to consequent cognitive decline. In this review, we summarize the characteristics of microvascular pericytes in brain blood flow regulation and outline the framework of a two-hit hypothesis in cognitive decline, where we emphasize how pericytes serve as targets of cerebral blood flow dysregulation that occurs with neurological challenges, ranging from genetic factors, aging, and pathological proteins to ischemic stress.
Collapse
Affiliation(s)
- Tong-Yao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|