1
|
Rossetti GMK, Dunster JL, Sohail A, Williams B, Cox KM, Rawlings S, Jewett E, Benford E, Lovegrove JA, Gibbins JM, Christakou A. Evidence for control of cerebral neurovascular function by circulating platelets in healthy older adults. J Physiol 2025; 603:3379-3404. [PMID: 40434152 DOI: 10.1113/jp288405] [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: 01/07/2025] [Accepted: 04/10/2025] [Indexed: 05/29/2025] Open
Abstract
Platelets play a vital role in preventing haemorrhage through haemostasis, but complications arise when platelets become overly reactive, leading to pathophysiology such as atherothrombosis. Elevated haemostatic markers are linked to dementia and predict its onset in long-term studies. Despite epidemiological evidence, the mechanism linking haemostasis with early brain pathophysiology remains unclear. Here, we aimed to determine whether a mechanistic association exists between platelet function and cerebral neurovascular function in 52 healthy mid- to older-age adults. To do this, we combined, for the first time, magnetic resonance imaging of cerebral neurovascular function, peripheral vascular physiology and in vitro platelet assaying. We show an association between platelet reactivity and cerebral neurovascular function that is both independent of vascular reactivity and mechanistically specific: Distinct platelet signalling mechanisms (ADP, collagen-related peptide, thrombin receptor activator peptide 6) were associated with different physiological components of the haemodynamic response to neuronal (visual) stimulation (full-width half-maximum, time to peak, area under the curve), an association that was not mediated by peripheral vascular effects. This finding challenges the previous belief that systemic vascular health determines the vascular component of cerebral neurovascular function, highlighting a specific link between circulating platelets and the neurovascular unit. Because altered cerebral neurovascular function marks the initial stages of neurodegenerative pathophysiology, understanding this novel association is now imperative, with the potential to lead to a significant advancement in our comprehension of early dementia pathophysiology. KEY POINTS: Haemostasis (platelet function) has been linked to the early stages of dementia, but the precise mechanisms are not well understood. This study considers whether a causal mechanism exists through atherothrombotic effects on the vasculature which can in turn affect brain health, or through platelet-specific interactions with brain physiology. Here, we show that elevated platelet reactivity is associated with blunted (delayed, shorter and smaller) cerebral blood flow responses to neuronal activation in healthy middle-aged and older adults. However, the association between platelet reactivity and cerebral neurovascular function was not mediated by systemic vascular reactivity. This finding challenges the previous belief that systemic vascular health determines the vascular component of cerebral neurovascular function, highlighting a specific link between circulating platelets and the neurovascular unit in early dementia pathophysiology.
Collapse
Affiliation(s)
- Gabriella M K Rossetti
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute of Sport, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, UK
| | - Joanne L Dunster
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Aamir Sohail
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Centre for Human Brain Health (CHBH), School of Psychology, University of Birmingham, Birmingham, UK
| | - Brendan Williams
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - Kiera M Cox
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Suzannah Rawlings
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Elysia Jewett
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Eleanor Benford
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Julie A Lovegrove
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
- Institute of Food and Nutritional Health (IFNH), Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Anastasia Christakou
- Centre for Integrative Neuroscience and Neurodynamics (CINN), School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| |
Collapse
|
2
|
Ashley J, Pasha EP, Tarumi T, Tomoto T, Zhang R. Lower diastolic velocity in the internal carotid artery mediates lower cerebral blood flow with age. J Appl Physiol (1985) 2025; 138:1385-1397. [PMID: 40353819 DOI: 10.1152/japplphysiol.00847.2024] [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: 10/30/2024] [Revised: 11/24/2024] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Advanced aging is characterized by reduced cerebral blood flow (CBF) and increased central arterial stiffness. Increased arterial stiffness is associated with increased CBF pulsatility, which is detrimental to cerebrovascular integrity. We examined the associations between central arterial stiffness, diastolic, systolic, and total CBF in healthy cognitively normal subjects (n = 163, age 20-81 yr, 62% female) who underwent color-coded duplex ultrasonography of the internal carotid (ICA) and the vertebral artery (VA) to measure pulsatile CBF and total CBF. Cerebral tissue oxygenation was measured using near-infrared spectroscopy (NIRS). Carotid β-stiffness index and carotid-femoral pulse wave velocity (cfPWV) were assessed via applanation tonometry and ultrasonography to assess central artery stiffness. Age was negatively associated with total CBF (R2 = 0.268, P < 0.001). ICA diastolic velocity was negatively associated with cfPWV (R2 = 0.163, P < 0.001) and carotid β-stiffness index (R2 = 0.134, P < 0.001). Higher carotid β-stiffness index was associated with lower CBF with age, which was mediated through lower ICA diastolic velocity. These findings suggest that central arterial stiffness with age may lead to reductions in ICA diastolic velocity, contributing to a reduction in CBF.NEW & NOTEWORTHY In this study, we demonstrated that higher central arterial stiffness is associated with lower diastolic velocity of the ICA, which mediates lower CBF with age. These findings provide new mechanistic insights into the reduction in CBF with age and add to the mounting evidence for the importance of age-related central arterial stiffening in brain health.
Collapse
Affiliation(s)
- John Ashley
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Evan P Pasha
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Takashi Tarumi
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Tsubasa Tomoto
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
3
|
Xu C, Wang Y, Tang C, Chen H, Cui L, Yu X, Wang F. Enhancing cognitive and emotional well-being in older adults through aerobic, strength, or balance training: insights from a randomized controlled trial. Acta Psychol (Amst) 2025; 257:105068. [PMID: 40381540 DOI: 10.1016/j.actpsy.2025.105068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Late-life depression (LLD) poses a significant public health challenge, profoundly affecting cognitive function and emotional well-being in the elderly. Current pharmacological treatments often have side effects, increasing interest in non-pharmacological interventions. OBJECTIVE This study evaluates the effectiveness of a 12-week aerobic, strength, or balance training intervention on depressive symptoms and cognitive function in older adults with mild to moderate LLD while also investigating the impact of different training frequencies (3×/week or 5×/week). METHODS We conducted a randomized controlled trial (RCT) involving 121 community-dwelling older adults aged 60 years and above diagnosed with mild to moderate LLD. Stratified randomization will be conducted based on baseline depression severity and key characteristics, dividing patients into the intervention groups (drug therapy and standard care combined with aerobic exercise, strength training, or balance training interventions, respectively) and the control group (drug therapy and standard care). The exercise intervention comprised aerobic, strength or balance exercises conducted 3×/week or 5×/week. Depression severity and cognitive function were assessed using the Hamilton Depression Rating Scale (HAMD-17) and Mini-Mental State Examination (MMSE) at baseline, after 12 weeks, and after a four-week follow-up. Additionally, physiological parameters, including weight, blood pressure (systolic and diastolic), resting heart rate, and fasting blood glucose levels, were assessed simultaneously to evaluate overall health improvements. Statistical analysis was performed using repeated measures ANOVA to assess within- and between-group changes. RESULTS The intervention group significantly improved depressive symptoms, cognitive function, and physiological parameters. Specifically, the HAMD-17 scores decreased by 6.5 ± 2.8 points (p < 0.001), while the MMSE scores increased by 5.7 ± 2.4 points (p < 0.001), indicating reduced depressive symptom severity and improved cognitive function, respectively. These improvements were significantly greater than those observed in the control group. Notably, high-frequency aerobic exercise (5×/week) yielded the most significant benefits, showing a reduction of 8.6 ± 2.7 points in HAMD-17 and an increase of 6.8 ± 2.1 points in MMSE (both p < 0.001). CONCLUSION The aerobic, strength, or balance training regimen effectively reduces depressive symptoms and improves cognitive function in older adults with LLD, offering a promising non-pharmacological treatment option. This study underscores the potential of exercise interventions to complement traditional therapies, paving the way for more holistic treatment strategies for late-life depression.
Collapse
Affiliation(s)
- Chen Xu
- Psychiatry Department, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Yuejia Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Nursing Department of Huzhou Third Municipal Hospital, Huzhou, Zhejiang 313000, China
| | - Chunxia Tang
- Psychiatry Department, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Haiqin Chen
- Psychogeriatrics Department, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Lijun Cui
- Key Laboratory of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Xuefen Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Fei Wang
- Psychogeriatrics Department, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China.
| |
Collapse
|
4
|
Palmer JA, Kaufman CS, Whitaker‐Hilbig AA, Billinger SA. APOE4 carriers display loss of anticipatory cerebrovascular regulation across the Alzheimer's disease continuum. Alzheimers Dement 2025; 21:e70229. [PMID: 40346723 PMCID: PMC12064416 DOI: 10.1002/alz.70229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Maintenance of cerebral blood flow during orthostasis is impaired with aging and associated with cognitive decline, but the effect of the apolipoprotein ɛ4 allele (APOE4) is unknown. METHODS Older adults (n = 108) (APOE4 carriers, n = 47; non-carriers, n = 61) diagnosed as having normal cognition (NC), mild cognitive impairment (MCI), or Alzheimer's disease (AD) underwent transcranial Doppler ultrasound assessment of middle cerebral artery blood velocity (MCAv) and beat-to-beat mean arterial blood pressure (MAP) during a sit-to-stand transition. Anticipatory and orthostasis-induced MCAv and MAP responses were compared between genotypes and diagnostic classifications. 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 non-carriers. MCAv and MAP responses were delayed and attenuated across the APOE4 disease continuum, with no differences between genotypes in MCI and AD. DISCUSSION Unique cerebral and peripheral vascular compensation observed in APOE4 carriers may be neuroprotective for AD development. HIGHLIGHTS APOE4 carriers with NC show greater anticipatory increases in MCAv prior to orthostasis and decreases during orthostasis. APOE4 carriers with NC show faster peripheral MAP responses during orthostasis. APOE4 carriers with MCI and AD display loss of anticipatory MCAv responses. APOE4 carriers with MCI and AD display slower peripheral MAP responses. Unique cerebral and peripheral vascular compensation observed in APOE4 carriers may be neuroprotective for AD development.
Collapse
Affiliation(s)
- Jacqueline A. Palmer
- Division of Physical Therapy and Rehabilitation ScienceUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- University of Kansas Alzheimer's Disease Research CenterFairwayKansasUSA
| | - Carolyn S. Kaufman
- Department of Molecular & Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Department of Internal MedicineStanford Health CareStanford UniversityPalo AltoCaliforniaUSA
| | - Alicen A. Whitaker‐Hilbig
- Department of Physical Medicine and RehabilitationMedical College of WisconsinMilwaukeeWisconsinUSA
- Cardiovascular CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Sandra A. Billinger
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- University of Kansas Alzheimer's Disease Research CenterFairwayKansasUSA
| |
Collapse
|
5
|
Farina S, Cattabiani A, Mandge D, Shichkova P, Isbister JB, Jacquemier J, King JG, Markram H, Keller D. A multiscale electro-metabolic model of a rat neocortical circuit reveals the impact of ageing on central cortical layers. PLoS Comput Biol 2025; 21:e1013070. [PMID: 40393041 DOI: 10.1371/journal.pcbi.1013070] [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: 12/11/2024] [Revised: 05/27/2025] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
The high energetic demands of the brain arise primarily from neuronal activity. Neurons consume substantial energy to transmit information as electrical signals and maintain their resting membrane potential. These energetic requirements are met by the neuro-glial-vascular (NGV) ensemble, which generates energy in a coupled metabolic process. In ageing, metabolic function becomes impaired, producing less energy and, consequently, the system is unable to sustain the neuronal energetic needs. We propose a multiscale model of electro-metabolic coupling in a reconstructed rat neocortex. This combines an electro-morphologically reconstructed electrophysiological model with a detailed NGV metabolic model. Our results demonstrate that the large-scale model effectively captures electro-metabolic processes at the circuit level, highlighting the importance of heterogeneity within the circuit, where energetic demands vary according to neuronal characteristics. Finally, in metabolic ageing, our model indicates that the middle cortical layers are particularly vulnerable to energy impairment.
Collapse
Affiliation(s)
- Sofia Farina
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Alessandro Cattabiani
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Darshan Mandge
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Polina Shichkova
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
- Biognosys AG, Schlieren, Switzerland
| | - James B Isbister
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Jean Jacquemier
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - James G King
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
- Brain Mind Institute, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Daniel Keller
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
6
|
Liu H, Sun Z, Zeng H, Han J, Hu M, Mao D, Tian X, Li R. Meta-analysis of the effects of multi-component exercise on cognitive function in older adults with cognitive impairment. Front Aging Neurosci 2025; 17:1551877. [PMID: 40297493 PMCID: PMC12034652 DOI: 10.3389/fnagi.2025.1551877] [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: 12/26/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Exercise has been widely recognized as an effective regimen in mitigating cognitive decline. However, the effect of multi-component exercise (i.e., combination of two or more types of exercise) on cognitive function and its subdomains in older adults remains unclear. This meta-analysis aimed to explore the effects of multi-component exercise on cognitive functions in elderly individuals with cognitive impairment and identify optimal prevention and treatment strategies. A systematic search was conducted on PubMed, EBSCOhost, Web of Science, and Embase to identify relevant randomized controlled trials assessing the effect of multi-component exercise on cognitive function in the elderly. Thirteen studies with 1,776 participants were included in the analysis using Revman 5.4 software. The results showed that multi-component exercise had a significant effect on mitigating cognitive function decline in the elderly, with a pooled effect size of SMD = 0.31 (95% CI: 0.08, 0.55; p = 0.009). The results of subgroup analysis showed that interventions with ≥3 days/week, 12-24 weeks duration, and ≤ 40 min/session were significantly superior to other frequencies, durations, and lengths, with all p-values <0.05. Additionally, multi-component exercise had the most pronounced effects on executive function, visual memory, and verbal memory in patients with mild cognitive impairment (MCI). In conclusion, multi-component exercise can delay the decline in cognitive function in the elderly, and the intervention effects are modulated by various variables. Optimal intervention effects were observed with an exercise frequency of three or more times per week, a duration of 12 to 24 weeks, and a time per session of 40 min or less, particularly for improving executive function, visual memory, and verbal memory in patients with MCI.
Collapse
Affiliation(s)
- Hualei Liu
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| | - Zhiyuan Sun
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
- Qufu Normal University, Jining, Shandong, China
| | - Haiqing Zeng
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| | - Jincheng Han
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| | - Mengqi Hu
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| | - Dewei Mao
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
- The Chinese University of Hong Kong, Shenzhen, China
| | - Xuewen Tian
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| | - Ran Li
- Shandong Sports Science Research Institute, Shandong Sports University, Jinan, Shandong, China
| |
Collapse
|
7
|
Dalakoti M, Chen CK, Sia CH, Poh KK. Frontiers in subclinical atherosclerosis and the latest in early life preventive cardiology. Singapore Med J 2025; 66:141-146. [PMID: 40116060 PMCID: PMC11991069 DOI: 10.4103/singaporemedj.smj-2024-169] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/01/2024] [Indexed: 03/23/2025]
Abstract
ABSTRACT Subclinical atherosclerosis underlies most cardiovascular diseases, manifesting before clinical symptoms and representing a key focus for early prevention strategies. Recent advancements highlight the importance of early detection and management of subclinical atherosclerosis. This review underscores that traditional risk factor levels considered safe, such as low-density lipoprotein cholesterol (LDL-C) and glycated haemoglobin (HbA1c), may still permit the development of atherosclerosis, suggesting a need for stricter thresholds. Early-life interventions are crucial, leveraging the brain's neuroplasticity to establish lifelong healthy habits. Preventive strategies should include more aggressive management of LDL-C and HbA1c from youth and persist into old age, supported by public health policies that promote healthy environments. Emphasising early education on cardiovascular health can fundamentally shift the trajectory of cardiovascular disease prevention and optimise long-term health outcomes.
Collapse
Affiliation(s)
- Mayank Dalakoti
- Cardiovascular Metabolic Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
- Department of Medicine, Ng Teng Fong General Hospital, Singapore
| | - Ching Kit Chen
- Cardiovascular Metabolic Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat – National University Children’s Medical Institute, National University Health System, Singapore
| | - Ching-Hui Sia
- Cardiovascular Metabolic Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| |
Collapse
|
8
|
Won J, Tomoto T, Tarumi T, Rodrigue KM, Kennedy KM, Park DC, Zhang R. Associations of cardiorespiratory fitness with cerebral cortical thickness and gray matter volume across the adult lifespan. J Appl Physiol (1985) 2025; 138:473-482. [PMID: 39818996 PMCID: PMC12179780 DOI: 10.1152/japplphysiol.00429.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/10/2024] [Accepted: 01/05/2025] [Indexed: 01/19/2025] Open
Abstract
High cardiorespiratory fitness (CRF) is associated with reduced cortical thinning and gray matter (GM) shrinkage in older adults. We investigated associations of CRF measured with peak oxygen consumption (V̇o2peak) with cortical thickness and GM volume across the adult lifespan. We hypothesized that higher CRF is associated with less cortical thinning and GM shrinkage across the adult lifespan, which is associated with better cognitive performance. This cross-sectional study recruited 172 sedentary yet healthy adults (65% women, 22-81 yr) who underwent treadmill exercise testing to measure V̇o2peak, structural magnetic resonance imaging to assess cortical thickness and GM volume, and a comprehensive cognitive test battery to assess fluid cognitive function. Linear regression models were used to examine the associations of total and regional cortical thickness and GM volume with age, V̇o2peak, and age × V̇o2peak interaction after adjusting for sex, education, and total intracranial volume, and the associations of cortical thickness and GM volume with fluid cognitive performance. Mean and regional cortical thickness and total GM volume were associated negatively with age, whereas no associations were observed with V̇o2peak. However, a significant interaction between age and V̇o2peak on the right superior parietal volume indicated that aging was associated with smaller right superior parietal volume in the lower CRF group, whereas no association was observed in the higher CRF group. Larger right superior parietal volume was associated with better fluid cognitive performance. These findings highlight the importance of maintaining CRF to prevent or slow brain aging from an adult lifespan perspective.NEW & NOTEWORTHY High cardiorespiratory fitness may mitigate regional gray matter shrinkage across the adult lifespan.
Collapse
Affiliation(s)
- Junyeon Won
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX, USA
| | - Tsubasa Tomoto
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Takashi Tarumi
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Karen M. Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Kristen M. Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Denise C. Park
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Rong Zhang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Brinkley TE, Garcia KR, Mitchell GF, Tegeler CH, Sarwal A, Bennett J, Kitzman DW, Leng I, Baker LD, Espeland MA, Snyder HM, Claassen JA, Bailey MJ, Shaltout HA. The U.S. POINTER neurovascular ancillary study: Study design and methods. Alzheimers Dement 2025; 21:e14574. [PMID: 39992278 PMCID: PMC11849405 DOI: 10.1002/alz.14574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025]
Abstract
INTRODUCTION POINTER Neurovascular (POINTER-NV) is an ancillary study that leverages the rich infrastructure and design of the U.S. Study to Protect Brain Health through Lifestyle Intervention to Reduce Risk (U.S. POINTER) to investigate neurovascular mechanisms that may underlie intervention effects on key brain outcomes. METHODS A comprehensive neurovascular assessment is conducted at baseline, Month 12, and Month 24 using a variety of complementary non-invasive techniques including transcranial Doppler ultrasound, carotid ultrasound, echocardiography, tonometry, and continuous blood pressure and heart rate monitoring. Measurements are acquired at rest and during orthostatic challenges, hyperventilation, and carbon dioxide inhalation. RESULTS The primary outcomes are baroreflex sensitivity and cerebral autoregulation. Secondary outcomes include aortic, carotid, and cerebral hemodynamics and various measures of autonomic function and vascular structure and function. DISCUSSION POINTER-NV will provide critical insight into neurovascular mechanisms that may change with intensive lifestyle modification and promote improvements in cognition and overall brain health. HIGHLIGHTS This study takes advantage of U.S. Study to Protect Brain Health through Lifestyle Intervention to Reduce Risk (U.S. POINTER) to address key gaps in the field. POINTER Neurovascular (POINTER-NV) will provide insight into neurovascular mechanisms underlying dementia. POINTER-NV may help shed light on modifiable vascular contributions to dementia.
Collapse
Affiliation(s)
- Tina E. Brinkley
- Department of Internal MedicineSection on Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Katelyn R. Garcia
- Department of Biostatistics and Data ScienceDivision of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | - Charles H. Tegeler
- Department of NeurologyWake Forest University School of Medicine, Medical Center BlvdWinston‐SalemNorth CarolinaUSA
| | - Aarti Sarwal
- Department of NeurologyWake Forest University School of Medicine, Medical Center BlvdWinston‐SalemNorth CarolinaUSA
| | - John Bennett
- Department of NeurologyWake Forest University School of Medicine, Medical Center BlvdWinston‐SalemNorth CarolinaUSA
| | - Dalane W. Kitzman
- Department of Internal MedicineSection on Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Internal MedicineSection on Cardiovascular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Iris Leng
- Department of Biostatistics and Data ScienceDivision of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Laura D. Baker
- Department of Internal MedicineSection on Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Social Sciences and Health PolicyDivision of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Mark A. Espeland
- Department of Internal MedicineSection on Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Heather M. Snyder
- Division of Medical and Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Jurgen A. Claassen
- Department of Geriatric MedicineRadboudumc Alzheimer CenterDonders Institute for BrainCognition and BehaviourRadboud University Medical CenterNijmegenNetherlands
| | - Margie J. Bailey
- Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Hossam A. Shaltout
- Hypertension and Vascular Research CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Obstetrics and GynecologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
10
|
Kalantari S, Soltani M, Maghbooli M, Khoshe Mehr FS, Kalantari Z, Borji S, Memari B, Hossein Heydari A, Elahi R, Bayat M, Salighehrad H. Cerebral blood flow alterations measured by ASL-MRI as a predictor of vascular dementia in small vessel ischemic disease. RADIOLOGIA 2025; 67:28-37. [PMID: 39978877 DOI: 10.1016/j.rxeng.2024.03.013] [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: 12/16/2023] [Accepted: 03/20/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND Cerebral small vessel ischemic disease (SVID) as a common age-related morbidity is the key mechanism of vascular cognitive impairment (VCI). This study uses Cerebral blood flow (CBF) measured by pseudo-continuous ASL MRI in SVID patients with and without cognitive impairment to differentiate VCI from normal aging. MATERIALS AND METHODS In this cross-sectional study, 74 SVID patients, including 35 with diagnosed VCI and 39 without cognitive impairment (control) underwent pCASL-MRI in the resting state. ROI-based approach pre-processing, denoising techniques, and correction for partial volume effects were performed. Regional CBF was compared between severe cognitive impairment (SCI), mild cognitive impairment (MCI), and SVID patients without cognitive impairment. RESULTS Total and regional CBF values in the thalamus, left cortex, hippocampus, post cingulate cortex, precuneus, insula, putamen, and middle temporal lobe was lower in VCI compared to SVID, also in SCI compared MCI group. There was a linear correlation between the Mini-Mental State Examination (MMSE) z score and CBF in the thalamus region in SVID participants and between the MMSE z score and CBF in the medial temporal region in MCI participants. The medial temporal atrophy )MTA( z score was significantly correlated with CBF values in the hippocampus and medial temporal regions in SCI and MCI also a significant correlation was seen between total CBF and Fazekas score. CONCLUSION Due to the growing prevalence of dementia and the role of CBF as a predictive biomarker, ASL-MRI as a non-invasive method can be easily added to diagnostic tools of cognitive impairment in individuals with SVID to recognize the initiation of vascular cognitive impairment.
Collapse
Affiliation(s)
- S Kalantari
- Departamento de Radiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - M Soltani
- Departamento de Radiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran.
| | - M Maghbooli
- Departamento de Radiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - F S Khoshe Mehr
- Departamento de Física Médica e Ingeniería Biomédica, Universidad de Ciencias Médicas de Teherán, Teheran, Iran
| | - Z Kalantari
- Departamento de Cardiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - S Borji
- Departamento de Radiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - B Memari
- Departamento de Radiología, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - A Hossein Heydari
- Facultad de Medicina, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran.
| | - R Elahi
- Facultad de Medicina, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - M Bayat
- Departamento de Radiología Técnica, Universidad de Ciencias Médicas de Zanjan, Zanjan, Iran
| | - H Salighehrad
- Grupo de Espectroscopia e Imágenes Cuantitativas por RM, Centro de Investigación de Imágenes Celulares y Moleculares, Universidad de Ciencias Médicas de Teherán, Teheran, Iran
| |
Collapse
|
11
|
Koep JL, Bond B, Taylor CE, Barker AR, Ruediger SL, Pizzey FK, Coombes JS, Bailey TG. The relationships between age, sex, and exercise intensity on cerebral artery hemodynamics during isometric handgrip exercise. Am J Physiol Regul Integr Comp Physiol 2025; 328:R1-R20. [PMID: 39437543 DOI: 10.1152/ajpregu.00014.2024] [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: 01/19/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Age and sex may alter the cerebral blood flow (CBF) responses to acute isometric exercise, via associated elevations in mean arterial pressure (MAP) and sympathetic activation. Our aim was to determine the relationships between age, sex, and exercise intensity on cerebrovascular responses to isometric handgrip exercise. In 78 healthy adults (18-80 yr, n = 42 females), cerebrovascular responses were assessed during 2-min isometric exercise bouts at three intensities [15, 30, 45% maximal voluntary contraction (MVC)]. Intracranial responses of the middle cerebral artery (MCA) and posterior cerebral artery (PCA) velocity (v) were measured using transcranial Doppler ultrasound. Extracranial responses of the internal carotid artery (ICA) and vertebral artery (VA) were assessed using Duplex ultrasound. Cardiopulmonary hemodynamic and neural parameters were measured throughout, including muscle sympathetic nerve activity, end-tidal carbon dioxide, and MAP. There were significant positive relationships between exercise intensity and the cerebral responses of the MCAv (P < 0.001) and PCAv (P = 0.005). There were no effects of intensity on ICA and VA responses (P > 0.05), despite intensity-dependent increases in MAP (P < 0.001). The increased MCAv response to exercise was blunted with advancing age (P = 0.01) with no influence of sex (P = 0.86). The present study provides data on age, sex, and intensity-specific relationships with intracranial and extracranial cerebrovascular responses to isometric exercise. Despite similar ICA, VA, and PCA responses, MCAv responses were attenuated with advancing age during handgrip exercise with no sex-dependent influence. Furthermore, intracranial responses were intensity dependent, whereas extracranial blood flow, shear-stress, and velocity responses were similarly increased at all intensities during handgrip exercise.NEW & NOTEWORTHY The influence of aging and sex on cerebral blood flow responses to isometric exercise are unknown. We observed intensity-dependent increases in velocity of the intracranial arteries, whereas the extracranial artery responses were similarly increased at all intensities during handgrip exercise in young and older individuals. Furthermore, we observed a blunted middle cerebral artery velocity response to handgrip exercise with advancing age, whereas the posterior circulation and extracranial responses were preserved across the lifespan in healthy individuals in males and females alike.
Collapse
Affiliation(s)
- Jodie L Koep
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- Children's Health and Exercise Research Centre, Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Bert Bond
- Children's Health and Exercise Research Centre, Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Chloe E Taylor
- School of Health Sciences, Western Sydney University, Sydney, New South Wales, Australia
| | - Alan R Barker
- Children's Health and Exercise Research Centre, Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Stefanie L Ruediger
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Faith K Pizzey
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jeff S Coombes
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Tom G Bailey
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- School of Nursing, Midwifery and Social Work, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Engstrom AC, Alitin JPM, 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 DJJ, Nation DA. Spontaneous cerebrovascular reactivity at rest in older adults with and without mild cognitive impairment and memory deficits. Alzheimers Dement 2025; 21:e14396. [PMID: 39555916 PMCID: PMC11772704 DOI: 10.1002/alz.14396] [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: 06/25/2024] [Revised: 09/25/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION 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 One hundred sixty-one older adults free of dementia or major neurological/psychiatric disorders were recruited. Participants underwent clinical interviews, cognitive testing, venipuncture for Alzheimer's disease (AD) biomarkers, and brain magnetic resonance imaging. Spontaneous CVR was quantified during 5 minutes of rest. Respiratory gases analyzed through nasal cannula to quantify end-tidal carbon dioxide (ETCO2) levels were used to estimate CVR. 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 AD biomarkers and vascular risk factors. DISCUSSION Spontaneous CVR deficits in the PHG are observed in older adults with MCI and memory impairment, suggesting medial temporal microvascular dysfunction is observed in cognitive decline. HIGHLIGHTS Aging is associated with decline in whole brain spontaneous cerebrovascular reactivity (CVR). Older adults with mild cognitive impairment exhibit deficits in spontaneous CVR in the parahippocampal gyrus (PHG). Memory impairment is correlated with reduced spontaneous CVR in the PHG.
Collapse
Affiliation(s)
- Allison C. Engstrom
- Department of Psychological ScienceUniversity of California, IrvineIrvineCaliforniaUSA
| | - John Paul M. Alitin
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Arunima Kapoor
- Department of Psychological ScienceUniversity of California, IrvineIrvineCaliforniaUSA
| | - Shubir Dutt
- Memory and Aging CenterWeill Institute for NeurosciencesDepartment of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Trevor Lohman
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Isabel J. Sible
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Anisa J. Marshall
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Fatemah Shenasa
- Department of Psychological ScienceUniversity of California, IrvineIrvineCaliforniaUSA
| | - Aimée Gaubert
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Farrah Ferrer
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Amy Nguyen
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - David Robert Bradford
- Center for Innovations in Brain ScienceDepartment of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen Rodgers
- Center for Innovations in Brain ScienceDepartment of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Lorena Sordo
- Department of Pathology and Laboratory MedicineUniversity of California, IrvineIrvineCaliforniaUSA
| | - Elizabeth Head
- Department of Pathology and Laboratory MedicineUniversity of California, IrvineIrvineCaliforniaUSA
| | - Xingfeng Shao
- Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Danny J. J. Wang
- Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Daniel A. Nation
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
13
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [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: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Fesharaki NJ, Taylor A, Mosby K, Li R, Kim JH, Ress D. Global Impact of Aging on the Hemodynamic Response Function in the Gray Matter of Human Cerebral Cortex. Hum Brain Mapp 2024; 45:e70100. [PMID: 39692126 PMCID: PMC11653092 DOI: 10.1002/hbm.70100] [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: 06/08/2024] [Revised: 10/20/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024] Open
Abstract
In functional magnetic resonance imaging, the hemodynamic response function (HRF) is a stereotypical response to local changes in cerebral hemodynamics and oxygen metabolism due to briefly (< 4 s) evoked neural activity. Accordingly, the HRF is often used as an impulse response with the assumption of linearity in data analysis. In cognitive aging studies, it has been very common to interpret differences in brain activation as age-related changes in neural activity. Contrary to this assumption, however, evidence has accrued that normal aging may also significantly affect the vasculature, thereby affecting cerebral hemodynamics and metabolism, confounding interpretation of fMRI cognitive aging studies. In this study, use was made of a multisensory task to evoke the HRF in ~87% of cerebral cortex in cognitively intact adults with ages ranging from 22 to 75 years. This widespread activation enabled us to investigate age trends in the spatial distributions of HRF characteristics within the majority of cortical gray matter, which we termed as global age trends. The task evoked both positive and negative HRFs, which were characterized using model-free parameters in native-space coordinates. We found significant global age trends in the distributions of HRF parameters in terms of both amplitudes (e.g., peak amplitude and contrast-to-noise ratio) and temporal dynamics (e.g., full-width-at-half-maximum). Our findings offer insight into how age-dependent changes affect neurovascular coupling and show promise for use of HRF parameters as non-invasive indicators for age-related pathology.
Collapse
Affiliation(s)
- Nooshin J. Fesharaki
- Department of NeurosurgeryUniversity of Texas Health Science CenterHoustonTexasUSA
- Department of Neuroscience, High Resolution Brain Imaging LabBaylor College of MedicineHoustonTexasUSA
| | - Amanda Taylor
- Department of Neuroscience, High Resolution Brain Imaging LabBaylor College of MedicineHoustonTexasUSA
| | - Keisjon Mosby
- Department of Neuroscience, High Resolution Brain Imaging LabBaylor College of MedicineHoustonTexasUSA
| | - Ruosha Li
- Department of NeurosurgeryUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Jung Hwan Kim
- Department of NeurosurgeryUniversity of Texas Health Science CenterHoustonTexasUSA
| | - David Ress
- Department of Neuroscience, High Resolution Brain Imaging LabBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
15
|
da C. Pinaffi-Langley AC, Szarvas Z, Peterfi A, Kaposzta Z, Mukli P, Shahriari A, Muranyi M, Pinto CB, Owens CD, Adams C, Karfonta B, Rohan M, Tarantini S, Yabluchanskiy A. Time-restricted eating for prevention of age-related vascular cognitive decline in older adults: A protocol for a single-arm open-label interventional trial. PLoS One 2024; 19:e0314871. [PMID: 39652561 PMCID: PMC11627372 DOI: 10.1371/journal.pone.0314871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024] Open
Abstract
Age-related cerebromicrovascular endothelial dysfunction underlies the initiation and progression of cognitive dysfunction and dementia, thus increasing the susceptibility of older adults to such conditions. Normal brain function requires dynamic adjustment of cerebral blood flow to meet the energetic demands of active neurons, which is achieved the homeostatic mechanism neurovascular coupling (NVC). In this context, therapeutical strategies aimed at rescuing or preserving NVC responses can delay the incidence or mitigate the severity of age-related cognitive dysfunction, and time-restricted eating (TRE) is a potential candidate for such a strategy. Studies have reported that TRE can improve cardiometabolic risk factors in older adults. However, the effect of TRE on cerebrovascular endothelial function remains unexplored. Thus, this protocol outlines the study procedures to test our hypothesis that a 6-month TRE regimen of 10-h eating window will improve NVC responses and endothelial function in community-dwelling older adults. This is a single-arm, open-label interventional trial. We aim to recruit 32 adults aged 55-80 years. Participants are instructed to maintain a TRE regimen of 10 h of free eating followed by 14 h of fasting for 6 months. Before and after fasting, participants are assessed for cognitive performance, peripheral micro- and macrovascular endothelial function, and NVC responses, as well as for several confounding factors, including body composition, dietary, and physical activity data. We expect that 6 months of TRE will improve NVC response and endothelial function in older adults compared with baseline, and that these improvements will be accompanied by improvements in cognitive performance. The study proposed herein will provide critical insight into a new potential therapeutical strategy for targeting age-related cognitive dysfunction. Ultimately, slowing down or alleviating cognitive decline will translate into improved quality of life and longer healthspan for aging adults. This study was prospectively registered at ClinicalTrials.gov (NCT06019195) on August 24, 2023.
Collapse
Affiliation(s)
- Ana Clara da C. Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Zalan Kaposzta
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Ali Shahriari
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Mihaly Muranyi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Camila B. Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Cheryl Adams
- Oklahoma Shared Clinical and Translational Resources, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Brittany Karfonta
- Oklahoma Shared Clinical and Translational Resources, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Michael Rohan
- Laureate Institute for Brain Research, Tulsa, OK, United States of America
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| |
Collapse
|
16
|
Rane Levendovszky S, Flores J, Peskind ER, Václavů L, van Osch MJP, Iliff J. Preliminary investigations into human neurofluid transport using multiple novel non-contrast MRI methods. J Cereb Blood Flow Metab 2024; 44:1580-1592. [PMID: 39053490 PMCID: PMC11572104 DOI: 10.1177/0271678x241264407] [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: 07/10/2023] [Revised: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 07/27/2024]
Abstract
We discuss two potential non-invasive MRI methods to study phenomena related to subarachnoid cerebrospinal fluid (CSF) motion and perivascular fluid transport, and their association with sleep and aging. We apply diffusion-based intravoxel incoherent motion (IVIM) imaging to evaluate pseudodiffusion coefficient, D*, or CSF movement across large spaces like the subarachnoid space (SAS). We also performed perfusion-based multi-echo, Hadamard encoded arterial spin labeling (ASL) to evaluate whole brain cortical cerebral blood flow (CBF) and trans-endothelial exchange (Tex) of water from the vasculature into the perivascular space and parenchyma. Both methods were used in young adults (N = 9, 6 F, 23 ± 3 years old) in the setting of sleep and sleep deprivation. To study aging, 10 older adults (6 F, 67 ± 3 years old) were imaged after a night of normal sleep and compared with the young adults. D* in SAS was significantly (p < 0.05) reduced with sleep deprivation (0.016 ± 0.001 mm2/s) compared to normal sleep (0.018 ± 0.001 mm2/s) and marginally reduced with aging (0.017 ± 0.001 mm2/s, p = 0.029). Cortical CBF and Tex were unchanged with sleep deprivation but significantly lower in older adults (37 ± 3 ml/100 g/min, 578 ± 61 ms) than in young adults (42 ± 2 ml/100 g/min, 696 ± 62 ms). IVIM was sensitive to sleep physiology and aging, and multi-echo, multi-delay ASL was sensitive to aging.
Collapse
Affiliation(s)
| | - Jaqueline Flores
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elaine R Peskind
- VISN 20 Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Healthcare System, Seattle, WA, USA
| | - Lena Václavů
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias JP van Osch
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeffrey Iliff
- VISN 20 Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Healthcare System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
17
|
Cheng H, Ding D, Dai J, Li G, Zhang K, Li J, Wei L, Zhang X, Hou J. Effect of a reduced arterial axial pre-stretch ratio during aging on the cardiac output and cerebral blood flow in the healthy elders. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108468. [PMID: 39442288 DOI: 10.1016/j.cmpb.2024.108468] [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: 06/26/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND OBJECTIVE It is an indisputable physiological phenomenon that the arterial axial pre-stretch ratio (AAPSR) decreases with age, but little attention has been paid to the effect of this reduction on chronic diseases during aging. METHODS Here we reported an experimental method to simulate arteries aging, developed a fluid-structure interaction model with the effect of AAPSR changes, and compared it with the anatomy data and structural parameters of the human thoracic aorta. RESULTS We showed that with the process of aging, the decrease of AAPSR leads to a decline of arterial elasticity, a decrease of arterial elastic strain energy, which weakens the ability to promote blood circulation, the corresponding decrease in cardiac output (CO) and cerebral blood flow (CBF) causes distal organ and body tissue ischemia, which is one of the main causes of increased blood pressure and decreased cerebral perfusion in the elderly. CONCLUSIONS Thus, reduced AAPSR is the one of main manifestation of arteries aging and has an important impact on hypertension and hypoperfusion of the brain in the process of human aging. The research contributes to a better understanding of the physiological and pathological mechanisms of aging-related diseases.
Collapse
Affiliation(s)
- Heming Cheng
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China..
| | - Dongfang Ding
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Jifeng Dai
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Gen Li
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Ke Zhang
- Department of Hydraulic Engineering, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Jianyun Li
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Liuchuang Wei
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Xue Zhang
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Jie Hou
- Department of Mechanics, Kunming University of Science and Technology, Kunming 650500, PR China..
| |
Collapse
|
18
|
Ahrens E, Wachtendorf LJ, Hill KP, Schaefer MS. Considerations for Anesthesia in Older Adults with Cannabis Use. Drugs Aging 2024; 41:933-943. [PMID: 39617807 DOI: 10.1007/s40266-024-01161-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/12/2024]
Abstract
Over the past decade, legislative changes occurred in the USA and the western world that were followed by a substantial increase in reported use of cannabis among the general population. Among patients undergoing anesthesia for surgery or interventional procedures, older patients-often defined as adults over 65 years-are one of the fastest-growing populations. Within this group, the prevalence of cannabis use almost tripled over the past decade. In addition to habitual cannabis use, recommendations for treatment of chronic pain with cannabinoids have become increasingly more common. The clinical relevance of cannabis use in older adults is supported by recent studies linking it to increased anesthetic requirements as well as respiratory, cardiovascular, and psychiatric complications following surgery. Still, evidence remains equivocal, as these associations may largely depend on the type, frequency, and route of cannabis administration, and current research is mostly limited to retrospective cohort studies. Multisystemic effects of cannabis can become especially relevant in patients of advanced age undergoing anesthesia, characterized by physiological and pharmacodynamic alterations as well as a higher risks of drug-to-drug interactions. Best-practice guidelines emphasize the need for detailed, systematic preoperative screening for habits of cannabis use, including the history, type, and frequency, to guide perioperative management in these patients. This review discusses considerations for anesthesia in older patients with habitual cannabis use while highlighting strategies and recommendations to ensure safe and effective anesthesia care.
Collapse
Affiliation(s)
- Elena Ahrens
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Luca J Wachtendorf
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kevin P Hill
- Division of Addiction Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Maximilian S Schaefer
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Anesthesiology, Duesseldorf University Hospital, Duesseldorf, Germany.
| |
Collapse
|
19
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong E, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-related decline in blood-brain barrier function is more pronounced in males than females in parietal and temporal regions. eLife 2024; 13:RP96155. [PMID: 39495221 PMCID: PMC11534331 DOI: 10.7554/elife.96155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), and shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here, we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8-92 years old, using a non-invasive diffusion-prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60 s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Ophthalmology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brian T Gold
- Department of Neuroscience, College of Medicine, University of KentuckyFrankfortUnited States
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of MedicineBaltimoreUnited States
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell MedicineNew YorkUnited States
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
20
|
Guo B, Chen Y, Lin J, Huang B, Bai X, Guo C, Gao B, Gong Q, Bai X. Self-supervised learning for accurately modelling hierarchical evolutionary patterns of cerebrovasculature. Nat Commun 2024; 15:9235. [PMID: 39455566 PMCID: PMC11511858 DOI: 10.1038/s41467-024-53550-5] [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: 07/28/2023] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Cerebrovascular abnormalities are critical indicators of stroke and neurodegenerative diseases like Alzheimer's disease (AD). Understanding the normal evolution of brain vessels is essential for detecting early deviations and enabling timely interventions. Here, for the first time, we proposed a pipeline exploring the joint evolution of cortical volumes (CVs) and arterial volumes (AVs) in a large cohort of 2841 individuals. Using advanced deep learning for vessel segmentation, we built normative models of CVs and AVs across spatially hierarchical brain regions. We found that while AVs generally decline with age, distinct trends appear in regions like the circle of Willis. Comparing healthy individuals with those affected by AD or stroke, we identified significant reductions in both CVs and AVs, wherein patients with AD showing the most severe impact. Our findings reveal gender-specific effects and provide critical insights into how these conditions alter brain structure, potentially guiding future clinical assessments and interventions.
Collapse
Affiliation(s)
- Bin Guo
- Xiamen Key Laboratory of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, China
- Image Processing Center, Beihang University, Beijing, China
| | - Ying Chen
- Image Processing Center, Beihang University, Beijing, China
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jinping Lin
- Xiamen Key Laboratory of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, China
| | - Bin Huang
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Xiangzhuo Bai
- Zhongxiang Hospital of Traditional Chinese Medicine, Hubei, China
| | | | - Bo Gao
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Qiyong Gong
- Xiamen Key Laboratory of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China.
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China.
| | - Xiangzhi Bai
- Image Processing Center, Beihang University, Beijing, China.
- State Key Laboratory of Virtual Reality Technology and Systems, Beihang University, Beijing, China.
- Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China.
| |
Collapse
|
21
|
Shichkova P, Coggan JS, Markram H, Keller D. Brain Metabolism in Health and Neurodegeneration: The Interplay Among Neurons and Astrocytes. Cells 2024; 13:1714. [PMID: 39451233 PMCID: PMC11506225 DOI: 10.3390/cells13201714] [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: 08/01/2024] [Revised: 08/31/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
The regulation of energy in the brain has garnered substantial attention in recent years due to its significant implications in various disorders and aging. The brain's energy metabolism is a dynamic and tightly regulated network that balances energy demand and supply by engaging complementary molecular pathways. The crosstalk among these pathways enables the system to switch its preferred fuel source based on substrate availability, activity levels, and cell state-related factors such as redox balance. Brain energy production relies on multi-cellular cooperation and is continuously supplied by fuel from the blood due to limited internal energy stores. Astrocytes, which interface with neurons and blood vessels, play a crucial role in coordinating the brain's metabolic activity, and their dysfunction can have detrimental effects on brain health. This review characterizes the major energy substrates (glucose, lactate, glycogen, ketones and lipids) in astrocyte metabolism and their role in brain health, focusing on recent developments in the field.
Collapse
Affiliation(s)
- Polina Shichkova
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Jay S. Coggan
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
- Laboratory of Neural Microcircuitry, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| |
Collapse
|
22
|
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
|
23
|
Sugawara J, Tarumi T, Tomoto T, Pasha E, Cullum CM, Zhang R. Patients with amnestic mild cognitive impairment have higher cerebrovascular impedance than cognitively normal older adults. J Appl Physiol (1985) 2024; 137:848-856. [PMID: 39116348 PMCID: PMC11486476 DOI: 10.1152/japplphysiol.00337.2024] [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: 05/08/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Brain hypoperfusion is associated with cognitive impairment. Higher cerebrovascular impedance modulus (Z) may contribute to brain hypoperfusion. We tested hypotheses that patients with amnestic mild cognitive impairment (aMCI) (i.e., those who have a high risk of developing Alzheimer's disease) have higher Z than age-matched cognitively normal individuals, and that high Z is correlated with brain hypoperfusion. Fifty-eight patients with aMCI (67 ± 7 yr) and 25 cognitively normal subjects (CN, 65 ± 6 yr) underwent simultaneous measurements of carotid artery pressure (CAP, via applanation tonometry) and middle cerebral arterial blood velocity (CBV, via transcranial Doppler). Z was quantified using cross-spectral and transfer function analyses between dynamic changes in CBV and CAP. Patients with aMCI exhibited higher Z than NC (1.18 ± 0.34 vs. 1.01 ± 0.35 mmHg/cm/s, P = 0.044) in the frequency range from 0.78 to 4.29 Hz. The averaged Z in the frequency range (0.78-3.13 Hz) of high coherence (>0.9) was inversely correlated with total cerebral blood flow measured with 2-D Doppler ultrasonography normalized by the brain tissue mass (via structural MRI) across both patients with aMCI and NC (r = -0.311, P = 0.007), and in patients with aMCI alone (r = -0.306, P = 0.007). Our findings suggest that patients with aMCI have higher cerebrovascular impedance than cognitively normal older adults and that increased cerebrovascular impedance is associated with brain hypoperfusion.NEW & NOTEWORTHY This is the first study to compare cerebrovascular impedance between patients with amnestic mild cognitive impairment (aMCI) and age-matched cognitively normal individuals. Patients with aMCI had higher cerebrovascular impedance modulus than age-matched cognitively normal individuals, which was correlated with brain hypoperfusion. These results suggest the presence of cerebrovascular dysfunction in the dynamic regulation of cerebral blood flow in older adults who have high risks of Alzheimer's disease.
Collapse
Affiliation(s)
- Jun Sugawara
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Takashi Tarumi
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Tsubasa Tomoto
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
| | - Evan Pasha
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - C Munro Cullum
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
24
|
Li R, Zhou B, Deng X, Tian W, Huang Y, Wang J, Xu L. α-Klotho: the hidden link between dietary inflammatory index and accelerated ageing. Br J Nutr 2024; 132:558-564. [PMID: 39300827 DOI: 10.1017/s0007114524001417] [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: 09/22/2024]
Abstract
Recent studies suggest an association between greater dietary inflammatory index (DII) and higher biological ageing. As α-Klotho has been considered as a longevity protein, we examined whether α-Klotho plays a role in the association between DII and ageing. We included 3054 participants from the National Health and Nutrition Examination Survey. The associations of DII with biological and phenotypic age were assessed by multivariable linear regression, and the mediating role of α-Klotho was evaluated by mediation analyses. Participants' mean age was 58·0 years (sd 11·0), with a median DII score of 1·85 and interquartile range from 0·44 to 2·79. After adjusting for age, sex, race/ethnicity, BMI, education, marital status, poverty income ratio, serum cotinine, alcohol, physical activity, a higher DII was associated with both older biological age and phenotypic age, with per DII score increment being associated with a 1·01-year increase in biological age (1·01 (95 % CI: 1·005, 1·02)) and 1·01-year increase in phenotypic age (1·01 (1·001, 1·02)). Negative associations of DII with α-Klotho (β = -1·01 pg/ml, 95 % CI: -1·02, -1·006) and α-Klotho with biological age (β= -1·07 years, 95 % CI: -1·13, -1·02) and phenotypic age (β= -1·03 years, 95 % CI: -1·05, -1·01) were found. Furthermore, α-Klotho mediated 10·13 % (P < 0·001) and 9·61 % (P < 0·001) of the association of DII with biological and phenotypic age, respectively. Higher DII was associated with older biological and phenotypic age, and the potential detrimental effects could be partly mediated through α-Klotho.
Collapse
Affiliation(s)
- Ruiqiang Li
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Baijing Zhou
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xueqing Deng
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wenbo Tian
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yingyue Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiao Wang
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
- School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
25
|
Abuelazm M, Khildj Y, Ibrahim AA, Mahmoud A, Amin AM, Gowaily I, Khan U, Abdelazeem B, Brašić JR. Intensive Blood Pressure Control After Endovascular Thrombectomy for Acute Ischemic Stroke: a Systematic Review and Meta-Analysis. Clin Neuroradiol 2024; 34:563-575. [PMID: 38453701 PMCID: PMC11339153 DOI: 10.1007/s00062-024-01391-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND AND PURPOSE Optimal clinical outcome with successful recanalization from endovascular thrombectomy (EVT) requires optimal blood pressure (BP) management. We aimed to evaluate the efficacy and safety of the intensive BP target (< 140 mm Hg) versus the standard BP target (< 180 mm Hg) after EVT for acute ischemic stroke. METHODS We conducted a systematic review and meta-analysis synthesizing evidence from randomized controlled trials (RCTs) obtained from PubMed, Embase Cochrane, Scopus, and WOS until September 7th, 2023. We used the fixed-effect model to report dichotomous outcomes using risk ratio (RR) and continuous outcomes using mean difference (MD), with a 95% confidence interval (CI). PROSPERO ID CRD42023463206. RESULTS We included four RCTs with 1559 patients. There was no difference between intensive BP and standard BP targets regarding the National Institutes of Health Stroke Scale (NIHSS) change after 24 h [MD: 0.44 with 95% CI (0.0, 0.87), P = 0.05]. However, the intensive BP target was significantly associated with a decreased risk of excellent neurological recovery (mRS ≤ 1) [RR: 0.87 with 95% CI (0.76, 0.99), P = 0.03], functional independence (mRS ≤ 2) [RR: 0.81 with 95% CI (0.73, 0.90), P = 0.0001] and independent ambulation (mRS ≤ 3) [RR: 0.85 with 95% CI (0.79, 0.92), P < 0.0001]. CONCLUSIONS An intensive BP target after EVT is associated with worse neurological recovery and significantly decreased rates of functional independence and independent ambulation compared to the standard BP target. Therefore, the intensive BP target should be avoided after EVT for acute ischemic stroke.
Collapse
Affiliation(s)
| | - Yehya Khildj
- Faculty of Medicine, University of Algiers, Algiers, Algeria
| | | | | | | | | | - Ubaid Khan
- Faculty of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, West Virginia, USA
| | - James Robert Brašić
- Section of High-Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Behavioral Health, New York City Health and Hospitals/Bellevue, New York, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| |
Collapse
|
26
|
Qin W, Fukuie M, Hoshi D, Mori S, Tomoto T, Sugawara J, Tarumi T. Dynamic cerebral autoregulation during repeated handgrip exercise: comparisons with spontaneous rest and sit-stand maneuvers. J Appl Physiol (1985) 2024; 137:718-727. [PMID: 39116347 DOI: 10.1152/japplphysiol.00217.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Induced arterial pressure oscillation may improve the assessment of dynamic cerebral autoregulation (dCA) with transfer function analysis (TFA). This study investigated dCA during repeated handgrip exercise (RHE) compared with spontaneous rest and sit-stand maneuvers (SSM), often used in cerebrovascular research. After a 5-min rest, 20 healthy young adults (10 women and 10 men) underwent 5 min of RHE (30% maximal voluntary contraction) and SSM at 0.05 Hz and 0.10 Hz each in random order. Power spectral density (PSD) and TFA gain, phase, coherence of mean arterial pressure (MAP), and blood velocity in the middle cerebral artery (MCAvmean) were measured in very low (VLF: 0.02-0.07 Hz) and low (LF: 0.07-0.20 Hz) frequencies. End-tidal CO2 (EtCO2) was continuously recorded throughout data collection. Compared with rest, RHE increased the PSD of MAP and MCAvmean in VLF (444% and 273%, respectively) and LF (1,571% and 1,765%, respectively) (all P < 0.001). Coherence increased during RHE (VLF: 131%, LF: 128%) and SSM (VLF: 166%, LF: 136%) compared with rest (all P < 0.05). TFA gain and phase were similar between RHE and rest, but VLF gain was higher, whereas VLF and LF phases were lower during SSM than RHE (all P < 0.05). EtCO2 was higher during SSM than rest and RHE (both P < 0.05), with the individual EtCO2 changes positively correlated with VLF gain (r = 0.538, P < 0.001). These results indicate that RHE significantly increases arterial pressure oscillation and TFA coherence and may improve dCA assessment in individuals unable to perform repeated postural changes.NEW & NOTEWORTHY This is the first study investigating dynamic cerebral autoregulation (dCA) during light-intensity repeated handgrip exercise (RHE) compared with rest and sit-stand maneuvers (SSM) using transfer function analysis (TFA). Compared with rest, RHE significantly increased oscillations of arterial blood pressure and cerebral blood velocity and coherence, whereas SSM exhibited the highest oscillations and coherence. These findings suggest that RHE may serve as an alternative method for assessing dCA in individuals unable to perform repeated postural changes.
Collapse
Affiliation(s)
- Wenxing Qin
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Marina Fukuie
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Daisuke Hoshi
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Shoya Mori
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tsubasa Tomoto
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
| | - Jun Sugawara
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Takashi Tarumi
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, Texas, United States
| |
Collapse
|
27
|
Ball JD, Davies A, Gurung D, Mankoo A, Panerai R, Minhas JS, Robinson T, Beishon L. The effect of posture on the age dependence of neurovascular coupling. Physiol Rep 2024; 12:e70031. [PMID: 39218618 PMCID: PMC11366444 DOI: 10.14814/phy2.70031] [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: 01/19/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Previous studies report contradicting age-related neurovascular coupling (NVC). Few studies assess postural effects, but less investigate relationships between age and NVC within different postures. Therefore, this study investigated the effect of age on NVC in different postures with varying cognitive stimuli. Beat-to-beat blood pressure, heart rate and end-tidal carbon dioxide were assessed alongside middle and posterior cerebral artery velocities (MCAv and PCAv, respectively) using transcranial Doppler ultrasonography in 78 participants (31 young-, 23 middle- and 24 older-aged) with visuospatial (VST) and attention tasks (AT) in various postures at two timepoints (T2 and T3). Between-group significance testing utilized one-way analysis-of-variance (ANOVA) (Tukey post-hoc). Mixed three-way/one-way ANOVAs explored task, posture, and age interactions. Significant effects of posture on NVC were driven by a 3.8% increase from seated to supine. For AT, mean supine %MCAv increase was greatest in younger (5.44%) versus middle (0.12%) and older-age (0.09%) at T3 (p = 0.005). For VST, mean supine %PCAv increase was greatest at T2 and T3 in middle (10.99%/10.12%) and older-age (17.36%/17.26%) versus younger (9.44%/8.89%) (p = 0.004/p = 0.002). We identified significant age-related NVC effects with VST-induced hyperactivation. This may reflect age-related compensatory processes in supine. Further work is required, using complex stimuli while standing/walking, examining NVC, aging and falls.
Collapse
Affiliation(s)
- James D. Ball
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Aaron Davies
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Dewakar Gurung
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Alex Mankoo
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Ronney Panerai
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Jatinder S. Minhas
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Thompson Robinson
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Lucy Beishon
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| |
Collapse
|
28
|
Davies A, Gurung D, Ladthavorlaphatt K, Mankoo A, Panerai RB, Robinson TG, Minhas JS, Beishon LC. The effect of CO 2 on the age dependence of neurovascular coupling. J Appl Physiol (1985) 2024; 137:445-459. [PMID: 38961823 DOI: 10.1152/japplphysiol.00695.2023] [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: 09/29/2023] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Prior studies have identified variable effects of aging on neurovascular coupling (NVC). Carbon dioxide (CO2) affects both cerebral blood velocity (CBv) and NVC, but the effects of age on NVC under different CO2 conditions are unknown. Therefore, we investigated the effects of aging on NVC in different CO2 states during cognitive paradigms. Seventy-eight participants (18-78 yr), with well-controlled comorbidities, underwent continuous recordings of CBv by bilateral insonation of middle (MCA) and posterior (PCA) cerebral arteries (transcranial Doppler), blood pressure, end-tidal CO2, and heart rate during poikilocapnia, hypercapnia (5% CO2 inhalation), and hypocapnia (paced hyperventilation). Neuroactivation via visuospatial (VS) and attention tasks (AT) was used to stimulate NVC. Peak percentage and absolute change in MCAv/PCAv, were compared between CO2 conditions and age groups (≤30, 31-60, and >60 yr). For the VS task, in poikilocapnia, younger adults had a lower NVC response compared with older adults [mean difference (MD): -7.92% (standard deviation (SD): 2.37), P = 0.004], but comparable between younger and middle-aged groups. In hypercapnia, both younger [MD: -4.75% (SD: 1.56), P = 0.009] and middle [MD: -4.58% (SD: 1.69), P = 0.023] age groups had lower NVC responses compared with older adults. Finally, in hypocapnia, both older [MD: 5.92% (SD: 2.21), P = 0.025] and middle [MD: 5.44% (SD: 2.27), P = 0.049] age groups had greater NVC responses, compared with younger adults. In conclusion, the magnitude of NVC response suppression from baseline during hyper- and hypocapnia, did not differ significantly between age groups. However, the middle age group demonstrated a different NVC response while under hypercapnic conditions, compared with hypocapnia.NEW & NOTEWORTHY This study describes the effects of age on neurovascular coupling under altered CO2 conditions. We demonstrated that both hypercapnia and hypocapnia suppress neurovascular coupling (NVC) responses. Furthermore, that middle age exhibits an NVC response comparable with younger adults under hypercapnia, and older adults under hypocapnia.
Collapse
Affiliation(s)
- Aaron Davies
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Dewarkar Gurung
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Kannaphob Ladthavorlaphatt
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Alex Mankoo
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Ronney B Panerai
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Research Centre, British Heart Foundation Cardiovascular Centre, Leicester, United Kingdom
| | - Thompson G Robinson
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Research Centre, British Heart Foundation Cardiovascular Centre, Leicester, United Kingdom
| | - Jatinder S Minhas
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Research Centre, British Heart Foundation Cardiovascular Centre, Leicester, United Kingdom
| | - Lucy C Beishon
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM) Research Group, Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Research Centre, British Heart Foundation Cardiovascular Centre, Leicester, United Kingdom
| |
Collapse
|
29
|
Allison EY, Al-Khazraji BK. Association of Arterial Stiffness Index and Brain Structure in the UK Biobank: A 10-Year Retrospective Analysis. Aging Dis 2024; 15:1872-1884. [PMID: 37307821 PMCID: PMC11272205 DOI: 10.14336/ad.2023.0419] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 06/14/2023] Open
Abstract
Arterial stiffening and changes in brain structure both occur with normal aging and can be exacerbated via acquired health conditions. While cross-sectional associations exist, the longitudinal relationship between arterial stiffness and brain structure remains unclear. In this study, we investigated 1) associations between baseline arterial stiffness index (ASI) and brain structure (global and regional grey matter volumes (GMV), white matter hyperintensities (WMH)) 10-years post-baseline (10.4±0.8 years) and 2) associations between the 10-year change in ASI from baseline and brain structure 10-years post-baseline in 650 healthy middle- to older-aged adults (53.4±7.5 years) from the UK Biobank. We observed significant associations between baseline ASI and GMV (p<0.001) and WMH (p=0.0036) 10-years post-baseline. No significant associations between 10-year change in ASI and brain structure (global GMV p=0.24; WMH volume p=0.87) were observed. There were significant associations of baseline ASI in 2 of 60 regional brain volumes analyzed (right posterior superior temporal gyrus p=0.001; left superior lateral occipital cortex p<0.001). Strong associations with baseline ASI, but not changes in ASI over 10-years, suggest arterial stiffness at the entry point of older adulthood is more impactful on brain structure 10-years later compared to age-related stiffening. Based on these associations, we suggest clinical monitoring and potential intervention for reducing arterial stiffness should occur in midlife to reduce vascular contributions to structural changes in the brain, supporting a healthy trajectory of brain aging. Our findings also support use of ASI as a surrogate for gold standard measures in showing overall relationships between arterial stiffness and brain structure.
Collapse
Affiliation(s)
- Elric Y Allison
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Baraa K Al-Khazraji
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
30
|
Kitthanyateerakul P, Tankumpuan T, Davidson PM. Cognitive dysfunction in older patients undergoing non-neurosurgery in the immediate postoperative period: A systematic review. Nurs Open 2024; 11:e70023. [PMID: 39189543 PMCID: PMC11348231 DOI: 10.1002/nop2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/28/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
AIM To explore the risk factors associated with postoperative cognitive dysfunction in older patients within the first 7 days after non-neurosurgical surgery and anaesthesia. DESIGN A systematic review. METHODS Following, PRISMA 2020 (Preferred Reporting Items for Systematic Reviews and Meta-analyses). Checklist, a systematic review of studies published from January 2018 to January 2024. The literature search was conducted across six electronic online databases, including PubMed, EMBASE, Scopus, Ovid, MEDLINE and Science Direct, and the Johns Hopkins Nursing Evidence-Based Practice Evidence Rating Scale was used for study appraisal. RESULTS The initial search yielded 1750 studies. The review included 19 studies which comprised prospective observational, case-control and retrospective studies. The prevalence of postoperative cognitive dysfunction ranged from 19% to 64% among older adults undergoing non-neurosurgery. The identified risk factors were classified into three phases including preoperative, intraoperative and postoperative. Preoperative risk factors were found in age, educational attainment, malnutrition, preoperative biomarkers and co-morbidities. Intraoperative risk factors were the duration of the operation, blood loss during the operation and anaesthesia used. Postoperative risk factors consisted of postoperative biomarkers and postoperative pain. PATIENT OR PUBLIC CONTRIBUTION The result from this review may assist researchers and healthcare providers in assessing the underlying causes and risk factors of postoperative cognitive dysfunction, and in formulating suitable preventative and therapeutic strategies for older adults with non-neurosurgery during the short-term postoperative period.
Collapse
Affiliation(s)
| | | | - Patricia M. Davidson
- The Vice‐Chancellor's UnitUniversity of WollongongWollongongNew South WalesAustralia
| |
Collapse
|
31
|
Dang Y, Lu B, Vanderwal T, Castellanos FX, Yan CG. Early-treatment cerebral blood flow change as a predictive biomarker of antidepressant treatment response: evidence from the EMBARC clinical trial. Psychol Med 2024; 54:3053-3062. [PMID: 38720516 DOI: 10.1017/s0033291724001156] [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] [Indexed: 02/20/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is one of the most prevalent and disabling illnesses worldwide. Treatment of MDD typically relies on trial-and-error to find an effective approach. Identifying early response-related biomarkers that predict response to antidepressants would help clinicians to decide, as early as possible, whether a particular treatment might be suitable for a given patient. METHODS Data were from the two-stage Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care (EMBARC) trial. A whole-brain, voxel-wise, mixed-effects model was applied to identify early-treatment cerebral blood flow (CBF) changes as biomarkers of treatment response. We examined changes in CBF measured with arterial spin labeling 1-week after initiating double-masked sertraline/placebo. We tested whether these early 1-week scans could be used to predict response observed after 8-weeks of treatment. RESULTS Response to 8-week placebo treatment was associated with increased cerebral perfusion in temporal cortex and reduced cerebral perfusion in postcentral region captured at 1-week of treatment. Additionally, CBF response in these brain regions was significantly correlated with improvement in Hamilton Depression Rating Scale score in the placebo group. No significant associations were found for selective serotonin reuptake inhibitor treatment. CONCLUSIONS We conclude that early CBF responses to placebo administration in multiple brain regions represent candidate neural biomarkers of longer-term antidepressant effects.
Collapse
Affiliation(s)
- Yi Dang
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Bin Lu
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- International Big-Data Center for Depression Research, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Tamara Vanderwal
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Francisco Xavier Castellanos
- Department of Child and Adolescent Psychiatry, NYU Robert I. Grossman School of Medicine, New York, NY, USA
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Chao-Gan Yan
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- International Big-Data Center for Depression Research, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Kalantari S, Soltani M, Maghbooli M, Khoshe Mehr F, Kalantari Z, Borji S, Memari B, Hossein Heydari A, Elahi R, Bayat M, Salighehrad H. Alteraciones del flujo sanguíneo cerebral medidas con RM-ASL como predictor de demencia vascular en la enfermedad isquémica de pequeño vaso. RADIOLOGIA 2024. [DOI: 10.1016/j.rx.2024.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Cunha S, Bicker J, Sereno J, Falcão A, Fortuna A. Blood brain barrier dysfunction in healthy aging and dementia: Why, how, what for? Ageing Res Rev 2024; 99:102395. [PMID: 38950867 DOI: 10.1016/j.arr.2024.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/03/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
The blood brain barrier (BBB) is an indispensable structure that maintains the central nervous system (CNS) microenvironment for a correct neuronal function. It is composed by highly specialized microvessels, surrounded by astrocytes, pericytes, neurons and microglia cells, which tightly control the influx and efflux of substances to the brain parenchyma. During aging, the BBB becomes impaired, and it may contribute to the development of neurodegenerative and neurological disorders including Alzheimer's disease and other dementias. Restoring the BBB can be a strategy to prevent disease onset and development, reducing the symptoms of these conditions. This work critically reviews the major mechanisms underlying BBB breakdown in healthy and unhealthy aging, as well as biomarkers and methodologies that accurately assess its impairment. Complementarily, potential therapeutic targets are discussed as new strategies to restore the normal function of the BBB in aging.
Collapse
Affiliation(s)
- Susana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - José Sereno
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
34
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
35
|
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
|
36
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong LE, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-Related Decline in Blood-Brain Barrier Function is More Pronounced in Males than Females in Parietal and Temporal Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575463. [PMID: 38293052 PMCID: PMC10827081 DOI: 10.1101/2024.01.12.575463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8 to 92 years old, using a non-invasive diffusion prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Ophthalmology, Keck School of Medicine, University of Southern California
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell Medicine
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| |
Collapse
|
37
|
Wahl D, Clayton ZS. Peripheral vascular dysfunction and the aging brain. Aging (Albany NY) 2024; 16:9280-9302. [PMID: 38805248 PMCID: PMC11164523 DOI: 10.18632/aging.205877] [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: 01/04/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024]
Abstract
Aging is the greatest non-modifiable risk factor for most diseases, including cardiovascular diseases (CVD), which remain the leading cause of mortality worldwide. Robust evidence indicates that CVD are a strong determinant for reduced brain health and all-cause dementia with advancing age. CVD are also closely linked with peripheral and cerebral vascular dysfunction, common contributors to the development and progression of all types of dementia, that are largely driven by excessive levels of oxidative stress (e.g., reactive oxygen species [ROS]). Emerging evidence suggests that several fundamental aging mechanisms (e.g., "hallmarks" of aging), including chronic low-grade inflammation, mitochondrial dysfunction, cellular senescence and deregulated nutrient sensing contribute to excessive ROS production and are common to both peripheral and cerebral vascular dysfunction. Therefore, targeting these mechanisms to reduce ROS-related oxidative stress and improve peripheral and/or cerebral vascular function may be a promising strategy to reduce dementia risk with aging. Investigating how certain lifestyle strategies (e.g., aerobic exercise and diet modulation) and/or select pharmacological agents (natural and synthetic) intersect with aging "hallmarks" to promote peripheral and/or cerebral vascular health represent a viable option for reducing dementia risk with aging. Therefore, the primary purpose of this review is to explore mechanistic links among peripheral vascular dysfunction, cerebral vascular dysfunction, and reduced brain health with aging. Such insight and assessments of non-invasive measures of peripheral and cerebral vascular health with aging might provide a new approach for assessing dementia risk in older adults.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science and Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Zachary S. Clayton
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Geriatric Medicine, Aurora, CO 80045, USA
| |
Collapse
|
38
|
Yang L, Du H, Zhang X, Zhang D, Su X, Qiao Z, Gao B. Evaluation of the correlation between cerebral hemodynamics and blood pressure by comparative analysis of variation in cerebral blood flow in hypertensive versus normotensive individuals: A systematic review and meta-analysis. BIOMOLECULES & BIOMEDICINE 2024; 24:775-786. [PMID: 38709773 PMCID: PMC11293236 DOI: 10.17305/bb.2024.10230] [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: 01/03/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/08/2024]
Abstract
Current understanding of the cerebral vascular response to variations in blood pressure (BP) among individuals with hypertension is limited. The aim of this meta-analysis was to determine the correlation between hypertension, risk of stroke, and cerebral blood flow (CBF). We reviewed studies published between 2000 and 2023 from PubMed, Google Scholar, and Science Direct that compared mean CBF in normotensive (NTN) and hypertensive (HTN) patients. A random effects model was used to construct the risk ratio (RR), 95% confidence interval (CI), forest plot, and inverse variance weighting. Additionally, a mixed-effects meta-regression was employed to examine the impact of study-specific patient variables. This meta-analysis included eight prospective cross-sectional studies published from 2002 to 2023. It revealed a significant average difference in the standard mean CBF of -0.45 (95% CI -0.60 to -0.30, I2 = 69%, P < 0.00001), distinguishing NTN from HTN subjects. A RR of 0.90 (95% CI 0.63 to 1.30, I2 = 89%, P = 0.04) indicated a significant decrease in CBF among individuals with hypertension. We found a statistically significant relationship between changes in diastolic and systolic BPs and the mean CBF (R -0.81, P = 0.001 and R = -0.90, P = 0.005, respectively). Our research demonstrates a strong relationship between elevated BP and reduced CBF, with hypertension reducing CBF compared to NTN individuals, by increasing cerebrovascular resistance.
Collapse
Affiliation(s)
- Lei Yang
- Department of Neurosurgery, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Hong Du
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuejing Zhang
- Center of Medical Research, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Dongliang Zhang
- Department of Neurosurgery, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Xianhui Su
- Department of Neurosurgery, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Zongrong Qiao
- Department of Neurosurgery, Shijiazhuang People’s Hospital, Shijiazhuang, China
| | - Bulang Gao
- Center of Medical Research, Shijiazhuang People’s Hospital, Shijiazhuang, China
| |
Collapse
|
39
|
Coverdale NS, Champagne AA, Allen MD, Tremblay JC, Ethier TS, Fernandez-Ruiz J, Marshall RA, MacPherson REK, Pyke KE, Cook DJ, Olver TD. Brain atrophy, reduced cerebral perfusion, arterial stiffening, and wall thickening with aging coincide with stimulus-specific changes in fMRI-BOLD responses. Am J Physiol Regul Integr Comp Physiol 2024; 326:R346-R356. [PMID: 38406844 DOI: 10.1152/ajpregu.00270.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024]
Abstract
The aim of this study was to investigate how aging affects blood flow and structure of the brain. It was hypothesized older individuals would have lower gray matter volume (GMV), resting cerebral blood flow (CBF0), and depressed responses to isometabolic and neurometabolic stimuli. In addition, increased carotid-femoral pulse-wave velocity (PWV), carotid intima-media thickness (IMT), and decreased brachial flow-mediated dilation (FMD) would be associated with lower CBF0, cerebrovascular reactivity (CVR), and GMV. Brain scans (magnetic resonance imaging) and cardiovascular examinations were conducted in young (age = 24 ± 3 yr, range = 22-28 yr; n = 13) and old (age = 71 ± 4 yr; range = 67-82 yr, n = 14) participants, and CBF0, CVR [isometabolic % blood oxygen level-dependent (BOLD) in response to a breath hold (BH)], brain activation patterns during a working memory task (neurometabolic %BOLD response to N-back trial), GMV, PWV, IMT, and FMD were measured. CBF0 and to a lesser extent CVRBH were lower in the old group (P ≤ 0.050); however, the increase in the %BOLD response to the memory task was not blunted (P ≥ 0.2867). Age-related differential activation patterns during the working memory task were characterized by disinhibition of the default mode network in the old group (P < 0.0001). Linear regression analyses revealed PWV, and IMT were negatively correlated with CBF0, CVRBH, and GMV across age groups, but within the old group alone only the relationships between PWV-CVRBH and IMT-GMV remained significant (P ≤ 0.0183). These findings suggest the impacts of age on cerebral %BOLD responses are stimulus specific, brain aging involves alterations in cerebrovascular and possibly neurocognitive control, and arterial stiffening and wall thickening may serve a role in cerebrovascular aging.NEW & NOTEWORTHY Cerebral perfusion was lower in old versus young adults. %Blood oxygen level-dependent (BOLD) responses to an isometabolic stimulus and gray matter volume were decreased in old versus young adults and associated with arterial stiffening and wall thickening. The increased %BOLD response to a neurometabolic stimulus appeared unaffected by age; however, the old group displayed disinhibition of the default mode network during the stimulus. Thus, age-related alterations in cerebral %BOLD responses were stimulus specific and related to arterial remodeling.
Collapse
Affiliation(s)
- Nicole S Coverdale
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Allen A Champagne
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Matti D Allen
- Department of Physical Medicine and Rehabilitation, Queen's University, Kingston, Ontario, Canada
| | - Joshua C Tremblay
- School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Tarrah S Ethier
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - Juan Fernandez-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| | - Rory A Marshall
- Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
- Department of Biomedical Sciences, Western College of Veterinary Medicine, the University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Kyra E Pyke
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - Douglas J Cook
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, the University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
40
|
Todorov-Völgyi K, González-Gallego J, Müller SA, Beaufort N, Malik R, Schifferer M, Todorov MI, Crusius D, Robinson S, Schmidt A, Körbelin J, Bareyre F, Ertürk A, Haass C, Simons M, Paquet D, Lichtenthaler SF, Dichgans M. Proteomics of mouse brain endothelium uncovers dysregulation of vesicular transport pathways during aging. NATURE AGING 2024; 4:595-612. [PMID: 38519806 DOI: 10.1038/s43587-024-00598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Age-related decline in brain endothelial cell (BEC) function contributes critically to neurological disease. Comprehensive atlases of the BEC transcriptome have become available, but results from proteomic profiling are lacking. To gain insights into endothelial pathways affected by aging, we developed a magnetic-activated cell sorting-based mouse BEC enrichment protocol compatible with proteomics and resolved the profiles of protein abundance changes during aging. Unsupervised cluster analysis revealed a segregation of age-related protein dynamics with biological functions, including a downregulation of vesicle-mediated transport. We found a dysregulation of key regulators of endocytosis and receptor recycling (most prominently Arf6), macropinocytosis and lysosomal degradation. In gene deletion and overexpression experiments, Arf6 affected endocytosis pathways in endothelial cells. Our approach uncovered changes not picked up by transcriptomic studies, such as accumulation of vesicle cargo and receptor ligands, including Apoe. Proteomic analysis of BECs from Apoe-deficient mice revealed a signature of accelerated aging. Our findings provide a resource for analysing BEC function during aging.
Collapse
Affiliation(s)
- Katalin Todorov-Völgyi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Judit González-Gallego
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mihail Ivilinov Todorov
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Dennis Crusius
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sophie Robinson
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florence Bareyre
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Ali Ertürk
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Division of Metabolic Biochemistry, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
41
|
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
|
42
|
Romay MC, Knutsen RH, Ma F, Mompeón A, Hernandez GE, Salvador J, Mirkov S, Batra A, Sullivan DP, Procissi D, Buchanan S, Kronquist E, Ferrante EA, Muller WA, Walshon J, Steffens A, McCortney K, Horbinski C, Tournier‑Lasserve E, Sonabend AM, Sorond FA, Wang MM, Boehm M, Kozel BA, Iruela-Arispe ML. Age-related loss of Notch3 underlies brain vascular contractility deficiencies, glymphatic dysfunction, and neurodegeneration in mice. J Clin Invest 2024; 134:e166134. [PMID: 38015629 PMCID: PMC10786701 DOI: 10.1172/jci166134] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Vascular aging affects multiple organ systems, including the brain, where it can lead to vascular dementia. However, a concrete understanding of how aging specifically affects the brain vasculature, along with molecular readouts, remains vastly incomplete. Here, we demonstrate that aging is associated with a marked decline in Notch3 signaling in both murine and human brain vessels. To clarify the consequences of Notch3 loss in the brain vasculature, we used single-cell transcriptomics and found that Notch3 inactivation alters regulation of calcium and contractile function and promotes a notable increase in extracellular matrix. These alterations adversely impact vascular reactivity, manifesting as dilation, tortuosity, microaneurysms, and decreased cerebral blood flow, as observed by MRI. Combined, these vascular impairments hinder glymphatic flow and result in buildup of glycosaminoglycans within the brain parenchyma. Remarkably, this phenomenon mirrors a key pathological feature found in brains of patients with CADASIL, a hereditary vascular dementia associated with NOTCH3 missense mutations. Additionally, single-cell RNA sequencing of the neuronal compartment in aging Notch3-null mice unveiled patterns reminiscent of those observed in neurodegenerative diseases. These findings offer direct evidence that age-related NOTCH3 deficiencies trigger a progressive decline in vascular function, subsequently affecting glymphatic flow and culminating in neurodegeneration.
Collapse
Affiliation(s)
- Milagros C. Romay
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ana Mompeón
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gloria E. Hernandez
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Jocelynda Salvador
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Snezana Mirkov
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ayush Batra
- Department of Pathology
- Department of Neurology, and
| | | | - Daniele Procissi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Samuel Buchanan
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elise Kronquist
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Elisa A. Ferrante
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Laboratory of Cardiovascular Regenerative Medicine, NIH, Bethesda, Maryland, USA
| | | | - Jordain Walshon
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alicia Steffens
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elisabeth Tournier‑Lasserve
- Inserm NeuroDiderot, Université Paris Cité, Paris, France
- Service de Génétique Neurovasculaire, Assistance Publique–Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Manfred Boehm
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Laboratory of Cardiovascular Regenerative Medicine, NIH, Bethesda, Maryland, USA
| | - Beth A. Kozel
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - M. Luisa Iruela-Arispe
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
43
|
Carson Smith J, Callow DD, Pena GS, Kommula Y, Arnold-Nedimala N, Won J, Nielson KA. Exercise and Protection from Age-Related Cognitive Decline. Curr Top Behav Neurosci 2024; 67:263-280. [PMID: 39080244 DOI: 10.1007/7854_2024_501] [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: 11/06/2024]
Abstract
In this chapter, we review the cross-sectional evidence in healthy human subjects for physical activity and cardiorespiratory fitness to offer neuroprotection and moderate cognitive decline in older age. The role of exercise training on cognition in healthy older adults and those diagnosed with mild cognitive impairment (MCI) is also discussed, including the evidence from neuroimaging studies that document changes to brain structure and function after a period of exercise training and improved fitness. Finally, in reference to animal models, the potential neurophysiological mechanisms for physical activity and exercise to impact human brain health are highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | - Junyeon Won
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX, USA
| | | |
Collapse
|
44
|
Shang J, Li W, Zhang H, Wang W, Liu N, Gao D, Wang F, Yan X, Gao C, Sun R, Zhang H, Ma K, Shao F, Zhang J. C-kit controls blood-brain barrier permeability by regulating caveolae-mediated transcytosis after chronic cerebral hypoperfusion. Biomed Pharmacother 2024; 170:115778. [PMID: 38141279 DOI: 10.1016/j.biopha.2023.115778] [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: 07/12/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/25/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction plays a pivotal role in the pathology of chronic cerebral hypoperfusion (CCH)-related neurodegenerative diseases. Continuous endothelial cells (EC) that line the blood vessels of the brain are important components of the BBB to strictly control the flow of substances and maintain the homeostatic environment of the brain. However, the molecular mechanisms from the perspective of EC-induced BBB dysfunction after CCH are largely unknown. In this study, the BBB function was assessed using immunostaining and transmission electron microscopy. The EC dysfunction profile was screened by using EC enrichment followed by RNA sequencing. After identified the key EC dysfunction factor, C-kit, we used the C-kit inhibition drug (imatinib) and C-kit down-regulation method (AAV-BR1-C-kit shRNA) to verify the role of C-kit on BBB integrity and EC transcytosis after CCH. Furthermore, we also activated C-kit with stem cell factor (SCF) to observe the effects of C-kit on BBB following CCH. We explored that macromolecular proteins entered the brain mainly through EC transcytosis after CCH and caused neuronal loss. Additionally, we identified receptor tyrosine kinase C-kit as a key EC dysfunction molecule. Furthermore, the pharmacological inhibition of C-kit with imatinib counteracted BBB leakage by reducing caveolae-mediated transcytosis. Moreover, treatment with AAV-BR1-C-kit shRNA, which targets brain EC to inhibit C-kit expression, also ameliorated BBB leakage by reducing caveolae-mediated transcytosis. Furthermore, the SCF increased the permeability of the BBB by actively increasing caveolae-mediated transcytosis. This study provides evidence that C-kit is a key BBB permeability regulator through caveolae-mediated transcytosis in EC after CCH.
Collapse
Affiliation(s)
- Junkui Shang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wei Li
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Huiwen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wan Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ning Liu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Dandan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430072, China
| | - Fengyu Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Xi Yan
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Chenhao Gao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Haohan Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Kai Ma
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Fengmin Shao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China; Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China.
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| |
Collapse
|
45
|
Popov A, Brazhe N, Morozova K, Yashin K, Bychkov M, Nosova O, Sutyagina O, Brazhe A, Parshina E, Li L, Medyanik I, Korzhevskii DE, Shenkarev Z, Lyukmanova E, Verkhratsky A, Semyanov A. Mitochondrial malfunction and atrophy of astrocytes in the aged human cerebral cortex. Nat Commun 2023; 14:8380. [PMID: 38104196 PMCID: PMC10725430 DOI: 10.1038/s41467-023-44192-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
How aging affects cells of the human brain active milieu remains largely unknown. Here, we analyze astrocytes and neurons in the neocortical tissue of younger (22-50 years) and older (51-72 years) adults. Aging decreases the amount of reduced mitochondrial cytochromes in astrocytes but not neurons. The protein-to-lipid ratio decreases in astrocytes and increases in neurons. Aged astrocytes show morphological atrophy quantified by the decreased length of branches, decreased volume fraction of leaflets, and shrinkage of the anatomical domain. Atrophy correlates with the loss of gap junction coupling between astrocytes and increased input resistance. Aging is accompanied by the upregulation of glial fibrillary acidic protein (GFAP) and downregulation of membrane-cytoskeleton linker ezrin associated with leaflets. No significant changes in neuronal excitability or spontaneous inhibitory postsynaptic signaling is observed. Thus, brain aging is associated with the impaired morphological presence and mitochondrial malfunction of cortical astrocytes, but not neurons.
Collapse
Affiliation(s)
- Alexander Popov
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Nadezda Brazhe
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Kseniia Morozova
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Konstantin Yashin
- Department of Neurosurgery, Privolzhskiy Research Medical University, Nizhny, Novgorod, 603005, Russia
| | - Maxim Bychkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Olga Nosova
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia
| | - Oksana Sutyagina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Alexey Brazhe
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Evgenia Parshina
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
| | - Li Li
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China
| | - Igor Medyanik
- Department of Neurosurgery, Privolzhskiy Research Medical University, Nizhny, Novgorod, 603005, Russia
| | | | - Zakhar Shenkarev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
| | - Ekaterina Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia
- Faculty of Biology, Shenzhen MSU-BIT University, 518172, Shenzhen, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
| | - Alexey Semyanov
- College of Medicine, Jiaxing University, 314001, Jiaxing, Zhejiang Pro, China.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya street 16/10, Moscow, 117997, Russia.
- Faculty of Biology, Moscow State University, Moscow, 119234, Russia.
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|
46
|
Yetim E, Jacoby J, Damestani NL, Lovely AE, Salat DH, Juttukonda MR. Mean Arterial Pressure and Cerebral Hemodynamics Across The Lifespan: A Cross-Sectional Study From Human Connectome Project-Aging. J Magn Reson Imaging 2023; 58:1892-1900. [PMID: 37040498 PMCID: PMC10564963 DOI: 10.1002/jmri.28722] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Cerebral perfusion is directly affected by systemic blood pressure, which has been shown to be negatively correlated with cerebral blood flow (CBF). The impact of aging on these effects is not fully understood. PURPOSE To determine whether the relationship between mean arterial pressure (MAP) and cerebral hemodynamics persists throughout the lifespan. STUDY TYPE Retrospective, cross-sectional study. POPULATION Six hundred and sixty-nine participants from the Human Connectome Project-Aging ranging between 36 and 100+ years and without a major neurological disorder. FIELD STRENGTH/SEQUENCE Imaging data was acquired at 3.0 Tesla using a 32-channel head coil. CBF and arterial transit time (ATT) were measured by multi-delay pseudo-continuous arterial spin labeling. ASSESSMENT The relationships between cerebral hemodynamic parameters and MAP were evaluated globally in gray and white matter and regionally using surface-based analysis in the whole group, separately within different age groups (young: <60 years; younger-old: 60-79 years; oldest-old: ≥80 years). STATISTICAL TESTS Chi-squared, Kruskal-Wallis, ANOVA, Spearman rank correlation and linear regression models. The general linear model setup in FreeSurfer was used for surface-based analyses. P < 0.05 was considered significant. RESULTS Globally, there was a significant negative correlation between MAP and CBF in both gray (ρ = -0.275) and white matter (ρ = -0.117). This association was most prominent in the younger-old [gray matter CBF (β = -0.271); white matter CBF (β = -0.241)]. In surface-based analyses, CBF exhibited a widespread significant negative association with MAP throughout the brain, whereas a limited number of regions showed significant prolongation in ATT with higher MAP. The associations between regional CBF and MAP in the younger-old showed a different topographic pattern in comparison to young subjects. DATA CONCLUSION These observations further emphasize the importance of cardiovascular health in mid-to-late adulthood for healthy brain aging. The differences in the topographic pattern with aging indicate a spatially heterogeneous relationship between high blood pressure and CBF. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY STAGE: 3.
Collapse
Affiliation(s)
- Ezgi Yetim
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
| | - John Jacoby
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
| | - Nikou L. Damestani
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
- Department of Radiology, Harvard Medical School, Boston MA
| | - Allison E. Lovely
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
| | - David H. Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
- Department of Radiology, Harvard Medical School, Boston MA
- Neuroimaging for Veterans Center, VA Boston Healthcare System, Boston MA
| | - Meher R. Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown MA
- Department of Radiology, Harvard Medical School, Boston MA
| |
Collapse
|
47
|
Dang Y, He Y, Zheng D, Wang X, Chen J, Zhou Y. Heritability of cerebral blood flow in adolescent and young adult twins: an arterial spin labeling perfusion imaging study. Cereb Cortex 2023; 33:10624-10633. [PMID: 37615361 DOI: 10.1093/cercor/bhad310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/25/2023] Open
Abstract
Blood perfusion is a fundamental physiological property of all organs and is closely linked to brain metabolism. Genetic factors were reported to have important influences on cerebral blood flow. However, the profile of genetic contributions to cerebral blood flow in adolescents or young adults was underexplored. In this study, we recruited a sample of 65 pairs of same-sex adolescent or young adult twins undergoing resting arterial spin labeling imaging to conduct heritability analyses. Our findings indicate that genetic factors modestly affect cerebral blood flow in adolescents or young adults in the territories of left anterior cerebral artery and right posterior cerebral artery, with the primary contribution being to the frontal regions, cingulate gyrus, and striatum, suggesting a profile of genetic contributions to specific brain regions. Notably, the regions in the left hemisphere demonstrate the highest heritability in most regions examined. These results expand our knowledge of the genetic basis of cerebral blood flow in the developing brain and emphasize the importance of regional analysis in understanding the heritability of cerebral blood flow. Such insights may contribute to our understanding of the underlying genetic mechanism of brain functions and altered cerebral blood flow observed in youths with brain disorders.
Collapse
Affiliation(s)
- Yi Dang
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuwen He
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
- Center for Cognitive and Brain Sciences, University of Macau, Macao SAR 999078, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR 999078, China
| | - Dang Zheng
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
- China National Children's Center, Beijing 100035, China
| | - Xiaoming Wang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
| | - Jie Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| | - Yuan Zhou
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing 100101, China
- Department of Psychology, University of the Chinese Academy of Sciences, Beijing 100101, China
- The National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
48
|
Le Sayec M, Carregosa D, Khalifa K, de Lucia C, Aarsland D, Santos CN, Rodriguez-Mateos A. Identification and quantification of (poly)phenol and methylxanthine metabolites in human cerebrospinal fluid: evidence of their ability to cross the BBB. Food Funct 2023; 14:8893-8902. [PMID: 37701930 PMCID: PMC10544810 DOI: 10.1039/d3fo01913f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023]
Abstract
Increasing evidence suggests that dietary (poly)phenols and methylxanthines have neuroprotective effects; however, little is known about whether they can cross the blood-brain barrier (BBB) and exert direct effects on the brain. We investigated the presence of (poly)phenol and methylxanthine metabolites in plasma and cerebrospinal fluid (CSF) from 90 individuals at risk of dementia using liquid chromatography-mass spectrometry and predicted their mechanism of transport across the BBB using in silico modelling techniques. A total of 123 and 127 metabolites were detected in CSF and plasma, respectively. In silico analysis suggests that 5 of the 20 metabolites quantified in CSF can cross the BBB by passive diffusion, while at least 9 metabolites require the aid of cell transporters to cross the BBB. Our results showed that (poly)phenols and methylxanthines are bioavailable, can cross the BBB via passive diffusion or transport carriers, and can reach brain tissues to exert neuroprotective effects.
Collapse
Affiliation(s)
- Melanie Le Sayec
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| | - Diogo Carregosa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Khadija Khalifa
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Chiara de Lucia
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Cláudia N Santos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
49
|
Owashi KP, Capel C, Balédent O. Cerebral arterial flow dynamics during systole and diastole phases in young and older healthy adults. Fluids Barriers CNS 2023; 20:65. [PMID: 37705096 PMCID: PMC10500860 DOI: 10.1186/s12987-023-00467-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Since arterial flow is the leading actor in neuro-fluids flow dynamics, it might be interesting to assess whether it is meaningful to study the arterial flow waveform in more detail and whether this provides new important information. Few studies have focused on determining the influence of heart rate variation over time on the arterial flow curve. Therefore, this study aimed to evaluate cerebral arterial flow waveforms at extracranial and intracranial compartments in young and elderly healthy adults, also considering systole and diastole phases. METHODS Cine phase-contrast magnetic resonance imaging (CINE-PC MRI) was performed on twenty-eight healthy young volunteers (HYV) and twenty healthy elderly volunteers (HEV) to measure arterial blood flows at the extracranial and intracranial planes. A semi-automated protocol using MATLAB scripts was implemented to identify the main representative points in the arterial flow waveforms. Representative arterial profiles were estimated for each group. Moreover, the effects of age and sex on flow times, amplitude-related parameters, and parameters related to systole and diastole phases were evaluated at the extracranial and intracranial compartments. Student's t-test or Wilcoxon's test (depending on the normality of the distribution) was used to detect significant differences. RESULTS In HYVs, significant differences were observed between extracranial and intracranial levels in parameters related to the AP1 amplitude. Besides the detected differences in pulsatility index (extracranial: 0.92 ± 0.20 vs. 1.28 ± 0.33; intracranial: 0.79 ± 0.15 vs. 1.14 ± 0.18, p < .001) and average flow (715 ± 136 vs. 607 ± 125 ml/min, p = .008) between HYV and HEV, differences in the amplitude value of the arterial flow profile feature points were also noted. Contrary to systole duration (HYV: 360 ± 29 ms; HEV: 364 ± 47 ms), diastole duration presented higher inter-individual variability in both populations (HYV: 472 ± 145 ms; HEV: 456 ± 106 ms). Our results also showed that, with age, it is mainly the diastolic phase that changes. Although no significant differences in duration were observed between the two populations, the mean flow value in the diastolic phase was significantly lower in HEV (extracranial: 628 ± 128 vs. 457 ± 111 ml/min; intracranial: 599 ± 121 vs. 473 ± 100 ml/min, p < .001). No significant differences were observed in the arterial flow parameters evaluated between females and males in either HYV or HEV. CONCLUSION Our study provides a novel contribution on the influence of the cardiac cycle phases on cerebral arterial flow. The main contribution in this study concerns the identification of age-related alterations in cerebral blood flow, which occur mainly during the diastolic phase. Specifically, we observed that mean flow significantly decreases with age during diastole, whereas mean flow during systole is consistent.
Collapse
Affiliation(s)
| | - Cyrille Capel
- CHIMERE UR 7516, Jules Verne University of Picardy, Amiens, France
- Neurosurgery Department, Amiens Picardy University Medical Center, Amiens, France
| | - Olivier Balédent
- CHIMERE UR 7516, Jules Verne University of Picardy, Amiens, France
- Medical Image Processing Department, Amiens Picardy University Medical Center, Amiens, France
| |
Collapse
|
50
|
Lee CH, Kwak HS, Kang HS, Jung KH, Jeong SK. Geometric versus Hemodynamic Indexes for Rupture-Destined Aneurysms: A Retrospective Cohort and a Repeated-Measures Study. Cerebrovasc Dis 2023; 53:327-334. [PMID: 37696264 DOI: 10.1159/000533167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/13/2023] [Indexed: 09/13/2023] Open
Abstract
INTRODUCTION A proper stratification of intracranial aneurysms is critical in identifying rupture-destined aneurysms and unruptured intracranial aneurysms. We aimed to determine the utility of geometric and hemodynamic indexes in differentiating two types of aneurysms and to examine the characteristics of natural evolutionary changes of unruptured aneurysms. METHODS Rupture-destined aneurysm refers to an aneurysm that undergoes subsequent aneurysmal subarachnoid hemorrhage (SAH). On the other hand, an unruptured intracranial aneurysm is characterized by an aneurysm that does not experience rupture during serial time-of-flight magnetic resonance angiography (TOF-MRA). In addition to geometric indexes, signal intensity gradient (SIG), an in vivo approximated wall shear stress from TOF-MRA, was measured in aneurysms. The difference between the maximum and minimum values of SIG in an aneurysm compared to parent arterial values was designated as the delta-SIG ratio. RESULTS This study analyzed 20 rupture-destined aneurysms in 20 patients and 45 unruptured intracranial aneurysms in 41 patients with follow-up TOF-MRA. While geometric indexes did not show differences between the two groups, the delta-SIG ratio was higher in the rupture-destined aneurysms (1.5 ± 0.6 vs. 1.1 ± 0.3, p = 0.032). The delta-SIG ratio showed a higher area under the receiver operating characteristic curve for SAH than the size ratio (0.72 [95% CI, 0.58-0.87] vs. 0.56 [95% CI, 0.41-0.72], p = 0.033). The longitudinal re-examination of TOF-MRA in the unruptured intracranial aneurysms revealed evidence of aneurysmal growth, while concurrently exhibiting hemodynamic stability. CONCLUSION The delta-SIG ratio showed higher discriminatory results between the two groups compared to geometric indexes. Aneurysmal rupture risk should be assessed by considering both geometric and hemodynamic information. This study was registered on ClinicalTrials.gov (NCT05450939).
Collapse
Affiliation(s)
- Chan-Hyuk Lee
- Department of Neurology, Asan Medical Center, Seoul, Republic of Korea
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Hyo Sung Kwak
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Hyun-Seung Kang
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keun-Hwa Jung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seul-Ki Jeong
- Seul-Ki Jeong Neurology Clinic, Seoul, Republic of Korea
| |
Collapse
|