1
|
Marshall AG, Neikirk K, Shao B, Crabtree A, Vue Z, Beasley HK, Garza-Lopez E, Scudese E, Wanjalla CN, Kirabo A, Albritton CF, Jamison S, Demirci M, Murray SA, Cooper AT, Taffet GE, Hinton AO, Reddy AK. Methods to Utilize Pulse Wave Velocity to Measure Alterations in Cerebral and Cardiovascular Parameters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.22.546154. [PMID: 38798364 PMCID: PMC11118486 DOI: 10.1101/2023.06.22.546154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Alzheimer's Disease (AD) is a global health issue, affecting over 6 million in the United States, with that number expected to increase as the aging population grows. As a neurodegenerative disorder that affects memory and cognitive functions, it is well established that AD is associated with cardiovascular risk factors beyond only cerebral decline. However, the study of cerebrovascular techniques for AD is still evolving. Here, we provide reproducible methods to measure impedance-based pulse wave velocity (PWV), a marker of arterial stiffness, in the systemic vascular (aortic PWV) and in the cerebral vascular (cerebral PWV) systems. Using aortic impedance and this relatively novel technique of cerebral impedance to comprehensively describe the systemic vascular and the cerebral vascular systems, we examined the sex-dependent differences in 5x transgenic mice (5XFAD) with AD under normal and high-fat diet, and in wild-type mice under a normal diet. Additionally, we validated our method for measuring cerebrovascular impedance in a model of induced stress in 5XFAD. Together, our results show that sex and diet differences in wildtype and 5XFAD mice account for very minimal differences in cerebral impedance. Interestingly, 5XFAD, and not wildtype, male mice on a chow diet show higher cerebral impedance, suggesting pathological differences. Opposingly, when we subjected 5XFAD mice to stress, we found that females showed elevated cerebral impedance. Using this validated method of measuring impedance-based aortic and cerebral PWV, future research may explore the effects of modifying factors including age, chronic diet, and acute stress, which may mediate cardiovascular risk in AD.
Collapse
Affiliation(s)
- Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil; Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Celestine N. Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Claude F Albritton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anthonya T. Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - George E Taffet
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Antentor O. Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Anilkumar K. Reddy
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
2
|
Sugg E, Gleeson E, Baker SN, Li P, Gao C, Mueller A, Deng H, Shen S, Franco-Garcia E, Saxena R, Musiek ES, Akeju O, Xie Z, Hu K, Gao L. Sleep and circadian biomarkers of postoperative delirium (SLEEP-POD): protocol for a prospective and observational cohort study. BMJ Open 2024; 14:e080796. [PMID: 38643014 PMCID: PMC11033637 DOI: 10.1136/bmjopen-2023-080796] [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: 10/10/2023] [Accepted: 03/06/2024] [Indexed: 04/22/2024] Open
Abstract
INTRODUCTION Surgical patients over 70 experience postoperative delirium (POD) complications in up to 50% of procedures. Sleep/circadian disruption has emerged as a potential risk factor for POD in epidemiological studies. This protocol presents a single-site, prospective observational study designed to examine the relationship between sleep/circadian regulation and POD and how this association could be moderated or mediated by Alzheimer's disease (AD) pathology and genetic risk for AD. METHODS AND ANALYSIS Study staff members will screen for eligible patients (age ≥70) seeking joint replacement or spinal surgery at Massachusetts General Hospital (MGH). At the inclusion visit, patients will be asked a series of questionnaires related to sleep and cognition, conduct a four-lead ECG recording and be fitted for an actigraphy watch to wear for 7 days before surgery. Blood samples will be collected preoperatively and postoperatively and will be used to gather information about AD variant genes (APOE-ε4) and AD-related pathology (total and phosphorylated tau). Confusion Assessment Method-Scale and Montreal Cognitive Assessment will be completed twice daily for 3 days after surgery. Seven-day actigraphy assessments and Patient-Reported Outcomes Measurement Information System questionnaires will be performed 1, 3 and 12 months after surgery. Relevant patient clinical data will be monitored and recorded throughout the study. ETHICS AND DISSEMINATION This study is approved by the IRB at MGH, Boston, and it is registered with the US National Institutes of Health on ClinicalTrials.gov (NCT06052397). Plans for dissemination include conference presentations at a variety of scientific institutions. Results from this study are intended to be published in peer-reviewed journals. Relevant updates will be made available on ClinicalTrials.gov. TRIAL REGISTRATION NUMBER NCT06052397.
Collapse
Affiliation(s)
- Elizabeth Sugg
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Medical Biodynamics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Elizabeth Gleeson
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah N Baker
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Peng Li
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Medical Biodynamics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chenlu Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Medical Biodynamics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ariel Mueller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Deng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Esteban Franco-Garcia
- Department of Internal Medicine, Division of Palliative Care and Geriatric Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Erik S Musiek
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
- Center on Biological Rhythms and Sleep (COBRAS), Washington University School of Medicine, St Louis, Missouri, USA
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kun Hu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Medical Biodynamics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Medical Biodynamics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Svensson JE, Bolin M, Thor D, Williams PA, Brautaset R, Carlsson M, Sörensson P, Marlevi D, Spin-Neto R, Probst M, Hagman G, Morén AF, Kivipelto M, Plavén-Sigray P. Evaluating the effect of rapamycin treatment in Alzheimer's disease and aging using in vivo imaging: the ERAP phase IIa clinical study protocol. BMC Neurol 2024; 24:111. [PMID: 38575854 PMCID: PMC10993488 DOI: 10.1186/s12883-024-03596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Rapamycin is an inhibitor of the mechanistic target of rapamycin (mTOR) protein kinase, and preclinical data demonstrate that it is a promising candidate for a general gero- and neuroprotective treatment in humans. Results from mouse models of Alzheimer's disease have shown beneficial effects of rapamycin, including preventing or reversing cognitive deficits, reducing amyloid oligomers and tauopathies and normalizing synaptic plasticity and cerebral glucose uptake. The "Evaluating Rapamycin Treatment in Alzheimer's Disease using Positron Emission Tomography" (ERAP) trial aims to test if these results translate to humans through evaluating the change in cerebral glucose uptake following six months of rapamycin treatment in participants with early-stage Alzheimer's disease. METHODS ERAP is a six-month-long, single-arm, open-label, phase IIa biomarker-driven study evaluating if the drug rapamycin can be repurposed to treat Alzheimer's disease. Fifteen patients will be included and treated with a weekly dose of 7 mg rapamycin for six months. The primary endpoint will be change in cerebral glucose uptake, measured using [18F]FDG positron emission tomography. Secondary endpoints include changes in cognitive measures, markers in cerebrospinal fluid as well as cerebral blood flow measured using magnetic resonance imaging. As exploratory outcomes, the study will assess change in multiple age-related pathological processes, such as periodontal inflammation, retinal degeneration, bone mineral density loss, atherosclerosis and decreased cardiac function. DISCUSSION The ERAP study is a clinical trial using in vivo imaging biomarkers to assess the repurposing of rapamycin for the treatment of Alzheimer's disease. If successful, the study would provide a strong rationale for large-scale evaluation of mTOR-inhibitors as a potential disease-modifying treatment in Alzheimer's disease. TRIAL REGISTRATION ClinicalTrials.gov ID NCT06022068, date of registration 2023-08-30.
Collapse
Affiliation(s)
- Jonas E Svensson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Bolin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Daniel Thor
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rune Brautaset
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Carlsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Peder Sörensson
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - David Marlevi
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rubens Spin-Neto
- Department of Dentistry and Oral Health, Section for Oral Radiology, Aarhus University, Aarhus C, Denmark
| | - Monika Probst
- Department of Diagnostic and Interventional Neuroradiology, Klinikum Rechts Der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Göran Hagman
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Anton Forsberg Morén
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Miia Kivipelto
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Pontus Plavén-Sigray
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
4
|
Stepanichev MY, Mamedova DI, Gulyaeva NV. Hippocampus under Pressure: Molecular Mechanisms of Development of Cognitive Impairments in SHR Rats. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:711-725. [PMID: 38831507 DOI: 10.1134/s0006297924040102] [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: 11/20/2023] [Revised: 11/20/2023] [Accepted: 03/14/2024] [Indexed: 06/05/2024]
Abstract
Data from clinical trials and animal experiments demonstrate relationship between chronic hypertension and development of cognitive impairments. Here, we review structural and biochemical alterations in the hippocampus of SHR rats with genetic hypertension, which are used as a model of essential hypertension and vascular dementia. In addition to hypertension, dysfunction of the hypothalamic-pituitary-adrenal system observed in SHR rats already at an early age may be a key factor of changes in the hippocampus at the structural and molecular levels. Global changes at the body level, such as hypertension and neurohumoral dysfunction, are associated with the development of vascular pathology and impairment of the blood-brain barrier. Changes in multiple biochemical glucocorticoid-dependent processes in the hippocampus, including dysfunction of steroid hormones receptors, impairments of neurotransmitter systems, BDNF deficiency, oxidative stress, and neuroinflammation are accompanied by the structural alterations, such as cellular signs of neuroinflammation micro- and astrogliosis, impairments of neurogenesis in the subgranular neurogenic zone, and neurodegenerative processes at the level of synapses, axons, and dendrites up to the death of neurons. The consequence of this is dysfunction of hippocampus, a key structure of the limbic system necessary for cognitive functions. Taking into account the available results at various levels starting from the body and brain structure (hippocampus) levels to molecular one, we can confirm translational validity of SHR rats for modeling mechanisms of vascular dementia.
Collapse
Affiliation(s)
- Mikhail Yu Stepanichev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
| | - Diana I Mamedova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| |
Collapse
|
5
|
Yuan S, Ling Y, Huang X, Tan S, Li W, Xu A, Lyu J. Associations between the use of common nonsteroidal anti-inflammatory drugs, genetic susceptibility and dementia in participants with chronic pain: A prospective study based on 194,758 participants from the UK Biobank. J Psychiatr Res 2024; 169:152-159. [PMID: 38039689 DOI: 10.1016/j.jpsychires.2023.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/01/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE To investigate the potential relationship between common nonsteroidal anti-inflammatory drugs (NSAIDs), genetic susceptibility and all-cause dementia (ACD), Alzheimer's disease (AD), and vascular dementia (VD) among individuals experiencing chronic pain. METHODS This study was based on 194,758 chronic pain participants form UK biobank with a median follow-up of 13.7 years. Participants were categorized into different NSAIDs painkiller regimen groups: No NSAIDs group, Aspirin group, Ibuprofen group, Paracetamol group, and 2-3 NSAIDs group. Cox proportional risk models were used to examine the correlation between regular NSAIDs usage and the risk of ACD, AD, and VD. In addition, we further performed subgroup analyses and sensitivity analyses. RESULTS 1) Compared to the No NSAIDs group, the aspirin group (HR = 1.12, 95% CI:1.01-1.24, P < 0.05), the paracetamol group (HR = 1.15, 95% CI:1.05-1.27, P < 0.01), and the 2-3 NSAIDs group (HR = 1.2, 95% CI:1.08-1.33, P < 0.05) showed a higher risk of ACD. Furthermore, the 2-3 NSAIDs group was also associated with a higher risk of VD (HR = 1.39, 95% CI: 1.08-1.33, P < 0.05). 2) At high dementia GRS participants with chronic pain, the paracetamol group (HR = 1.2, 95% CI: 1.03-1.43, P < 0.05) and the NSAIDs group (HR = 1.3, 95% CI: 1.07-1.59, P < 0.05) were associated with a higher risk of ACD compared to the no painkiller group. 3) There was no significant association between ibuprofen use and higher risk of dementia. CONCLUSION In individuals with chronic pain, the use of aspirin and paracetamol was associated with a higher risk of ACD, whereas the use of ibuprofen was not significantly associated with a higher risk of ACD.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Yitong Ling
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Xiaxuan Huang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Shanyuan Tan
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Wanyue Li
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Anding Xu
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China.
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|
6
|
Sanjana F, Delgorio PL, DeConne TM, Hiscox LV, Pohlig RT, Johnson CL, Martens CR. Vascular determinants of hippocampal viscoelastic properties in healthy adults across the lifespan. J Cereb Blood Flow Metab 2023; 43:1931-1941. [PMID: 37395479 PMCID: PMC10676145 DOI: 10.1177/0271678x231186571] [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: 12/20/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
Arterial stiffness and cerebrovascular pulsatility are non-traditional risk factors of Alzheimer's disease. However, there is a gap in understanding the earliest mechanisms that link these vascular determinants to brain aging. Changes to mechanical tissue properties of the hippocampus (HC), a brain structure essential for memory encoding, may reflect the impact of vascular dysfunction on brain aging. We tested the hypothesis that arterial stiffness and cerebrovascular pulsatility are related to HC tissue properties in healthy adults across the lifespan. Twenty-five adults underwent measurements of brachial blood pressure (BP), large elastic artery stiffness, middle cerebral artery pulsatility index (MCAv PI), and magnetic resonance elastography (MRE), a sensitive measure of HC viscoelasticity. Individuals with higher carotid pulse pressure (PP) exhibited lower HC stiffness (β = -0.39, r = -0.41, p = 0.05), independent of age and sex. Collectively, carotid PP and MCAv PI significantly explained a large portion of the total variance in HC stiffness (adjusted R2 = 0.41, p = 0.005) in the absence of associations with HC volumes. These cross-sectional findings suggest that the earliest reductions in HC tissue properties are associated with alterations in vascular function.
Collapse
Affiliation(s)
- Faria Sanjana
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| | - Peyton L Delgorio
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Theodore M DeConne
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| | - Lucy V Hiscox
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
| | - Ryan T Pohlig
- Department of Epidemiology, University of Delaware, Newark, DE, USA
| | - Curtis L Johnson
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Christopher R Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| |
Collapse
|
7
|
Liu X, Halvorsen S, Blanke N, Downs M, Stein TD, Bigio IJ, Zaia J, Zhang Y. Progressive mechanical and structural changes in anterior cerebral arteries with Alzheimer's disease. Alzheimers Res Ther 2023; 15:185. [PMID: 37891618 PMCID: PMC10605786 DOI: 10.1186/s13195-023-01331-5] [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: 08/21/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the main cause for dementia. The irreversible neurodegeneration leads to a gradual loss of brain function characterized predominantly by memory loss. Cerebrovascular changes are common neuropathologic findings in aged subjects with dementia. Cerebrovascular integrity is critical for proper metabolism and perfusion of the brain, as cerebrovascular remodeling may render the brain more susceptible to pulse pressure and may be associated with poorer cognitive performance and greater risk of cerebrovascular events. The objective of this study is to provide understanding of cerebrovascular remodeling with AD progression. Anterior cerebral arteries (ACAs) from a total of 19 brain donor participants from controls and pathologically diagnosed AD groups (early-Braak stages I-II; intermediate-Braak stages III-IV; and advanced-Braak stages V-VI) were included in this study. Mechanical testing, histology, advanced optical imaging, and mass spectrometry were performed to study the progressive structural and functional changes of ACAs with AD progression. Biaxial extension-inflation tests showed that ACAs became progressively less compliant, and the longitudinal stress in the intermediate and advanced AD groups was significantly higher than that from the control group. With pathological AD development, the inner and outer diameters of the ACAs remained almost unchanged; however, histology study revealed progressive smooth muscle cell atrophy and loss of elastic fibers which led to compromised structural integrity of the arterial wall. Multiphoton imaging demonstrated elastin degradation at the media-adventitia interface, which led to the formation of an empty band of 21.0 ± 15.4 μm and 32.8 ± 9.24 μm in width for the intermediate and advanced AD groups, respectively. Furthermore, quantitative birefringence microscopy showed disorganized adventitial collagen with AD development. Mass spectrometry analysis provided further evidence of altered collagen content and other extracellular matrix (ECM) molecule and smooth muscle cell changes that were consistent with the mechanical and structural alterations. Collectively, our study provides understanding of the mechanical and structural cerebrovascular deterioration in cerebral arteries with AD, which may be related to neurodegenration and pathology in the brain.
Collapse
Affiliation(s)
- Xiaozhu Liu
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA, 02215, USA
| | - Samuel Halvorsen
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA, 02215, USA
| | - Nathan Blanke
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Margaret Downs
- Department of Biochemistry and Cell Biology, Boston University, Avedisian School of Medicine, Chobanian &, Boston, MA, USA
| | - Thor D Stein
- Pathology and Laboratory Medicine, Boston University, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA, USA
| | - Irving J Bigio
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Joseph Zaia
- Department of Biochemistry and Cell Biology, Boston University, Avedisian School of Medicine, Chobanian &, Boston, MA, USA
| | - Yanhang Zhang
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA, 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Division of Materials Science & Engineering, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
8
|
Liu X, Halvorsen S, Blanke N, Downs M, Stein TD, Bigio IJ, Zaia J, Zhang Y. Progressive Mechanical and Structural Changes in Anterior Cerebral Arteries with Alzheimer's Disease. RESEARCH SQUARE 2023:rs.3.rs-3283587. [PMID: 37693508 PMCID: PMC10491325 DOI: 10.21203/rs.3.rs-3283587/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Alzheimer disease (AD) is a neurodegenerative disease and the main cause for dementia. The irreversible neurodegeneration leads to a gradual loss of brain function characterized predominantly by memory loss. Cerebrovascular changes are common neuropathologic findings in aged subjects with dementia. Cerebrovascular integrity is critical for proper metabolism and perfusion of the brain, as cerebrovascular remodeling may render the brain more susceptible to pulse pressure and may be associated with poorer cognitive performance and greater risk of cerebrovascular events. The objective of this study is to provide understanding of cerebrovascular remodeling with AD progression. A total of 28 brain donor participants with human anterior cerebral artery (ACA) from controls and pathologically diagnosed AD groups (early - Braak stages I-II; intermediate - Braak stages III-IV; and advanced - Braak stages V-VI) were included in this study. Mechanical testing, histology, advanced optical imaging, and mass spectrometry were performed to study the progressive structural and functional changes of ACAs with AD progression. Biaxial extension-inflation tests showed that ACAs became progressively less compliant, and the longitudinal stress in the intermediate& advanced AD groups was significantly higher than that from the control group. With pathological AD development, the inner and outer diameter of ACA remained almost unchanged; however, histology study revealed progressive smooth muscle cell atrophy and loss of elastic fibers which led to compromised structural integrity of the arterial wall. Multiphoton imaging demonstrated elastin degradation at the media-adventitia interface, which led to the formation of an empty band of 21.0 ± 15.4 μm and 32.8 ± 9.24 μm in width for the intermediate& advanced AD groups, respectively. Furthermore, quantitative birefringence microscopy showed disorganized adventitial collagen with AD development. Mass spectrometry analysis provided further evidence of altered collagen content and other extracellular matrix (ECM) molecule and smooth muscle cell changes that were consistent with the mechanical and structural alterations. Collectively, our study provides understanding of the mechanical and structural cerebrovascular deterioration in cerebral arteries with AD, which may be related to neurodegenration and pathology in the brain.
Collapse
Affiliation(s)
| | | | | | - Margaret Downs
- Boston University Chobanian & Avedisian School of Medicine
| | | | | | - Joseph Zaia
- Boston University Chobanian & Avedisian School of Medicine
| | | |
Collapse
|
9
|
Kelly L, Sharp MM, Thomas I, Brown C, Schrag M, Antunes LV, Solopova E, Martinez-Gonzalez J, Rodríguez C, Carare RO. Targeting lysyl-oxidase (LOX) may facilitate intramural periarterial drainage for the treatment of Alzheimer's disease. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100171. [PMID: 37457664 PMCID: PMC10338210 DOI: 10.1016/j.cccb.2023.100171] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease is the commonest form of dementia. It is likely that a lack of clearance of amyloid beta (Aβ) results in its accumulation in the parenchyma as Aβ oligomers and insoluble plaques, and within the walls of blood vessels as cerebral amyloid angiopathy (CAA). The drainage of Aβ along the basement membranes of blood vessels as intramural periarterial drainage (IPAD), could be improved if the driving force behind IPAD could be augmented, therefore reducing Aβ accumulation. There are alterations in the composition of the vascular basement membrane in Alzheimer's disease. Lysyl oxidase (LOX) is an enzyme involved in the remodelling of the extracellular matrix and its expression and function is altered in various disease states. The expression of LOX is increased in Alzheimer's disease, but it is unclear whether this is a contributory factor in the impairment of IPAD in Alzheimer's disease. The pharmacological inhibition of LOX may be a strategy to improve IPAD and reduce the accumulation of Aβ in the parenchyma and within the walls of blood vessels.
Collapse
Affiliation(s)
- Louise Kelly
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | | | | | - Christopher Brown
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Lissa Ventura Antunes
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Elena Solopova
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - José Martinez-Gonzalez
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | | |
Collapse
|
10
|
Waigi EW, Webb RC, Moss MA, Uline MJ, McCarthy CG, Wenceslau CF. Soluble and insoluble protein aggregates, endoplasmic reticulum stress, and vascular dysfunction in Alzheimer's disease and cardiovascular diseases. GeroScience 2023; 45:1411-1438. [PMID: 36823398 PMCID: PMC10400528 DOI: 10.1007/s11357-023-00748-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/28/2023] [Indexed: 02/25/2023] Open
Abstract
Dementia refers to a particular group of symptoms characterized by difficulties with memory, language, problem-solving, and other thinking skills that affect a person's ability to perform everyday activities. Alzheimer's disease (AD) is the most common form of dementia, affecting about 6.2 million Americans aged 65 years and older. Likewise, cardiovascular diseases (CVDs) are a major cause of disability and premature death, impacting 126.9 million adults in the USA, a number that increases with age. Consequently, CVDs and cardiovascular risk factors are associated with an increased risk of AD and cognitive impairment. They share important age-related cardiometabolic and lifestyle risk factors, that make them among the leading causes of death. Additionally, there are several premises and hypotheses about the mechanisms underlying the association between AD and CVD. Although AD and CVD may be considered deleterious to health, the study of their combination constitutes a clinical challenge, and investigations to understand the mechanistic pathways for the cause-effect and/or shared pathology between these two disease constellations remains an active area of research. AD pathology is propagated by the amyloid β (Aβ) peptides. These peptides give rise to small, toxic, and soluble Aβ oligomers (SPOs) that are nonfibrillar, and it is their levels that show a robust correlation with the extent of cognitive impairment. This review will elucidate the interplay between the effects of accumulating SPOs in AD and CVDs, the resulting ER stress response, and their role in vascular dysfunction. We will also address the potential underlying mechanisms, including the possibility that SPOs are among the causes of vascular injury in CVD associated with cognitive decline. By revealing common mechanistic underpinnings of AD and CVD, we hope that novel experimental therapeutics can be designed to reduce the burden of these devastating diseases. Graphical abstract Alzheimer's disease (AD) pathology leads to the release of Aβ peptides, and their accumulation in the peripheral organs has varying effects on various components of the cardiovascular system including endoplasmic reticulum (ER) stress and vascular damage. Image created with BioRender.com.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Melissa A Moss
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Mark J Uline
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA.
| |
Collapse
|
11
|
Parada H, Bergstrom J, Bangen KJ, Reas ET. Pulse pressure trajectories predict brain microstructure in community-dwelling older adults: Associations with executive function and modification by APOE. Alzheimers Dement 2023; 19:1963-1973. [PMID: 36377803 PMCID: PMC10182213 DOI: 10.1002/alz.12844] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/16/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Effects of chronic arterial stiffness on brain aging remain unclear. We, therefore, examined whether long-term trajectories of pulse pressure (PP) predicted brain microstructure, microstructure mediated PP-executive function associations, and APOE genotype modified PP-microstructure associations. METHODS We examined associations of PP trajectories with brain microstructure measured using restriction spectrum imaging in 146 community-dwelling older adults, whether microstructure mediated PP trajectory-executive function associations, and whether PP-restriction spectrum imaging correlations were modified by APOE-ε4 status. RESULTS Participants with trajectories of high PP had lower restricted isotropic diffusion (RI) compared to those with low PP trajectories and PP-executive function associations were mediated by subcortical and white matter RI. High PP more strongly correlated with lower RI and higher hindered diffusion among APOE-ε4 carriers than non-carriers. DISCUSSION Prolonged elevated PP predicts microstructural abnormalities which may contribute to impaired executive function. APOE-ε4 carriers may be most vulnerable to the adverse effects of PP on brain microstructure.
Collapse
Affiliation(s)
- Humberto Parada
- Division of Epidemiology and Biostatistics, School of Public Health, San Diego State University, San Diego, CA, USA
- University of California, San Diego Moores Cancer Center, La Jolla, CA, USA
- Department of Radiation Medicine & Applied Science, University of California, San Diego, La Jolla, CA, USA
| | - Jaclyn Bergstrom
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Katherine J. Bangen
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Emilie T. Reas
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
12
|
Laporte JP, Faulkner ME, Gong Z, Palchamy E, Akhonda MA, Bouhrara M. Investigation of the association between central arterial stiffness and aggregate g-ratio in cognitively unimpaired adults. Front Neurol 2023; 14:1170457. [PMID: 37181577 PMCID: PMC10167487 DOI: 10.3389/fneur.2023.1170457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Stiffness of the large arteries has been shown to impact cerebral white matter (WM) microstructure in both younger and older adults. However, no study has yet demonstrated an association between arterial stiffness and aggregate g-ratio, a specific magnetic resonance imaging (MRI) measure of axonal myelination that is highly correlated with neuronal signal conduction speed. In a cohort of 38 well-documented cognitively unimpaired adults spanning a wide age range, we investigated the association between central arterial stiffness, measured using pulse wave velocity (PWV), and aggregate g-ratio, measured using our recent advanced quantitative MRI methodology, in several cerebral WM structures. After adjusting for age, sex, smoking status, and systolic blood pressure, our results indicate that higher PWV values, that is, elevated arterial stiffness, were associated with lower aggregate g-ratio values, that is, lower microstructural integrity of WM. Compared to other brain regions, these associations were stronger and highly significant in the splenium of the corpus callosum and the internal capsules, which have been consistently documented as very sensitive to elevated arterial stiffness. Moreover, our detailed analysis indicates that these associations were mainly driven by differences in myelination, measured using myelin volume fraction, rather than axonal density, measured using axonal volume fraction. Our findings suggest that arterial stiffness is associated with myelin degeneration, and encourages further longitudinal studies in larger study cohorts. Controlling arterial stiffness may represent a therapeutic target in maintaining the health of WM tissue in cerebral normative aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
13
|
Madsen LS, Parbo P, Ismail R, Gottrup H, Østergaard L, Brooks DJ, Eskildsen SF. Capillary dysfunction correlates with cortical amyloid load in early Alzheimer's disease. Neurobiol Aging 2023; 123:1-9. [PMID: 36610198 DOI: 10.1016/j.neurobiolaging.2022.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Alterations in cerebral perfusion is increasingly considered to play a crucial role in Alzheimer's disease (AD) and together with accumulated amyloid-β, deficiencies in the brain microvascular circulation may result in local hypoxia. Here, we studied alterations in cerebral circulation and the correlation between amyloid-β load and cerebral perfusion in prodromal AD (pAD). Using dynamic susceptibility contrast MRI and PET, we evaluated cerebral perfusion and amyloid-β levels in 19 individuals with mild cognitive impairment (MCI) and high amyloid-β load (pAD-MCI), 13 MCI individuals without AD pathology and 21 healthy controls. The pAD-MCI group showed significantly lower microvascular blood flow and significantly higher heterogeneity of microvascular blood transit times (p < 0.01) compared with the other 2 groups. Additionally, in the pAD-MCI group raised amyloid-β levels correlated with decreased microvascular blood flow and increased heterogeneity of microvascular blood flow in frontal and temporal areas (p < 0.01). These results indicate a close connection between levels of amyloid-β deposition and brain microvascular perfusion in pAD. A vicious cycle may be established where amyloid-β load and deficiencies in brain perfusion may reinforce each other.
Collapse
Affiliation(s)
- Lasse S Madsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine, Sygehus Lillebaelt, Kolding, Denmark
| | - Hanne Gottrup
- Dementia Clinic, Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle, UK
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Worley ML, Reed EL, Chapman CL, Kueck P, Seymour L, Fitts T, Zazulak H, Schlader ZJ, Johnson BD. Acute beetroot juice consumption does not alter cerebral autoregulation or cardiovagal baroreflex sensitivity during lower-body negative pressure in healthy adults. Front Hum Neurosci 2023; 17:1115355. [PMID: 36742355 PMCID: PMC9892911 DOI: 10.3389/fnhum.2023.1115355] [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: 12/03/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Introduction Beetroot juice (BRJ) improves peripheral endothelial function and vascular compliance, likely due to increased nitric oxide bioavailability. It is unknown if BRJ alters cerebrovascular function and cardiovagal baroreflex control in healthy individuals. Purpose We tested the hypotheses that BRJ consumption improves cerebral autoregulation (CA) and cardiovagal baroreflex sensitivity (cBRS) during lower-body negative pressure (LBNP). Methods Thirteen healthy adults (age: 26 ± 4 years; 5 women) performed oscillatory (O-LBNP) and static LBNP (S-LBNP) before (PRE) and 3 h after consuming 500 mL of BRJ (POST). Participants inhaled 3% CO2 (21% O2, 76% N2) during a 5 min baseline and throughout LBNP to attenuate reductions in end-tidal CO2 tension (PETCO2). O-LBNP was conducted at ∼0.02 Hz for six cycles (-70 mmHg), followed by a 3-min recovery before S-LBNP (-40 mmHg) for 7 min. Beat-to-beat middle cerebral artery blood velocity (MCAv) (transcranial Doppler) and blood pressure were continuously recorded. CA was assessed using transfer function analysis to calculate coherence, gain, and phase in the very-low-frequency (VLF; 0.020-0.070 Hz) and low-frequency bands (LF; 0.07-0.20 Hz). cBRS was calculated using the sequence method. Comparisons between POST vs. PRE are reported as mean ± SD. Results During O-LBNP, coherence VLF was greater at POST (0.55 ± 0.06 vs. 0.46 ± 0.08; P < 0.01), but phase VLF (P = 0.17) and gain VLF (P = 0.69) were not different. Coherence LF and phase LF were not different, but gain LF was lower at POST (1.03 ± 0.20 vs. 1.12 ± 0.30 cm/s/mmHg; P = 0.05). During S-LBNP, CA was not different in the VLF or LF bands (all P > 0.10). Up-cBRS and Down-cBRS were not different during both LBNP protocols. Conclusion These preliminary data indicate that CA and cBRS during LBNP in healthy, young adults is largely unaffected by an acute bolus of BRJ.
Collapse
Affiliation(s)
- Morgan L. Worley
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Emma L. Reed
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Human Physiology, Bowerman Sports Science Center, University of Oregon, Eugene, OR, United States
| | - Christopher L. Chapman
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Human Physiology, Bowerman Sports Science Center, University of Oregon, Eugene, OR, United States
| | - Paul Kueck
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Lauren Seymour
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Triniti Fitts
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Hannah Zazulak
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Zachary J. Schlader
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Kinesiology, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| | - Blair D. Johnson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- Department of Kinesiology, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
15
|
Liu C, Pan H, Kong F, Yang S, Shubhra QTH, Li D, Chen S. Association of arterial stiffness with all-cause and cause-specific mortality in the diabetic population: A national cohort study. Front Endocrinol (Lausanne) 2023; 14:1145914. [PMID: 36967807 PMCID: PMC10031114 DOI: 10.3389/fendo.2023.1145914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Estimated pulse wave velocity (ePWV) has been proposed as a potential alternative to carotid-femoral pulse wave velocity to assess the degree of aortic stiffness, and may predict cardiovascular disease (CVD) outcomes and mortality in the general population. However, whether arterial stiffness estimated by ePWV predicts all-cause and cause-specific mortality in patients with diabetes mellitus (DM) has not been reported. METHODS This was a prospective cohort study with data from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2014 and followed up until the end of December 2019. 5,235U.S. adults with DM (age≥20years) were included in the study. Arterial stiffness was estimated by ePWV. Survey-weighted Cox proportional hazards models were performed to assess the hazard ratios (HRs), and 95% confidence intervals (CIs) for the associations of ePWV with all-cause and cause-specific mortality. Meanwhile, the generalized additive model was used to visually assess the dose-dependent relationship between ePWV and mortality. As a complementary analysis, the relationship between mean blood pressure (MBP) and risk of mortality was also examined. Multiple imputations accounted for missing data. RESULTS For the 5,235 DM patients, the weighted mean age was 57.4 years, and 51.07% were male. During a median follow-up period of 115 months (interquartile range 81-155 months; 53,159 person-years), 1,604 all-cause deaths were recorded. In the fully adjusted Cox regression model, every 1 m/s increase in ePWV was associated with 56% (HR 1.56; 95% CI, 1.44 to 1.69) increase in the risk of all-cause. In addition, a nonlinear relationship between ePWV and all-cause mortality was observed (P for non-linear=0.033). Similar results were obtained after subgroup analysis and multiple imputations. Besides, the risk of most cause-specific mortality, except for accident and renal disease-specific mortality, increased from 53% to 102% for every 1 m/s increase in ePWV. CONCLUSIONS In the diabetic population, ePWV is independently associated with all-cause and most cause-specific mortality risks. ePWV may be a useful tool for assessing mortality risk.
Collapse
Affiliation(s)
- Cun Liu
- Department of Hypertension and Plateau Disease, Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Qinghai, China
| | - Huachun Pan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fanliang Kong
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August University, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Shumin Yang
- State Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Quazi T. H. Shubhra
- Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Translational Medicine Engineering Research Center of Guangdong Province, Foshan First People’s Hospital, Foshan, China
| | - Dandan Li
- Department of Nursing, Zaozhuang Vocational College, Shandong, China
| | - Siwei Chen
- Department of Cardiovascular Medicine, The Third Hospital of Nanchang, Jiangxi, China
- *Correspondence: Siwei Chen,
| |
Collapse
|
16
|
Carson MY, Barinas‐Mitchell E, Maki PM, Thurston RC. Childhood Maltreatment and Arterial Stiffness Among Midlife Women. J Am Heart Assoc 2022; 11:e026081. [PMID: 36314495 PMCID: PMC9673641 DOI: 10.1161/jaha.122.026081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Background Childhood maltreatment has been associated with arterial stiffness. This relationship has not been examined specifically among women at midlife, a time of increased arterial stiffness in women. This study tested whether childhood maltreatment is associated with arterial stiffness among a cohort of midlife women. Methods and Results A total of 162 nonsmoking perimenopausal and postmenopausal women free of clinical cardiovascular disease (mean age, 54 years; 72% White race, 23% Black race, and 5% Asian/Pacific Islander or Mixed race) completed the Child Trauma Questionnaire at baseline. At a follow-up visit 5 years later, blood pressure and carotid-femoral pulse wave velocity (a measure of arterial stiffness) were assessed. Relationships between childhood maltreatment and carotid-femoral pulse wave velocity were tested in linear regression models, adjusting for time between visits, age, race and ethnicity, education, body mass index, heart rate, hypertension medication, and diastolic blood pressure. Seventy-one women (44% of the sample) met criteria for a history of childhood maltreatment. Women with a history of childhood maltreatment had higher carotid-femoral pulse wave velocity (B [SE]=0.47 [0.21]; P=0.03) than women without this history, controlling for time between visits, age, race and ethnicity, education, body mass index, heart rate, hypertension medication, and diastolic blood pressure. Conclusions Among these midlife women, childhood maltreatment was associated with arterial stiffness, highlighting the potential long-term cardiovascular implications of childhood maltreatment.
Collapse
Affiliation(s)
| | - Emma Barinas‐Mitchell
- Department of EpidemiologyUniversity of Pittsburgh Graduate School of Public HealthPittsburghPA
| | - Pauline M. Maki
- Department of Psychiatry, Psychology, and Obstetrics & GynecologyUniversity of Illinois at ChicagoChicagoIL
| | - Rebecca C. Thurston
- Department of PsychologyUniversity of PittsburghPA,Department of EpidemiologyUniversity of Pittsburgh Graduate School of Public HealthPittsburghPA,Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPA
| |
Collapse
|
17
|
Deshpande A, Elliott J, Kari N, Jiang B, Michel P, Toosizadeh N, Fahadan PT, Kidwell C, Wintermark M, Laksari K. Novel imaging markers for altered cerebrovascular morphology in aging, stroke, and Alzheimer's disease. J Neuroimaging 2022; 32:956-967. [PMID: 35838658 PMCID: PMC9474631 DOI: 10.1111/jon.13023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Altered brain vasculature is a key phenomenon in several neurologic disorders. This paper presents a quantitative assessment of the anatomical variations in the Circle of Willis (CoW) and vascular morphology in healthy aging, acute ischemic stroke (AIS) and Alzheimer's Disease (AD). METHODS We used our novel automatic method to segment and extract geometric features of the cerebral vasculature from MR angiography scans of 175 healthy subjects, which were used to create a probabilistic atlas of cerebrovasculature and to study normal aging and intersubject variations in CoW anatomy. Subsequently, we quantified and analyzed vascular alterations in 45AIS and 50 AD patients, two prominent cerebrovascular and neurodegenerative disorders. RESULTS In the sampled cohort, we determined that the CoW is fully formed in only 35% of healthy adults and found significantly (p < .05) increased tortuosity and fractality, with increasing age and also with disease in both AIS and AD. We also found significantly lower vessel length, volume, and number of branches in AIS patients, as expected. The AD cerebral vessels exhibited significantly smaller diameter and more complex branching patterns, compared to age-matched healthy adults. These changes were significantly heightened (p < .05) among healthy, early onset mild AD, and moderate/severe dementia groups. CONCLUSION Although our study does not include longitudinal data due to paucity of such datasets, the specific geometric features and quantitative comparisons demonstrate the potential for using vascular morphology as a noninvasive imaging biomarker for neurologic disorders.
Collapse
Affiliation(s)
| | - Jordan Elliott
- Department of Biomedical Engineering, University of Arizona
| | - Nitya Kari
- Department of Biomedical Engineering, University of Arizona
| | - Bin Jiang
- Department of Radiology, Stanford University
| | - Patrik Michel
- Department of Neurology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nima Toosizadeh
- Department of Biomedical Engineering, University of Arizona
- Arizona Center on Aging, Department of Medicine, University of Arizona
| | - Pouya Tahsili Fahadan
- Neuroscience Intensive Care Unit, Medical Critical Care Service and Department of Medical Education, University of Virginia School of Medicine, Inova Fairfax Medical Campus
- Departments of Neurology, Johns Hopkins University School of Medicine
| | | | | | - Kaveh Laksari
- Department of Biomedical Engineering, University of Arizona
- Department of Aerospace and Mechanical Engineering, University of Arizona
| |
Collapse
|
18
|
Clayton ZS, Craighead DH, Darvish S, Coppock M, Ludwig KR, Brunt VE, Seals DR, Rossman MJ. Promoting healthy cardiovascular aging: emerging topics. THE JOURNAL OF CARDIOVASCULAR AGING 2022; 2:43. [PMID: 36337728 PMCID: PMC9632540 DOI: 10.20517/jca.2022.27] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The development of age-related cardiovascular (CV) dysfunction increases the risk of CV disease as well as other chronic age-associated disorders, including chronic kidney disease, and Alzheimer's disease and related dementias. Major manifestations of age-associated CV dysfunction that increase disease risk are vascular dysfunction, primarily vascular endothelial dysfunction and arterial stiffening, and elevated systolic blood pressure. Declines in nitric oxide bioavailability secondary to increased oxidative stress and inflammation are established mechanisms of CV dysfunction with aging. Moreover, fundamental mechanisms of aging, termed the "hallmarks of aging" extend to the CV system and, as such, may be considered "hallmarks of CV aging". These mechanisms represent viable therapeutic targets for treating CV dysfunction with aging. Healthy lifestyle behaviors, such as regular aerobic exercise and certain dietary patterns, are considered "first-line" strategies to prevent and/or treat age-associated CV dysfunction. Despite the well-established benefits of these strategies, many older adults do not meet the recommended guidelines for exercise or consume a healthy diet. Therefore, it is important to establish alternative and/or complementary evidence-based approaches to prevent or reverse age-related CV dysfunction. Targeting fundamental mechanisms of CV aging with interventions such as time-efficient exercise training, food-derived molecules, termed nutraceuticals, or select synthetic pharmacological agents represents a promising approach. In the present review, we will highlight emerging topics in the field of healthy CV aging with a specific focus on how exercise, nutrition/dietary patterns, nutraceuticals and select synthetic pharmacological compounds may promote healthy CV aging, in part, by targeting the hallmarks of CV aging.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Daniel H Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Sanna Darvish
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - McKinley Coppock
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Katelyn R Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
19
|
Iqbal K, Hasanain M, Ahmed J, Iqbal A, Rathore SS, Monis A, Baig MD, Ul Haq ZG. Association of Motoric Cognitive Risk Syndrome with Cardiovascular and Noncardiovascular Factors: A Systematic Review and Meta-Analysis. J Am Med Dir Assoc 2021; 23:810-822. [PMID: 34973959 DOI: 10.1016/j.jamda.2021.11.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/09/2021] [Accepted: 11/27/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Motoric cognitive risk syndrome (MCR) is a recently proposed predementia syndrome characterized by subjective cognitive impairment and slow gait. We aim to assess the cardiovascular and noncardiovascular factors associated with MCR. DESIGN Systematic review and meta-analysis. SETTING AND PARTICIPANTS Studies comparing patients with MCR to those without MCR, and identifying the factors associated with MCR. METHODS We used databases, including PubMed, Cochrane CENTRAL, and Embase, to identify studies evaluating the factors associated with MCR. Mean differences, odds ratios (ORs), risk ratios (RRs), and hazard ratios (HRs) with 95% CIs were calculated using Review Manager. RESULTS Meta-analysis revealed that all cardiovascular factors, including diabetes (21 studies; OR 1.50, 95% CI 1.37, 1.64), hypertension (21 studies; OR 1.20, 95% CI 1.08, 1.33), stroke (16 studies; OR 2.03, 95% CI 1.70, 2.42), heart disease (7 studies; OR 1.45, 95% CI 1.13, 1.86), coronary artery disease (5 studies; OR 1.49, 95% CI 1.16, 1.91), smoking (13 studies; OR 1.28, 95% CI 1.04, 1.58), and obesity (12 studies; OR 1.34, 95% CI 1.13, 1.59) were significantly higher in the MCR than the non-MCR group. Noncardiovascular factors, including age (22 studies; MD = 1.08, 95% CI 0.55, 1.61), education (8 studies; OR 2.04, 95% CI 1.28, 3.25), depression (17 studies; OR 2.19, 95% CI 1.65, 2.91), prior falls (9 studies; OR 1.45, 95% CI 1.17, 1.80), arthritis (6 studies; OR 1.35, 95% CI 1.07, 1.70), polypharmacy (5 studies; OR 1.65, 95% CI 1.07, 2.54), and sedentary lifestyle (11 studies; OR 2.00, 95% CI 1.59, 2.52), were significantly higher in the MCR than in the non-MCR group. Alcohol consumption (6 studies; OR 0.84, 95% CI 0.72, 0.98), however, favored the MCR over the non-MCR group. Additionally, there was no significant association of MCR with gender (22 studies; OR 1.04, 95% CI 0.94, 1.15) and cancer (3 studies; OR 2.39, 95% CI 0.69, 8.28). MCR was also significantly associated with an increased likelihood of incident dementia (5 studies; HR 2.84, 95% CI 1.77, 4.56; P < .001), incident cognitive impairment [2 studies; adjusted hazard ratio (aHR) 1.76, 95% CI 1.44, 2.15], incident falls (4 studies; RR 1.37, 95% CI 1.17, 1.60), and mortality (2 studies; aHR 1.58, 95% CI 1.35, 1.85). CONCLUSIONS AND IMPLICATIONS MCR syndrome was significantly associated with diabetes, hypertension, stroke, obesity, smoking, low education, sedentary lifestyle, and depression. Moreover, MCR significantly increased the risk of incident dementia, cognitive impairment, falls, and mortality.
Collapse
Affiliation(s)
- Kinza Iqbal
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan.
| | - Muhammad Hasanain
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Jawad Ahmed
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ayman Iqbal
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | | | | - Mirza Daniyal Baig
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
20
|
Wu L, Wang W, Tian S, Zheng H, Liu P, Wu W. Identification of Hub Genes in Patients with Alzheimer Disease and Obstructive Sleep Apnea Syndrome Using Integrated Bioinformatics Analysis. Int J Gen Med 2021; 14:9491-9502. [PMID: 34916831 PMCID: PMC8668230 DOI: 10.2147/ijgm.s341078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background Obstructive sleep apnea syndrome (OSA) is associated with an increased risk of Alzheimer’s disease (AD). This study aimed to identify the key common genes in AD and OSA and explore molecular mechanism value in AD. Methods Expression profiles GSE5281 and GSE135917 were acquired from Gene Expression Omnibus (GEO) database, respectively. Weighted gene co-expression network analysis (WGCNA) and R 4.0.2 software were used for identifying differentially expressed genes (DEGs) related to AD and OSA. Function enrichment analyses using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and the protein–protein interaction network (PPI) using the STRING database were subsequently performed on the shared DEGs. Finally, the hub genes were screened from the PPI network using the MCC algorithm of CytoHubba plugin. Results Seven modules and four modules were the most significant with AD and OSA by WGCNA, respectively. A total of 33 common genes were screened in AD and OSA by VENN. Functional enrichment analysis indicated that DEGs were mainly involved in cellular response to oxidative stress, neuroinflammation. Among these DEGs, the top 10 hub genes (high scores in cytoHubba) were selected in the PPI network, including AREG, SPP1, CXCL2, ITGAX, DUSP1, COL1A1, SCD, ACTA2, CCND2, ATF3. Conclusion This study presented ten target genes on the basis of common genes to AD and OSA. These candidate genes may provide a novel perspective to explore the underlying mechanism that OSA leads to an increased risk of AD at the transcriptome level.
Collapse
Affiliation(s)
- Lanxiang Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Wenjun Wang
- Department of Respiratory Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Sheng Tian
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Heqing Zheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Pan Liu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Wei Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| |
Collapse
|
21
|
Inserra F, Forcada P, Castellaro A, Castellaro C. Chronic Kidney Disease and Arterial Stiffness: A Two-Way Path. Front Med (Lausanne) 2021; 8:765924. [PMID: 34888327 PMCID: PMC8650118 DOI: 10.3389/fmed.2021.765924] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
The kidney-heart relationship has raised interest for the medical population since its vast and complex interaction significantly impacts health. Chronic kidney disease (CKD) generates vascular structure and function changes, with significant hemodynamic effects. The early arterial stiffening in CKD patients is a consequence of the interaction between oxidative stress and chronic vascular inflammation, leading to an accelerated deterioration of left ventricular function and alteration in tissue perfusion. CKD amplifies the inflammatory cascade's activation and is responsible for altering the endothelium function, increasing the vascular tone, wall thickening, and favors calcium deposits in the arterial wall. Simultaneously, the autonomic imbalance, and alteration in other hormonal systems, also favor the overactivation of inflammatory and fibrotic mediators. Thus, hormonal disarrangement also contributes to structural and functional lesions throughout the arterial wall. On the other hand, a rise in arterial stiffening and volume overload generates high left ventricular afterload. It increases the left ventricular burden with consequent myocardial remodeling, development of left ventricular hypertrophy and, in turn, heart failure. It is noteworthy that reduction in glomerular mass of renal diseases generates a compensatory glomerular filtration overdriven associated with large-arteries stiffness and high cardiovascular events. Furthermore, we consider that the consequent alterations of the arterial system's mechanical properties are crucial for altering tissue perfusion, mainly in low resistance. Thus, increasing the knowledge of these processes may help the reader to integrate them from a pathophysiological perspective, providing a comprehensive idea of this two-way path between arterial stiffness and renal dysfunction and their impact at the cardiovascular level.
Collapse
Affiliation(s)
- Felipe Inserra
- Advisor of Academic Vice-Rectory Department, Maimonides University, Buenos Aires, Argentina.,Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina
| | - Pedro Forcada
- Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina.,Non-Invasive Vascular Labs, CardioArenales and Diagnóstico Integral Médico (DIM) Prevención Cardiovascular, Buenos Aires, Argentina
| | - Agustina Castellaro
- Pediatric Medicine of Prof. Dr. Juan P Garrahan Hospital, Buenos Aires, Argentina
| | - Carlos Castellaro
- Master Vascular Mechanics and Arterial Hypertension, Postgraduate Department, Hypertension, Austral University, Buenos Aires, Argentina.,Department of Nephrology, Centro de Educación Médica e Investigaciones Clínicas Norberto Quirno (CEMIC) Hospital, Buenos Aires, Argentina
| |
Collapse
|
22
|
Sapkota S, McFall GP, Masellis M, Dixon RA, Black SE. Differential Cognitive Decline in Alzheimer's Disease Is Predicted by Changes in Ventricular Size but Moderated by Apolipoprotein E and Pulse Pressure. J Alzheimers Dis 2021; 85:545-560. [PMID: 34864669 DOI: 10.3233/jad-215068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Differential cognitive trajectories in Alzheimer's disease (AD) may be predicted by biomarkers from multiple domains. OBJECTIVE In a longitudinal sample of AD and AD-related dementias patients (n = 312), we tested whether 1) change in brain morphometry (ventricular enlargement) predicts differential cognitive trajectories, 2) further risk is contributed by genetic (Apolipoprotein E [APOE] ɛ4+) and vascular (pulse pressure [PP]) factors separately, and 3) the genetic + vascular risk moderates this pattern. METHODS We applied a dynamic computational approach (parallel process models) to test both concurrent and change-related associations between predictor (ventricular size) and cognition (executive function [EF]/attention). We then tested these associations as stratified by APOE (ɛ4-/ɛ4+), PP (low/high), and APOE+ PP (low/intermediate/high) risk. RESULTS First, concurrently, higher ventricular size predicted lower EF/attention performance and, longitudinally, increasing ventricular size predicted steeper EF/attention decline. Second, concurrently, higher ventricular size predicted lower EF/attention performance selectively in APOEɛ4+ carriers, and longitudinally, increasing ventricular size predicted steeper EF/attention decline selectively in the low PP group. Third, ventricular size and EF/attention associations were absent in the high APOE+ PP risk group both concurrently and longitudinally. CONCLUSION As AD progresses, a threshold effect may be present in which ventricular enlargement in the context of exacerbated APOE+ PP risk does not produce further cognitive decline.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - G Peggy McFall
- Department of Psychology (Science), University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| | - Roger A Dixon
- Department of Psychology (Science), University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Hendrickx JO, De Moudt S, Van Dam D, De Deyn PP, Fransen P, De Meyer GRY. Altered stress hormone levels affect in vivo vascular function in the hAPP23 +/- overexpressing mouse model of Alzheimer's disease. Am J Physiol Heart Circ Physiol 2021; 321:H905-H919. [PMID: 34506227 DOI: 10.1152/ajpheart.00254.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) has long been considered a brain-specific dementia syndrome. However, in recent decades, the occurrence of cardiovascular (CV) disease in the progression of AD has been confirmed by increasing epidemiological evidence. In this study, we conducted an in-depth cardiovascular characterization of a humanized amyloid precursor protein (APP) overexpressing mouse model (hAPP23+/-), which overexpresses the Swedish mutation (KM670/671NL). At the age of 6 mo, hAPP23+/- mice had a lower survival, lower body weight, and increased corticosterone and VMA levels compared with C57BL/6 littermates. Systolic blood pressure was increased in hAPP23+/- animals compared with C57BL/6 littermates, but diastolic blood pressure was not statistically different. Pulse pressure remained unchanged but abdominal and carotid pulse-wave velocity (aPWV and cPWV) were increased in hAPP23+/- compared with C57BL/6 mice. Echocardiography showed no differences in systolic or diastolic cardiac function. Ex vivo evaluation of vascular function showed decreased adreno receptor dependent vasoconstriction of hAPP23+/- aortic segments, although the isobaric biomechanics of the aortic wall were similar to C57BL/6 aortic segments. In conclusion, hAPP23+/- mice exhibited high serum corticosterone levels, elevated systolic blood pressure, and increased arterial stiffness in vivo. However, ex vivo aortic stiffness of hAPP23+/- aortic segments was not changed and vascular reactivity to α1-adrenoceptor stimulation was attenuated. These findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of AD.NEW & NOTEWORTHY We showed that male amyloid precursor protein (APP) transgenic mice have higher circulating stress hormone levels. As a result, higher systolic blood pressure and pulse-wave velocity were measured in vivo in addition to a smaller α-adrenergic receptor-dependent contraction upon ex vivo stimulation with phenylephrine. Our findings highlight the need for more frequent assessment of circulating stress hormone levels and PWV measurements in daily clinical practice for people at risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Jhana O Hendrickx
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Paul Fransen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
25
|
Bangen KJ, Smirnov DS, Delano-Wood L, Wierenga CE, Bondi MW, Salmon DP, Galasko D. Arterial stiffening acts synergistically with APOE genotype and AD biomarker status to influence memory in older adults without dementia. Alzheimers Res Ther 2021; 13:121. [PMID: 34210365 PMCID: PMC8246656 DOI: 10.1186/s13195-021-00851-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/31/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND Arterial stiffening has emerged as an important risk factor for Alzheimer's disease (AD) and related dementias. Carotid-femoral pulse wave velocity has been proposed as a non-invasive and reproducible method to assess arterial stiffness. However, the association of pulse wave velocity with performance across multiple cognitive domains as well as interactions with in vivo AD biomarkers and apolipoprotein E (APOE) genotype has received limited study. METHOD We studied 193 older adult volunteers (167 with normal cognition and 26 with mild cognitive impairment) who underwent comprehensive medical and neuropsychological evaluation at the University of California, San Diego Alzheimer's Disease Research Center. Cerebrospinal fluid (CSF) biomarkers were available on 123 participants (63%). Linear models examined whether pulse wave velocity significantly interacted with APOE ε4 status and CSF AD biomarker positivity (based on the ratio of total tau over beta-amyloid [tau/Aβ42]) on memory, language, executive functioning, attention, and visuospatial abilities. RESULTS After adjusting for demographic characteristics and vascular risk burden, across the entire sample, pulse wave velocity was associated with poorer executive functioning but not the performance in the other cognitive domains. When the modifying effects of AD genetic risk and CSF AD biomarkers were considered, pulse wave velocity interacted with APOE genotype and CSF tau/Aβ ratio such that a stronger association between elevated pulse wave velocity and poorer memory performance was found among those positive for CSF and genetic AD markers. There were no significant interaction effects for non-memory cognitive domains. CONCLUSION The findings suggest that pulse wave velocity, a non-invasive method to assess arterial wall properties, may be a useful marker of risk for cognitive decline, particularly among individuals who are APOE ε4 carriers or CSF AD biomarke0r-positive.
Collapse
Affiliation(s)
- Katherine J Bangen
- Research Service, VA San Diego Healthcare System, Building 13, 3350 La Jolla Village Drive (151A), San Diego, CA, 92161, USA.
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
| | - Denis S Smirnov
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Lisa Delano-Wood
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Christina E Wierenga
- Research Service, VA San Diego Healthcare System, Building 13, 3350 La Jolla Village Drive (151A), San Diego, CA, 92161, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - David P Salmon
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Kim HJ, Cho H, Park M, Kim JW, Ahn SJ, Lyoo CH, Suh SH, Ryu YH. MRI-Visible Perivascular Spaces in the Centrum Semiovale Are Associated with Brain Amyloid Deposition in Patients with Alzheimer Disease-Related Cognitive Impairment. AJNR Am J Neuroradiol 2021; 42:1231-1238. [PMID: 33985952 DOI: 10.3174/ajnr.a7155] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/21/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The association of perivascular spaces in the centrum semiovale with amyloid accumulation among patients with Alzheimer disease-related cognitive impairment is unknown. We evaluated this association in patients with Alzheimer disease-related cognitive impairment and β-amyloid deposition, assessed with [18F] florbetaben PET/CT. MATERIALS AND METHODS MR imaging and [18F] florbetaben PET/CT images of 144 patients with Alzheimer disease-related cognitive impairment were retrospectively evaluated. MR imaging-visible perivascular spaces were rated on a 4-point visual scale: a score of ≥3 or <3 indicated a high or low degree of MR imaging-visible perivascular spaces, respectively. Amyloid deposition was evaluated using the brain β-amyloid plaque load scoring system. RESULTS Compared with patients negative for β-amyloid, those positive for it were older and more likely to have lower cognitive function, a diagnosis of Alzheimer disease, white matter hyperintensity, the Apolipoprotein E ε4 allele, and a high degree of MR imaging-visible perivascular spaces in the centrum semiovale. Multivariable analysis, adjusted for age and Apolipoprotein E status, revealed that a high degree of MR imaging-visible perivascular spaces in the centrum semiovale was independently associated with β-amyloid positivity (odds ratio, 2.307; 95% CI, 1.036-5.136; P = .041). CONCLUSIONS A high degree of MR imaging-visible perivascular spaces in the centrum semiovale independently predicted β-amyloid positivity in patients with Alzheimer disease-related cognitive impairment. Thus, MR imaging-visible perivascular spaces in the centrum semiovale are associated with amyloid pathology of the brain and could be an indirect imaging marker of amyloid burden in patients with Alzheimer disease-related cognitive impairment.
Collapse
Affiliation(s)
- H J Kim
- From the Department of Nuclear Medicine (H.J.K., Y.H.R.)
- Department of Nuclear Medicine (H.J.K.), Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, South Korea
| | | | - M Park
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - J W Kim
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - S J Ahn
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | | | - S H Suh
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Y H Ryu
- From the Department of Nuclear Medicine (H.J.K., Y.H.R.)
| |
Collapse
|
27
|
Triantafyllou A, Ferreira JP, Kobayashi M, Micard E, Xie Y, Kearney-Schwartz A, Hossu G, Rossignol P, Bracard S, Benetos A. Longer Duration of Hypertension and MRI Microvascular Brain Alterations Are Associated with Lower Hippocampal Volumes in Older Individuals with Hypertension. J Alzheimers Dis 2021; 74:227-235. [PMID: 32039844 PMCID: PMC7175941 DOI: 10.3233/jad-190842] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hippocampal atrophy is associated with cognitive decline. Determining the clinical features associated with hippocampal volume (HV)/atrophy may help in tailoring preventive strategies. OBJECTIVE This study was aimed to investigate the association between HV (at visit 2) and vascular status (both at visit 1 and visit 2) in a cohort of individuals aged 60+ with hypertension and without overt cognitive impairment at visit 1 (visit 1 and visit 2 were separated by approximately 8 years). METHODS Hippocampal volume was estimated in brain MRIs as HV both clinically with the Scheltens' Medial Temporal Atrophy score, and automatically with the Free Surfer Software application. A detailed medical history, somatometric measurements, cognitive tests, leukoaraiosis severity (Fazekas score), vascular parameters including pulse wave velocity, central blood pressure, and carotid artery plaques, as well as several biochemical parameters were also measured. RESULTS 113 hypertensive patients, 47% male, aged 75.1±5.6 years, participated in both visit 1 and visit 2 of the ADELAHYDE study. Age (β= -0.30) and hypertension duration (β= -0.20) at visit 1 were independently associated with smaller HV at visit 2 (p < 0.05 for all). In addition to these variables, low body mass index (β= 0.18), high MRI Fazekas score (β= -0.20), and low Gröber-Buschke total recall (β= 0.27) were associated with smaller HV at visit 2 (p < 0.05 for all). CONCLUSION In a cohort of older individuals without cognitive impairment at baseline, we described several factors associated with lower HV, of which hypertension duration can potentially be modified.
Collapse
Affiliation(s)
- Areti Triantafyllou
- Department of Geriatric Medicine and Memory Clinic, CMRR Nancy-Lorraine CHU-Nancy, Nancy, France
| | - João Pedro Ferreira
- Université de Lorraine, INSERM CIC-P 1433, CHRU de Nancy, INSERM U1116, and FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France.,Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Masatake Kobayashi
- Université de Lorraine, INSERM CIC-P 1433, CHRU de Nancy, INSERM U1116, and FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Emilien Micard
- CHRU-Nancy, Inserm, Université de Lorraine, CIC, Innovation Technologique, Nancy, France
| | - Yu Xie
- Université de Lorraine, Inserm, IADI, F-54000 Nancy, France
| | - Anna Kearney-Schwartz
- Department of Geriatric Medicine and Memory Clinic, CMRR Nancy-Lorraine CHU-Nancy, Nancy, France
| | - Gabriela Hossu
- CHRU-Nancy, Inserm, Université de Lorraine, CIC, Innovation Technologique, Nancy, France.,Université de Lorraine, Inserm, IADI, F-54000 Nancy, France
| | - Patrick Rossignol
- Université de Lorraine, INSERM CIC-P 1433, CHRU de Nancy, INSERM U1116, and FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Serge Bracard
- Université de Lorraine, Inserm, IADI, F-54000 Nancy, France.,Department of Neuroradiology, CHU-Nancy, Nancy, France
| | - Athanase Benetos
- Department of Geriatric Medicine and Memory Clinic, CMRR Nancy-Lorraine CHU-Nancy, Nancy, France.,INSERM, U1116, Université de Lorraine, Vandoeuvre-les-Nancy, France
| |
Collapse
|
28
|
Pasha EP, Rutjes E, Tomoto T, Tarumi T, Stowe A, Claassen JAHR, Munro Cullum C, Zhu DC, Zhang R. Carotid Stiffness is Associated with Brain Amyloid-β Burden in Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2021; 74:925-935. [PMID: 32083583 DOI: 10.3233/jad-191073] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Vascular dysfunction has been implicated in the onset and progression of Alzheimer's disease (AD), yet the relationship of arterial stiffening with brain amyloid-β (Aβ) burden in at risk patients is unclear. OBJECTIVE We aimed to determine the relationship of aortic and carotid arterial stiffening with Aβ burden in patients with amnestic mild cognitive impairment (aMCI), a proposed transitional stage between normal aging and AD. METHODS Thirty-two older adults with aMCI underwent 18Florbetapir PET amyloid imaging to ascertain Aβ burden via standardized uptake value ratio (SUVR). Carotid-femoral pulse wave velocity (cfPWV), which reflects aortic stiffness, and carotid β stiffness index and distensibility, which reflect local cerebral arterial stiffness, thus having direct impact on the cerebral circulation, were measured using applanation tonometry and ultrasonography. RESULTS Region-of-interest based analysis showed that precuneus and mean cortex Aβ SUVR were correlated positively with carotid β stiffness index and negatively with carotid distensibility after adjusting for age, sex, mean arterial pressure (MAP), pulse pressure (PP), and APOE4 status. Whole-brain voxel-wise analysis showed that Aβ SUVR was positively correlated with carotid β stiffness index, and negatively with carotid distensibility at the precuneus/cingulate gyrus after multiple comparison correction. cfPWV was not correlated with Aβ SUVR. CONCLUSIONS Carotid rather than aortic stiffening was independently associated with brain Aβ burden in patients with aMCI after adjusting for age, sex, MAP, PP, and APOE4 status. These findings provide evidence that arterial stiffening, particularly carotid artery stiffening, may contribute to AD pathology in patients with aMCI.
Collapse
Affiliation(s)
- Evan P Pasha
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, TX, USA.,The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elmer Rutjes
- Radboud University Medical Center, Nijmegen, Netherlands
| | - Tsubasa Tomoto
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, TX, USA.,The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Takashi Tarumi
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, TX, USA.,The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann Stowe
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, TX, USA
| | | | - C Munro Cullum
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David C Zhu
- Michigan State University, East Lansing, MI, USA
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, TX, USA.,The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
29
|
Hendrickx JO, Martinet W, Van Dam D, De Meyer GRY. Inflammation, Nitro-Oxidative Stress, Impaired Autophagy, and Insulin Resistance as a Mechanistic Convergence Between Arterial Stiffness and Alzheimer's Disease. Front Mol Biosci 2021; 8:651215. [PMID: 33855048 PMCID: PMC8039307 DOI: 10.3389/fmolb.2021.651215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
The average age of the world's elderly population is steadily increasing. This unprecedented rise in the aged world population will increase the prevalence of age-related disorders such as cardiovascular disease (CVD) and neurodegeneration. In recent years, there has been an increased interest in the potential interplay between CVDs and neurodegenerative syndromes, as several vascular risk factors have been associated with Alzheimer's disease (AD). Along these lines, arterial stiffness is an independent risk factor for both CVD and AD. In this review, we discuss several inflammaging-related disease mechanisms including acute tissue-specific inflammation, nitro-oxidative stress, impaired autophagy, and insulin resistance which may contribute to the proposed synergism between arterial stiffness and AD.
Collapse
Affiliation(s)
- Jhana O. Hendrickx
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
30
|
Heidari Pahlavian S, Cen SY, Bi X, Wang DJJ, Chui HC, Yan L. Assessment of carotid stiffness by measuring carotid pulse wave velocity using a single-slice oblique-sagittal phase-contrast MRI. Magn Reson Med 2021; 86:442-455. [PMID: 33543788 DOI: 10.1002/mrm.28677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 11/06/2022]
Abstract
PURPOSE Increased arterial stiffness has been shown to be one of the earliest markers of cerebrovascular dysfunction. As a surrogate marker of arterial stiffness, pulse wave velocity (PWV) quantifications are generally carried out on central and peripheral arteries. The purpose of this study was to develop and evaluate an MRI approach to assess carotid stiffness by measuring carotid PWV (cPWV) using a fast oblique-sagittal phase-contrast MRI sequence. METHODS In 29 volunteers, a single-slice oblique-sagittal phase-contrast MRI sequence with retrospective cardiac gating was used to quantify blood velocity waveforms along a vessel segment covering the common carotid artery (CCA) and the internal carotid artery (ICA). The CCA-ICA segment length was measured from a region of interest selected on the magnitude image. Phase-contrast MRI-measured velocities were also used to quantify the ICA pulsatility index along with cPWV quantification. RESULTS The mean value of cPWV calculated using the middle upslope area algorithm was 2.86 ± 0.71 and 3.97 ± 1.14 m/s in young and elderly subjects, respectively. Oblique-sagittal phase-contrast MRI-derived cPWV measurements showed excellent intrascan and interscan repeatability. cPWV and ICA pulsatility index were significantly greater in older subjects compared to those in the young subjects (P < .01 and P = .01, respectively). Also, increased cPWV values were associated with elevated systolic blood pressure (β = 0.05, P = .03). CONCLUSION This study demonstrated that oblique-sagittal phase-contrast MRI is a feasible technique for the quantification of both cPWV and ICA pulsatility index and showed their potential utility in evaluating cerebroarterial aging and age-related neurovascular disorders.
Collapse
Affiliation(s)
- Soroush Heidari Pahlavian
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Steven Yong Cen
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Xiaoming Bi
- Siemens Medical Solutions USA, Inc., Los Angeles, California, USA
| | - Danny J J Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Helena Chang Chui
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Lirong Yan
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
31
|
Jones AM, Vanhatalo A, Seals DR, Rossman MJ, Piknova B, Jonvik KL. Dietary Nitrate and Nitric Oxide Metabolism: Mouth, Circulation, Skeletal Muscle, and Exercise Performance. Med Sci Sports Exerc 2021; 53:280-294. [PMID: 32735111 DOI: 10.1249/mss.0000000000002470] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) is a gaseous signaling molecule that plays an important role in myriad physiological processes, including the regulation of vascular tone, neurotransmission, mitochondrial respiration, and skeletal muscle contractile function. NO may be produced via the canonical NO synthase-catalyzed oxidation of l-arginine and also by the sequential reduction of nitrate to nitrite and then NO. The body's nitrate stores can be augmented by the ingestion of nitrate-rich foods (primarily green leafy vegetables). NO bioavailability is greatly enhanced by the activity of bacteria residing in the mouth, which reduce nitrate to nitrite, thereby increasing the concentration of circulating nitrite, which can be reduced further to NO in regions of low oxygen availability. Recent investigations have focused on promoting this nitrate-nitrite-NO pathway to positively affect indices of cardiovascular health and exercise tolerance. It has been reported that dietary nitrate supplementation with beetroot juice lowers blood pressure in hypertensive patients, and sodium nitrite supplementation improves vascular endothelial function and reduces the stiffening of large elastic arteries in older humans. Nitrate supplementation has also been shown to enhance skeletal muscle function and to improve exercise performance in some circumstances. Recently, it has been established that nitrate concentration in skeletal muscle is much higher than that in blood and that muscle nitrate stores are exquisitely sensitive to dietary nitrate supplementation and deprivation. In this review, we consider the possibility that nitrate represents an essential storage form of NO and discuss the integrated function of the oral microbiome, circulation, and skeletal muscle in nitrate-nitrite-NO metabolism, as well as the practical relevance for health and performance.
Collapse
Affiliation(s)
- Andrew M Jones
- Department of Sport and Health Sciences, University of Exeter, Exeter, UNITED KINGDOM
| | - Anni Vanhatalo
- Department of Sport and Health Sciences, University of Exeter, Exeter, UNITED KINGDOM
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Barbora Piknova
- Molecular Medicine Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
32
|
Badji A, Westman E. Cerebrovascular pathology in Alzheimer's disease: Hopes and gaps. Psychiatry Res Neuroimaging 2020; 306:111184. [PMID: 32950333 DOI: 10.1016/j.pscychresns.2020.111184] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/27/2020] [Accepted: 09/03/2020] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease (AD) is recognized as multifactorial and heterogeneous disease with multiple contributors to its pathophysiology, including vascular dysfunction. Given that a revision of the AT(N) classification is expected in the near future, the present work supports the importance to add an additional vascular (V) category to the framework. In particular, we attempt to shed light on the vascular markers and risk factors that are currently ready-to-be-added to the framework: i) lacunes, ii) white matter hyperintensities and iii) microbleeds seen in Flair, T2* weighted imaging and susceptibility images (SWI). Next, we discuss the added value of other types of imaging, such as diffusion-based metrics and advanced perfusion sequences to encompass more subtle vascular dysfunction. Finally, we highlight the importance to add information about the following cardiovascular risk factors to the framework: history of hypertension, obesity, and diabetes. We believe that adding a V category to the AT(N) framework will improve AD classification and foster efforts to apply the right drug(s) at the right time in the right AD subgroups. Brief communication The present work supports the importance to add an additional vascular (V) category to the AT(N) framework and shed light on the vascular MRI markers and risk factors that are currently ready-to-be-added to the framework.
Collapse
Affiliation(s)
- Atef Badji
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montréal, QC, Canada; Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Korte N, Nortley R, Attwell D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer's disease. Acta Neuropathol 2020; 140:793-810. [PMID: 32865691 PMCID: PMC7666276 DOI: 10.1007/s00401-020-02215-w] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/15/2020] [Accepted: 08/15/2020] [Indexed: 02/08/2023]
Abstract
Therapies targeting late events in Alzheimer's disease (AD), including aggregation of amyloid beta (Aβ) and hyperphosphorylated tau, have largely failed, probably because they are given after significant neuronal damage has occurred. Biomarkers suggest that the earliest event in AD is a decrease of cerebral blood flow (CBF). This is caused by constriction of capillaries by contractile pericytes, probably evoked by oligomeric Aβ. CBF is also reduced by neutrophil trapping in capillaries and clot formation, perhaps secondary to the capillary constriction. The fall in CBF potentiates neurodegeneration by upregulating the BACE1 enzyme that makes Aβ and by promoting tau hyperphosphorylation. Surprisingly, therefore, CBF reduction may play a crucial role in driving cognitive decline by initiating the amyloid cascade itself, or being caused by and amplifying Aβ production. Here, we review developments in this area that are neglected in current approaches to AD, with the aim of promoting novel mechanism-based therapeutic approaches.
Collapse
Affiliation(s)
- Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ross Nortley
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
34
|
Presa JL, Saravia F, Bagi Z, Filosa JA. Vasculo-Neuronal Coupling and Neurovascular Coupling at the Neurovascular Unit: Impact of Hypertension. Front Physiol 2020; 11:584135. [PMID: 33101063 PMCID: PMC7546852 DOI: 10.3389/fphys.2020.584135] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Components of the neurovascular unit (NVU) establish dynamic crosstalk that regulates cerebral blood flow and maintain brain homeostasis. Here, we describe accumulating evidence for cellular elements of the NVU contributing to critical physiological processes such as cerebral autoregulation, neurovascular coupling, and vasculo-neuronal coupling. We discuss how alterations in the cellular mechanisms governing NVU homeostasis can lead to pathological changes in which vascular endothelial and smooth muscle cell, pericyte and astrocyte function may play a key role. Because hypertension is a modifiable risk factor for stroke and accelerated cognitive decline in aging, we focus on hypertension-associated changes on cerebral arteriole function and structure, and the molecular mechanisms through which these may contribute to cognitive decline. We gather recent emerging evidence concerning cognitive loss in hypertension and the link with vascular dementia and Alzheimer’s disease. Collectively, we summarize how vascular dysfunction, chronic hypoperfusion, oxidative stress, and inflammatory processes can uncouple communication at the NVU impairing cerebral perfusion and contributing to neurodegeneration.
Collapse
Affiliation(s)
- Jessica L Presa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
35
|
Muhire G, Iulita MF, Vallerand D, Youwakim J, Gratuze M, Petry FR, Planel E, Ferland G, Girouard H. Arterial Stiffness Due to Carotid Calcification Disrupts Cerebral Blood Flow Regulation and Leads to Cognitive Deficits. J Am Heart Assoc 2020; 8:e011630. [PMID: 31057061 PMCID: PMC6512142 DOI: 10.1161/jaha.118.011630] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Arterial stiffness is associated with cognitive decline and dementia; however, the precise mechanisms by which it affects the brain remain unclear. Methods and Results Using a mouse model based on carotid calcification this study characterized mechanisms that could contribute to brain degeneration due to arterial stiffness. At 2 weeks postcalcification, carotid stiffness attenuated resting cerebral blood flow in several brain regions including the perirhinal/entorhinal cortex, hippocampus, and thalamus, determined by autoradiography (P<0.05). Carotid calcification impaired cerebral autoregulation and diminished cerebral blood flow responses to neuronal activity and to acetylcholine, examined by laser Doppler flowmetry (P<0.05, P<0.01). Carotid stiffness significantly affected spatial memory at 3 weeks (P<0.05), but not at 2 weeks, suggesting that cerebrovascular impairments precede cognitive dysfunction. In line with the endothelial deficits, carotid stiffness led to increased blood‐brain barrier permeability in the hippocampus (P<0.01). This region also exhibited reductions in vessel number containing collagen IV (P<0.01), as did the somatosensory cortex (P<0.05). No evidence of cerebral microhemorrhages was present. Carotid stiffness did not affect the production of mouse amyloid‐β (Aβ) or tau phosphorylation, although it led to a modest increase in the Aβ40/Aβ42 ratio in frontal cortex (P<0.01). Conclusions These findings suggest that carotid stiffness alters brain microcirculation and increases blood‐brain barrier permeability associated with cognitive impairments. Therefore, arterial stiffness should be considered a relevant target to protect the brain and prevent cognitive dysfunctions.
Collapse
Affiliation(s)
- Gervais Muhire
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - M Florencia Iulita
- 2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,3 Département de Neurosciences Université de Montréal Québec Canada
| | - Diane Vallerand
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Jessica Youwakim
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Maud Gratuze
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Franck R Petry
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Emmanuel Planel
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada.,5 Centre de Recherche du CHU de Québec Québec Canada
| | - Guylaine Ferland
- 6 Département de Nutrition Université de Montréal Québec Canada.,7 Centre de Recherche de l'Institut de Cardiologie de Montréal Montréal Québec Canada
| | - Hélène Girouard
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada.,2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,8 Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal Montréal Québec Canada
| |
Collapse
|
36
|
Targeting mitochondrial fitness as a strategy for healthy vascular aging. Clin Sci (Lond) 2020; 134:1491-1519. [PMID: 32584404 DOI: 10.1042/cs20190559] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide and aging is the primary risk factor for CVD. The development of vascular dysfunction, including endothelial dysfunction and stiffening of the large elastic arteries (i.e., the aorta and carotid arteries), contribute importantly to the age-related increase in CVD risk. Vascular aging is driven in large part by oxidative stress, which reduces bioavailability of nitric oxide and promotes alterations in the extracellular matrix. A key upstream driver of vascular oxidative stress is age-associated mitochondrial dysfunction. This review will focus on vascular mitochondria, mitochondrial dysregulation and mitochondrial reactive oxygen species (ROS) production and discuss current evidence for prevention and treatment of vascular aging via lifestyle and pharmacological strategies that improve mitochondrial health. We will also identify promising areas and important considerations ('research gaps') for future investigation.
Collapse
|
37
|
Rosa G, Giannotti C, Martella L, Massa F, Serafini G, Pardini M, Nobili FM, Monacelli F. Brain Aging, Cardiovascular Diseases, Mixed Dementia, and Frailty in the Oldest Old: From Brain Phenotype to Clinical Expression. J Alzheimers Dis 2020; 75:1083-1103. [DOI: 10.3233/jad-191075] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Gianmarco Rosa
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Cardiovascular Diseases, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Giannotti
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Martella
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Flavio Mariano Nobili
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
38
|
Large-Artery Stiffness in Health and Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 74:1237-1263. [PMID: 31466622 DOI: 10.1016/j.jacc.2019.07.012] [Citation(s) in RCA: 511] [Impact Index Per Article: 127.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/28/2019] [Accepted: 07/08/2019] [Indexed: 12/27/2022]
Abstract
A healthy aorta exerts a powerful cushioning function, which limits arterial pulsatility and protects the microvasculature from potentially harmful fluctuations in pressure and blood flow. Large-artery (aortic) stiffening, which occurs with aging and various pathologic states, impairs this cushioning function, and has important consequences on cardiovascular health, including isolated systolic hypertension, excessive penetration of pulsatile energy into the microvasculature of target organs that operate at low vascular resistance, and abnormal ventricular-arterial interactions that promote left ventricular remodeling, dysfunction, and failure. Large-artery stiffness independently predicts cardiovascular risk and represents a high-priority therapeutic target to ameliorate the global burden of cardiovascular disease. This paper provides an overview of key physiologic and biophysical principles related to arterial stiffness, the impact of aortic stiffening on target organs, noninvasive methods for the measurement of arterial stiffness, mechanisms leading to aortic stiffening, therapeutic approaches to reduce it, and clinical applications of arterial stiffness measurements.
Collapse
|
39
|
Abstract
Hypertension, particularly midlife high blood pressure, has been related to a higher risk of cognitive decline and dementia, including Alzheimer disease. However, these associations are complex and not fully elucidated. Cerebral small vessel disease emerges as one of the most important causes. Several observational studies have shown the potential beneficial role of antihypertensive treatment in preventing cognitive decline. However, randomized clinical trials (RCTs) have shown controversial results without proving nor disproving the association. On the other hand, in very elderly or frail people some studies have observed a relationship between low blood pressure and worse cognitive function. The optimal systolic and diastolic blood pressure values for protecting cognitive function, especially in elderly people, are not known.
Collapse
Affiliation(s)
- Cristina Sierra
- Hypertension & Vascular Risk Unit, Internal Medicine Department, Hospital Clinic of Barcelona (IDIBAPS), University of Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Gepner AD, Lazar K, Hulle CV, Korcarz CE, Asthana S, Carlsson CM. Effects of Simvastatin on Augmentation Index Are Transient: Outcomes From a Randomized Controlled Trial. J Am Heart Assoc 2019; 8:e009792. [PMID: 31607205 PMCID: PMC6818042 DOI: 10.1161/jaha.118.009792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Statins improve endothelial function, but their effects on arterial stiffness and aortic blood pressure in middle‐aged adults are uncertain. Methods and Results This was a prospective, randomized, double‐blind, placebo‐controlled trial of middle‐aged (40‐72 years old) adults who were randomly assigned to receive simvastatin 40 mg (n=44) or placebo (n=44) daily for 18 months to evaluate impact on dementia‐related biomarkers (primary end points) and measures of vascular health (secondary end points). This analysis focuses on the predetermined secondary end points of changes in central aortic blood pressure, aortic augmentation index, and brachial artery flow‐mediated dilation. Measurements were performed at baseline and after 6, 12, and 18 months. Multivariable models were used to identify predictors of these prespecified vascular end points. Study groups were similar at baseline; low‐density lipoprotein cholesterol declined in the statin group but not in the placebo group (P<0.01). There were no significant differences in changes in central blood pressure parameters or flow‐mediated dilation (all P>0.2). After 12 months, augmentation index decreased from baseline in the statin group compared with the placebo group (−2.3% [5.5%] versus 1.2% [5.7%], P=0.007), but by 18 months the response in both groups trend toward baseline (−1.1% [5.8%] versus 0.2% [4.8%], P=0.3). Low‐density lipoprotein cholesterol was not associated with changes in augmentation index at any time point. Conclusions Statin therapy led to a short‐term reduction in augmentation index after 12 months, but this effect did not persist after 18 months despite continued reduction in low‐density lipoprotein cholesterol levels. These findings suggest that statins may have a transient effect on aortic stiffness. Clinical Trial Registration URL: https://www.clinicaltrials.gov/. Unique identifier: NCT00939822.
Collapse
Affiliation(s)
- Adam D. Gepner
- William S. Middleton Memorial Veterans HospitalMadisonWI
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| | - Karen Lazar
- William S. Middleton Memorial Veterans HospitalMadisonWI
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| | - Carol Van Hulle
- William S. Middleton Memorial Veterans HospitalMadisonWI
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| | - Claudia E. Korcarz
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| | - Sanjay Asthana
- William S. Middleton Memorial Veterans HospitalMadisonWI
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| | - Cynthia M. Carlsson
- William S. Middleton Memorial Veterans HospitalMadisonWI
- Department of MedicineSchool of Medicine and Public HealthUniversity of WisconsinMadisonWI
| |
Collapse
|
41
|
Cui C, Sekikawa A, Kuller LH, Lopez OL, Newman AB, Kuipers AL, Mackey RH. Aortic Stiffness is Associated with Increased Risk of Incident Dementia in Older Adults. J Alzheimers Dis 2019; 66:297-306. [PMID: 30282361 DOI: 10.3233/jad-180449] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease risk factors, including age, hypertension, and diabetes, contribute to aortic stiffness and subclinical cardiovascular and brain disease, increasing dementia risk. Aortic stiffness, measured by carotid-femoral pulse wave velocity (cfPWV), reduces the buffering of pulsatile blood flow, exposing cerebral small arteries to microvascular damage. High cfPWV is related to white matter hyperintensities and brain amyloid deposition, and to cognitive decline, but it is unclear whether cfPWV independently predicts incident dementia. Therefore, we tested the hypothesis that cfPWV predicts incident dementia in older adults, independent of potential confounders. The Cardiovascular Health Study Cognition Study followed 532 non-demented older adults with annual cognitive exams from 1998-99 through 2013. CfPWV was measured on 356 (mean age = 78, 59% women) between 1996-2000. Over 15 years, 212 (59.6%) developed dementia (median time from cfPWV measurement = 4 years). In age and sex-adjusted Cox models, cfPWV was significantly associated with increased risk of dementia, but systolic blood pressure, mean arterial pressure and pulse pressure were not. CfPWV (transformed as - 1/cfPWV) remained significantly associated with dementia risk when further adjusted for education, race, APOEɛ4, diabetes, body mass index, mean arterial pressure, and anti-hypertensive medication (hazard ratio = 1.60, 95% CI = 1.02, 2.51). Results were similar when further adjusted for baseline global cognition, subclinical brain measures, and coronary artery calcification. Finally, higher cfPWV was related to lower physical activity intensity and higher systolic blood pressure, heart rate, and waist circumference measured 5 years prior. An important unanswered question is whether interventions to slow arterial stiffening can reduce the risk of dementia.
Collapse
Affiliation(s)
- Chendi Cui
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Akira Sekikawa
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Allison L Kuipers
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Rachel H Mackey
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
42
|
Brothers RM, Fadel PJ, Keller DM. Racial disparities in cardiovascular disease risk: mechanisms of vascular dysfunction. Am J Physiol Heart Circ Physiol 2019; 317:H777-H789. [PMID: 31397168 DOI: 10.1152/ajpheart.00126.2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) accounts for a third of all deaths in the United States making it the leading cause of morbidity and mortality. Although CVD affects individuals of all races/ethnicities, the prevalence of CVD is highest in non-Hispanic black (BL) individuals relative to other populations. The mechanism(s) responsible for elevated CVD risk in the BL population remains incompletely understood. However, impaired vascular vasodilator capacity and exaggerated vascular vasoconstrictor responsiveness are likely contributing factors, both of which are present even in young, otherwise healthy BL individuals. Within this review, we highlight some historical and recent data, collected from our laboratories, of impaired vascular function, in terms of reduced vasodilator capacity and heightened vasoconstrictor responsiveness, in the peripheral and cerebral circulations in BL individuals. We provide data that such impairments may be related to elevated oxidative stress and subsequent reduction in nitric oxide bioavailability. In addition, divergent mechanisms of impaired vasodilatory capacity between BL men and women are discussed. Finally, we propose several directions where future research is needed to fill in knowledge gaps, which will allow for better understanding of the mechanisms contributing to impaired vascular function in this population. Ultimately, this information will allow for better lifestyle and therapeutic approaches to be implemented in an effort to minimize the increased CVD burden in the BL population.
Collapse
Affiliation(s)
- R Matthew Brothers
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - David M Keller
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
43
|
Russell-Buckland J, Barnes CP, Tachtsidis I. A Bayesian framework for the analysis of systems biology models of the brain. PLoS Comput Biol 2019; 15:e1006631. [PMID: 31026277 PMCID: PMC6505968 DOI: 10.1371/journal.pcbi.1006631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/08/2019] [Accepted: 02/23/2019] [Indexed: 01/11/2023] Open
Abstract
Systems biology models are used to understand complex biological and physiological systems. Interpretation of these models is an important part of developing this understanding. These models are often fit to experimental data in order to understand how the system has produced various phenomena or behaviour that are seen in the data. In this paper, we have outlined a framework that can be used to perform Bayesian analysis of complex systems biology models. In particular, we have focussed on analysing a systems biology of the brain using both simulated and measured data. By using a combination of sensitivity analysis and approximate Bayesian computation, we have shown that it is possible to obtain distributions of parameters that can better guard against misinterpretation of results, as compared to a maximum likelihood estimate based approach. This is done through analysis of simulated and experimental data. NIRS measurements were simulated using the same simulated systemic input data for the model in a ‘healthy’ and ‘impaired’ state. By analysing both of these datasets, we show that different parameter spaces can be distinguished and compared between different physiological states or conditions. Finally, we analyse experimental data using the new Bayesian framework and the previous maximum likelihood estimate approach, showing that the Bayesian approach provides a more complete understanding of the parameter space. Systems biology models are mathematical representations of biological processes that reproduce the overall behaviour of a biological system. They are comprised by a number of parameters representing biological information. We use them to understand the behaviour of biological systems, such as the brain. We do this by fitting the model’s parameter to observed or simulated data; and by looking at how these values change during the fitting process we investigate the behaviour of our system. We are interested in understanding differences between a healthy and an injured brain. Here we outline a statistical framework that uses a Bayesian approach during the fitting process that can provide us with a distribution of parameters rather than single parameter number. We apply this method when simulating the physiological responses between a healthy and a vascular compromised brain to a drop in oxygenation. We then use experimental data that demonstrates the healthy brain response to an increase in arterial CO2 and fit our brain model predictions to the measurements. In both instances we show that our approach provides more information about the overlap between healthy and unhealthy brain states than a fitting process that provides a single value parameter estimate.
Collapse
Affiliation(s)
- Joshua Russell-Buckland
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, United Kingdom
- * E-mail:
| | - Christopher P. Barnes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Ilias Tachtsidis
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| |
Collapse
|
44
|
Pasha EP, Birdsill AC, Oleson S, Haley AP, Tanaka H. Physical activity mitigates adverse effect of metabolic syndrome on vessels and brain. Brain Imaging Behav 2019; 12:1658-1668. [PMID: 29374355 DOI: 10.1007/s11682-018-9830-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Metabolic syndrome (MetS) adversely affects the vasculature and cerebral white matter (CWM) integrity. Arterial stiffening has been associated with diminished CWM integrity. Physical activity (PA) can ameliorate components of MetS and subsequently affect arterial stiffening and CWM integrity. Our aim was to determine the role of PA on mitigating the adverse influence of MetS on arterial stiffness and CWM integrity. In a cross-sectional study design, sixty-six middle-aged adults (40-62 years) composed of 18 sedentary MetS (Sed MetS), 21 physically active MetS (Active MetS), and 27 healthy individuals absent of MetS risk factors were studied. Carotid artery stiffness was assessed via simultaneous ultrasound and tonometry. CWM integrity was measured using diffusion tensor imaging (DTI) through metrics of fractional anisotropy (FA) and mean diffusivity (MD). Carotid β-stiffness index in Active MetS was lower than Sed MetS but was not different from Healthy controls (6.6 ± 1.5, 7.7 ± 2.1, and 5.6 ± 1.6 au, p = 0.001). CWM integrity was significantly greater in Active MetS subjects compared to Sed MetS subjects but statistically equal to Healthy controls in the anterior limb of the internal capsule, and splenium of the corpus callosum, uncinate fasciculus, and superior corona radiata (all p < 0.05). Middle-aged individuals with MetS who habitually perform PA demonstrated lower arterial stiffness and more favorable CWM integrity than their sedentary peers, indicating that PA may be effective in mitigating the adverse effects of MetS on the vasculature and brain at midlife.
Collapse
Affiliation(s)
- Evan P Pasha
- Cardiovascular Aging Research Laboratory, Department of Kinesiology and Health Education, The University of Texas at Austin, 2109 San Jacinto Blvd, D3700, Austin, TX, 78712, USA.
| | - Alex C Birdsill
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Stephanie Oleson
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andreana P Haley
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
- Imaging Research Center, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hirofumi Tanaka
- Cardiovascular Aging Research Laboratory, Department of Kinesiology and Health Education, The University of Texas at Austin, 2109 San Jacinto Blvd, D3700, Austin, TX, 78712, USA
| |
Collapse
|
45
|
Hypertension Assessment Using Photoplethysmography: A Risk Stratification Approach. J Clin Med 2018; 8:jcm8010012. [PMID: 30577637 PMCID: PMC6352119 DOI: 10.3390/jcm8010012] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/29/2018] [Accepted: 12/14/2018] [Indexed: 12/01/2022] Open
Abstract
Hypertension is a common chronic cardiovascular disease (CVD). Early screening and diagnosis of hypertension plays a major role in its prevention and in the control of CVDs. Our study discusses the early screening of hypertension while using the morphological features of photoplethysmography (PPG). Numerous morphological features of PPG and its derivative waves were defined and extracted. Six types of feature selection methods were chosen to screen and evaluate these PPG morphological features. The optimal features were comprehensively analyzed in relation to the physiological processes of the cardiovascular circulatory system. Particularly, the intrinsic relation and physiological significance between the formation process of systolic blood pressure (SBP) and PPG morphology features were analyzed in depth. A variety of linear and nonlinear classification models were established for the comparison trials. The F1 scores for the normotension versus prehypertension, normotension and prehypertension versus hypertension, and normotension versus hypertension trials were 72.97%, 81.82%, and 92.31%, respectively. In summary, this study established a PPG characteristic analysis model and established the intrinsic relationship between SBP and PPG characteristics. Finally, the risk stratification of hypertension at different stages was examined and compared based on the optimal feature subset.
Collapse
|
46
|
Rossman MJ, LaRocca TJ, Martens CR, Seals DR. Healthy lifestyle-based approaches for successful vascular aging. J Appl Physiol (1985) 2018; 125:1888-1900. [PMID: 30212305 PMCID: PMC6842891 DOI: 10.1152/japplphysiol.00521.2018] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 12/19/2022] Open
Abstract
This review summarizes a presentation given at the 2016 Gerontological Society of America Annual Meeting as part of the Vascular Aging Workshop. The development of age-related vascular dysfunction increases the risk of cardiovascular disease as well as other chronic age-associated disorders, including chronic kidney disease and Alzheimer's disease. Healthy lifestyle behaviors, most notably regular aerobic exercise and certain dietary patterns, are considered "first-line" strategies for the prevention and/or treatment of vascular dysfunction with aging. Despite the well-established benefits of these strategies, however, many older adults do not meet the recommended guidelines for exercise or consume a healthy diet. Therefore, it is important to establish alternative and/or complementary evidence-based approaches to prevent or reverse age-related vascular dysfunction. Time-efficient forms of exercise training, hormetic exposure to mild environmental stress, fasting "mimicking" dietary paradigms, and nutraceutical/pharmaceutical approaches to favorably modulate cellular and molecular pathways activated by exercise and healthy dietary patterns may hold promise as such alternative approaches. Determining the efficacy of these novel strategies is important to provide alternatives for adults with low adherence to conventional healthy lifestyle practices for healthy vascular aging.
Collapse
Affiliation(s)
- Matthew J Rossman
- Department of Integrative Physiology, University of Colorado-Boulder , Boulder, Colorado
| | - Thomas J LaRocca
- Department of Integrative Physiology, University of Colorado-Boulder , Boulder, Colorado
| | - Christopher R Martens
- Department of Integrative Physiology, University of Colorado-Boulder , Boulder, Colorado
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado-Boulder , Boulder, Colorado
| |
Collapse
|
47
|
Liang Y, Elgendi M, Chen Z, Ward R. An optimal filter for short photoplethysmogram signals. Sci Data 2018; 5:180076. [PMID: 29714722 PMCID: PMC5928853 DOI: 10.1038/sdata.2018.76] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022] Open
Abstract
A photoplethysmogram (PPG) contains a wealth of cardiovascular system information, and with the development of wearable technology, it has become the basic technique for evaluating cardiovascular health and detecting diseases. However, due to the varying environments in which wearable devices are used and, consequently, their varying susceptibility to noise interference, effective processing of PPG signals is challenging. Thus, the aim of this study was to determine the optimal filter and filter order to be used for PPG signal processing to make the systolic and diastolic waves more salient in the filtered PPG signal using the skewness quality index. Nine types of filters with 10 different orders were used to filter 219 (2.1s) short PPG signals. The signals were divided into three categories by PPG experts according to their noise levels: excellent, acceptable, or unfit. Results show that the Chebyshev II filter can improve the PPG signal quality more effectively than other types of filters and that the optimal order for the Chebyshev II filter is the 4th order.
Collapse
Affiliation(s)
- Yongbo Liang
- School of Electrical and Computer Engineering, University of British Columbia, Vancouver, V6T 1Z4, Canada.,School of Electronic Engineering and Automation, Guilin University of Electronic Technology, Guilin 541004, PR China.,School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin 541004, PR China
| | - Mohamed Elgendi
- School of Electrical and Computer Engineering, University of British Columbia, Vancouver, V6T 1Z4, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, V6T 1Z3, Canada.,BC Children's & Women's Hospital, Vancouver, V6H 3N1, Canada
| | - Zhencheng Chen
- School of Electronic Engineering and Automation, Guilin University of Electronic Technology, Guilin 541004, PR China.,School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin 541004, PR China
| | - Rabab Ward
- School of Electrical and Computer Engineering, University of British Columbia, Vancouver, V6T 1Z4, Canada
| |
Collapse
|
48
|
Hughes TM, Wagenknecht LE, Craft S, Mintz A, Heiss G, Palta P, Wong D, Zhou Y, Knopman D, Mosley TH, Gottesman RF. Arterial stiffness and dementia pathology: Atherosclerosis Risk in Communities (ARIC)-PET Study. Neurology 2018; 90:e1248-e1256. [PMID: 29549223 PMCID: PMC5890613 DOI: 10.1212/wnl.0000000000005259] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/20/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Arterial stiffness has been associated with evidence of cerebral small vessel disease (cSVD) and fibrillar β-amyloid (Aβ) deposition in the brain. These complex relationships have not been examined in racially and cognitively diverse cohorts. METHODS The Atherosclerosis Risk in Communities (ARIC)-Neurocognitive Study collected detailed cognitive testing for adjudication of dementia and mild cognitive impairment (MCI), brain MRI, and arterial stiffness by pulse wave velocity (PWV, carotid-femoral [cfPWV] and heart-carotid [hcPWV]). The ARIC-PET ancillary study added Aβ imaging using florbetapir ([18F]-AV-45) to obtain standardized uptake volume ratios and defined global Aβ-positivity as standardized uptake volume ratio >1.2. One-SD increase in PWV was related to brain volume, MRI-defined cSVD (e.g., cerebral microbleeds and white matter hyperintensity), and cortical Aβ deposition adjusted for age, body mass index, sex, race, and APOE ε4 status. We examined the cross-sectional relationships including interactions by race, APOE ε4 status, and cognition. RESULTS Among the 320 ARIC-PET participants (76 [5] years, 45% black, 27% MCI), greater central stiffness (hcPWV) was associated with greater Aβ deposition (odds ratio [OR] = 1.31, 95% confidence interval [CI] 1.01-1.71). Greater central stiffness (cfPWV) was significantly associated with having lower brain volumes in Alzheimer disease-susceptible regions (in mm3, β = -1.5 [0.7 SD], p = 0.03) and high white matter hyperintensity burden (OR = 1.6, 95% CI 1.2-2.1). Furthermore, cfPWV was associated with a higher odds of concomitant high white matter hyperintensity and Aβ-positive scans (OR = 1.4, 95% CI 1.1-2.1). These associations were strongest among individuals with MCI and did not differ by race or APOE ε4 status. CONCLUSIONS Arterial stiffness, measured by PWV, is an emerging risk factor for dementia through its repeated relationships with cognition, cSVD, and Aβ deposition.
Collapse
Affiliation(s)
- Timothy M Hughes
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson.
| | - Lynne E Wagenknecht
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Suzanne Craft
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Akiva Mintz
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Gerardo Heiss
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Priya Palta
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Dean Wong
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Yun Zhou
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - David Knopman
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Thomas H Mosley
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| | - Rebecca F Gottesman
- From the Departments of Internal Medicine (T.M.H., S.C.) and Radiology (A.M.), and Division of Public Health Sciences (L.E.W.), Wake Forest School of Medicine, Winston-Salem, NC; Department of Epidemiology (G.H., P.P.), University of North Carolina at Chapel Hill; Departments of Radiology (D.W., Y.Z.) and Neurology (R.F.G.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (D.K.), Mayo Clinic, Rochester, MN; and Department of Medicine (T.H.M.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
49
|
Hughes TM, Lockhart SN, Smagula SF. Blood Pressure's Role in Alzheimer Disease Pathology. Am J Geriatr Psychiatry 2018; 26:23-24. [PMID: 29050913 PMCID: PMC5759321 DOI: 10.1016/j.jagp.2017.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 09/18/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC.
| | - Samuel N Lockhart
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Stephen F Smagula
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
50
|
Rouzaud-Laborde C, Lafitte P, Balardy L, Czosnyka Z, Schmidt EA. ICP and Antihypertensive Drugs. ACTA NEUROCHIRURGICA. SUPPLEMENT 2018; 126:163-165. [PMID: 29492554 DOI: 10.1007/978-3-319-65798-1_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVES Arterial hypertension is among the leading risks for mortality. This burden requires in hypertensive patients the use of single, double or more antihypertensive drugs. The relationship between intracranial pressure (ICP) and arterial blood pressure is complex and still under debate. The impact of antihypertensive drugs on ICP is unknown. We wanted to understand whether the use of antihypertensive drugs has a significant influence on ICP and cerebrospinal fluid (CSF)/brain related parameters. MATERIALS AND METHODS In a cohort of 95 patients with suspected normal pressure hydrocephalus, we prospectively collected drug details according to the Anatomical Therapeutic Chemical (ATC) classification. Lumbar infusion studies were performed. Using ICM+ software, we calculated at baseline and plateau ICP and pulse amplitude, resistance to CSF outflow, elastance, and pressure in the sagittal sinus and CSF production rate. We studied the influence of the administration of 1, 2, 3 or more antihypertensive drugs on ICP-derived parameters. We compared the data using Student's and Mann-Whitney tests or Chi-squared and Fisher's exact test. RESULTS Elastance is significantly higher in patients with at least one antihypertensive drug compared with patients without medication. On the contrary, pressure volume index (PVI) is significantly decreased in patients with antihypertensive drugs compared with patients not on these medications. However, the number of antihypertensive drugs does not seem to influence other ICP parameters. CONCLUSIONS Patients on antihypertensive drugs seem to have a stiffer brain than those not on them.
Collapse
Affiliation(s)
| | - Pierre Lafitte
- Department of Pharmacy, University Hospital Toulouse, Toulouse, France
| | - Laurent Balardy
- Department of Geriatry, University Hospital Toulouse, Toulouse, France
| | - Zofia Czosnyka
- Brain Physics Lab, Academic Neurosurgery, University of Cambridge, Cambridge, UK
| | - Eric A Schmidt
- Department of Neurosurgery, University Hospital Toulouse, Toulouse, France
| |
Collapse
|