1
|
Gassner GM, Damestani NL, Wheeler NS, Kufer JA, Yadav SM, Mellen SF, Maina KN, Salat DH, Juttukonda MR. Cerebral microvascular physiology associated with white matter lesion burden differs by level of vascular risk in typically aging older adults. J Cereb Blood Flow Metab 2025; 45:885-896. [PMID: 39568243 PMCID: PMC11580122 DOI: 10.1177/0271678x241300394] [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: 03/21/2024] [Revised: 09/16/2024] [Accepted: 10/13/2024] [Indexed: 11/22/2024]
Abstract
White matter lesions (WMLs) are prevalent with aging, and higher WML burden has been observed in older adults with vascular diseases. While the physiology underlying the formation of WMLs is not known, various risk factors are associated with high WML burden. Here, we investigated the relationship between vascular risk factors and microvascular physiology (i.e., oxygen supply and oxygen extraction fraction [OEF]) and their association with WML burden. Forty-one typically aging adults (60-80 years) were classified into high or low vascular risk based on common modifiable vascular risk factors (hypertension, diabetes, hyperlipidemia, and overweight). These groups were subdivided into high or low WML burden. Differences in microvascular physiology (oxygen supply and OEF) were then compared between and within groups. Overall, OEF was significantly higher in the high vascular risk group compared to the low vascular risk group (p < 0.01). In the low vascular risk subgroup, OEF was uniquely lower in the individuals with high WML versus low WML burden (p = 0.02), despite no differences in oxygen supply between these subgroups (p = 0.87). The coupling of impaired OEF with the absence of compensatory physiology, such as elevated oxygen supply, may represent an important mechanism underlying WML burden in individuals with low vascular risk factors.
Collapse
Affiliation(s)
- Gabriele M Gassner
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Faculty of Medicine, Kiel University, Kiel, Germany
| | - Nikou L Damestani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Natalie S Wheeler
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jan A Kufer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Shrikanth M Yadav
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sarah F Mellen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Katherine N Maina
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - David H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Neuroimaging for Veterans Center, VA Boston Healthcare System, Boston, MA, USA
| | - Meher R Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Küpeli Akkol E, Karatoprak GŞ, Dumlupınar B, Bahadır Acıkara Ö, Arıcı R, Yücel Ç, Aynal LC, Sobarzo Sánchez E. Stilbenes Against Alzheimer's Disease: A Comprehensive Review of Preclinical Studies of Natural and Synthetic Compounds Combined with the Contributions of Developed Nanodrug Delivery Systems. Molecules 2025; 30:1982. [PMID: 40363789 PMCID: PMC12073496 DOI: 10.3390/molecules30091982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
This review covers preclinical studies of stilbene derivative compounds (both natural and synthetic) with potential preventive and therapeutic effects against Alzheimer's disease (AD). AD is a worldwide neurodegenerative disease characterized by the destruction of nerve cells in the brain and the loss of cognitive function due to aging. Stilbenes are a unique class of natural phenolic compounds distinguished by a C6-C2-C6 (1,2-diphenylethylene) structure and two aromatic rings connected by an ethylene bridge. Stilbenes' distinct features make them an intriguing subject for pharmacological research and development. Several preclinical studies have suggested that stilbenes may have neuroprotective effects by reducing Aβ generation and oligomerization, enhancing Aβ clearance, and regulating tau neuropathology through the prevention of aberrant tau phosphorylation and aggregation, as well as scavenging reactive oxygen species. Synthetic stilbene derivatives also target multiple pathways involved in neuroprotection and have demonstrated promising biological activity in vitro. However, some properties of stilbenes, such as sensitivity to physiological conditions, low solubility, poor permeability, instability, and low bioavailability, limit their usefulness in clinical applications. To address this issue, current investigations have developed new drug delivery systems based on stilbene derivative molecules. This review aims to shed light on the development of next-generation treatment strategies by examining in detail the role of stilbenes in Alzheimer's pathophysiology and their therapeutic potential.
Collapse
Affiliation(s)
- Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara 06330, Türkiye
| | - Gökçe Şeker Karatoprak
- Department of Pharmacognosy, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Türkiye;
| | - Berrak Dumlupınar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Okan University, İstanbul 34959, Türkiye;
| | - Özlem Bahadır Acıkara
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara University, Ankara 06560, Türkiye;
| | - Reyhan Arıcı
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara Medipol University, Ankara 06570, Türkiye;
| | - Çiğdem Yücel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Türkiye;
| | - Leyli Can Aynal
- Etlik City Hospital, Department of Neurology, Ankara 06170, Türkiye;
| | - Eduardo Sobarzo Sánchez
- Centro de Investigación en Ingeniería de Materiales, Facultad de Medicina y Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
3
|
Moran KL, Smith CJ, McManus E, Allan SM, Montaldi D, Muhlert N. Cerebrovascular health impacts processing speed through anterior white matter alterations: a UK biobank study. Sci Rep 2025; 15:9860. [PMID: 40119105 PMCID: PMC11928594 DOI: 10.1038/s41598-025-93399-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Cerebrovascular disease is associated with an increased likelihood of developing dementia. Cerebrovascular risk factors are modifiable and may reduce the risk of later-life cognitive dysfunction, however, the relationship between cerebrovascular risk factors, brain integrity and cognition remains poorly characterised. Using a UK Biobank sample of mid-to-old aged adults, without neurological disease, our structural equation mediation models showed that poor cerebrovascular health, indicated by the presence of cerebrovascular risk factors, was associated with slowed processing speed. This effect was best explained by anterior white matter microstructure (e.g. genu, anterior corona radiata), rather than posterior (e.g. splenium, posterior corona radiata)-the mediatory effect of anterior white matter strengthened further with age. Effects were also significantly reduced when considering other forms of cognition, demonstrating both regional- and cognitive-specificity. Our findings also illustrate that cerebrovascular risk factors cross-sectionally predict cognitive processing speed performance, which can be further strengthened by accounting for risk factor duration, particularly hypertensive duration. In summary, our study highlights the vulnerability of anterior regions and sensitivity of processing speed performance to cerebrovascular burden, and show this effect is amplified with age. We also highlight an improved method of cerebrovascular burden quantification, which accounts for factor duration, as well as risk factor presence and degree. Future work will aim to establish the role of medication and effective risk factor control in alleviating or preventing white matter pathology and cognitive dysfunction.
Collapse
Affiliation(s)
- Katie L Moran
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK.
| | - Craig J Smith
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Elizabeth McManus
- Division of Psychology and Mental Health, School of Health Sciences, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Daniela Montaldi
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
| | - Nils Muhlert
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
| |
Collapse
|
4
|
He Y, Zhu T, Bei E, Xiang G, Xi D, Meng H, Bei Y. Hyperuricemia reduces the risk of MCI but not dementia: a cross-sectional study in Liuyang. Front Neurol 2025; 16:1555587. [PMID: 40166637 PMCID: PMC11955452 DOI: 10.3389/fneur.2025.1555587] [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: 01/05/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Background Cognitive impairments, including mild cognitive impairment (MCI) and dementia, significantly affect patients' daily functions and quality of life, contributing to a substantial societal and economic burden. The role of uric acid in cognitive function is controversial, with some studies suggesting protective effects, while others indicate increased risk of cognitive decline. Methods A total of 1,098 participants with an average age of 69 years were included in the study. Cognitive function was assessed using the Mini-Mental State Examination. Hyperuricemia was defined as blood uric acid concentrations >420 μmol/L. Logistic regression and restricted cubic spline analysis were performed to assess the association between hyperuricemia and cognitive impairment, including MCI and dementia. Results Hyperuricemia was associated with a lower risk of cognitive impairment (OR = 0.51, 95% CI = 0.31-0.80) and MCI (OR = 0.39, 95% CI = 0.21-0.69), particularly in individuals younger than 70 years, males, and those without hypertension or diabetes. No significant association was found between hyperuricemia and dementia (OR = 0.94, 95% CI = 0.44-1.89). There is no evidence of a non-linear relationship between hyperuricemia and cognitive impairment. Conclusion Hyperuricemia appears to have a protective effect on cognitive function, particularly in reducing the risk of MCI, but not dementia, in specific populations.
Collapse
Affiliation(s)
- Yong He
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Neurology, Liuyang Jili Hospital, Changsha, China
| | - Tieshi Zhu
- Department of Neurology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Erxinxian Bei
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Guangpeng Xiang
- Department of Hematology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Danyang Xi
- Department of Anesthesiology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Heng Meng
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yuzhang Bei
- Department of Neurology, Liuyang Jili Hospital, Changsha, China
| |
Collapse
|
5
|
Miller T, Bittner N, Moebus S, Caspers S. Identifying sources of bias when testing three available algorithms for quantifying white matter lesions: BIANCA, LPA and LGA. GeroScience 2025; 47:1221-1237. [PMID: 39115640 PMCID: PMC11872996 DOI: 10.1007/s11357-024-01306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/29/2024] [Indexed: 03/04/2025] Open
Abstract
Brain magnetic resonance imaging frequently reveals white matter lesions (WMLs) in older adults. They are often associated with cognitive impairment and risk of dementia. Given the continuous search for the optimal segmentation algorithm, we broke down this question by exploring whether the output of algorithms frequently used might be biased by the presence of different influencing factors. We studied the impact of age, sex, blood glucose levels, diabetes, systolic blood pressure and hypertension on automatic WML segmentation algorithms. We evaluated three widely used algorithms (BIANCA, LPA and LGA) using the population-based 1000BRAINS cohort (N = 1166, aged 18-87, 523 females, 643 males). We analysed two main aspects. Firstly, we examined whether training data (TD) characteristics influenced WML estimations, assessing the impact of relevant factors in the TD. Secondly, algorithm's output and performance within selected subgroups defined by these factors were assessed. Results revealed that BIANCA's WML estimations are influenced by the characteristics present in the TD. LPA and LGA consistently provided lower WML estimations compared to BIANCA's output when tested on participants under 67 years of age without risk cardiovascular factors. Notably, LPA and LGA showed reduced accuracy for these participants. However, LPA and LGA showed better performance for older participants presenting cardiovascular risk factors. Results suggest that incorporating comprehensive cohort factors like diverse age, sex and participants with and without hypertension in the TD could enhance WML-based analyses and mitigate potential sources of bias. LPA and LGA are a fast and valid option for older participants with cardiovascular risk factors.
Collapse
Affiliation(s)
- Tatiana Miller
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Nora Bittner
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany.
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany.
| | - Susanne Moebus
- Institute for Urban Public Health, University Hospital Essen and University Duisburg-Essen, Essen, Germany
| | - Svenja Caspers
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| |
Collapse
|
6
|
Jiang X, Schreiner PJ, Gunderson EP, Yaffe K. Hypertensive Disorders of Pregnancy and Brain Health in Midlife: The CARDIA Study. Hypertension 2025; 82:197-205. [PMID: 39162043 PMCID: PMC11735310 DOI: 10.1161/hypertensionaha.124.22857] [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: 02/06/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND To understand the role of hypertensive disorders of pregnancy (HDP), including preeclampsia and gestational hypertension (GH), in brain health earlier in life, we investigated the association of HDP with midlife cognition and brain health. METHODS We studied a prospective cohort of women, baseline age 18 to 30 years, who were assessed at study years 25 and 30 with a cognitive battery and a subset with brain magnetic resonance imaging. A history of HDP was defined based on self-report. We conducted linear regression to assess the association of a history of preeclampsia, GH, or no HDP with cognition and brain magnetic resonance imaging white matter hyperintensities. RESULTS Among 1441 women (mean age, 55.2±3.6 years), 202 reported preeclampsia and 112 reported GH. GH was associated with worse cognitive performance: global cognition (mean score, 23.2 versus 24.0; P=0.018), processing speed (67.5 versus 71.3; P=0.01), verbal fluency (29.5 versus 31.1; P=0.033), and a trend for executive function (24.3 versus 22.6; P=0.09), after multivariable adjustment. GH was associated with a greater 5-year decline in processing speed (mean change, -4.9 versus -2.7; P=0.049) and executive function (-1.7 versus 0.3; P=0.047); preeclampsia was associated with a greater 5-year decline on delayed verbal memory (-0.3 versus 0.1; P=0.041). GH and preeclampsia were associated with greater white matter hyperintensities in the parietal and frontal lobes, respectively. CONCLUSIONS GH and preeclampsia are associated with cognition and white matter hyperintensities during midlife, with differences in cognitive domains and brain lobes. Women with HDP may need to be closely monitored for adverse brain outcomes starting in midlife.
Collapse
Affiliation(s)
- Xiaqing Jiang
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco
| | | | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco
- Department of Neurology, Epidemiology and Biostatistics, University of California San Francisco
- San Francisco VA Health Care System
| |
Collapse
|
7
|
Hou YC, Tsai CC, Chen RM, Liu YC, Lu KC, Chen YL, Shen TW, Wang JJ. Cerebral white matter damage in patients with end-stage kidney disease associates with cognitive impairment. Clin Kidney J 2025; 18:sfae283. [PMID: 39781476 PMCID: PMC11704783 DOI: 10.1093/ckj/sfae283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 01/12/2025] Open
Abstract
Background Damage to brain white matter often occurs in individuals with chronic kidney disease, which might be related to their cognitive decline. This study aims to investigate tract-specific white matter damage in patients with end-stage kidney disease by using fixel-based analysis. Methods Images of 31 end-stage kidney disease patients and 16 normal controls (aged: 61.1 ± 10.4 years; 11 men) were acquired from a 1.5T magnetic resonance scanner. The patients were subsequently divided into with normal cognition (N = 17, aged: 66.9 ± 7.2 years; 10 men) and cognitive impairment (N = 14, aged: 72.4 ± 9.4 years; 7 men). Cognitive assessment, and neurologic, hematologic and biochemical samples were collected. Fixel-based analysis was used to examine the tract-specific damage within white matter. Differences between groups were evaluated through connectivity-based fixel enhancement and non-parametric permutation testing. Correlation with biomarkers was conducted through general linear model. Significance was determined with family-wise error-corrected P-value <.05. Results Reduced fixel-based metrics were observed in specific tract located the cerebral peduncle, internal capsule, corpus callosum, fornix and superior corona radiata in patients when compared with normal controls, indicating a reduction in fiber content. The fibers crossing the corpus callosum and the fornix/stria terminalis are particularly vulnerable sites, which can be associated with the decrease in both Mini-Mental State Examination (R2 ranged between 0.420 and 0.556) and Montreal Cognitive Assessment (R2 ranged between 0.425 and 0.509), as well as the plasma concentration of calcium (R2 ranged between 0.207 and 0.322). The plasma concentration of indoxyl sulfate was associated with the descending tracts from right posterior limb of internal capsule to cerebral peduncle (R2 ranged between 0.262 and 0.335). Conclusions Tract-specific white matter damage can be noticed in the patients with end-stage kidney disease, and could be associated with their cognitive decline.
Collapse
Affiliation(s)
- Yi-Chou Hou
- Department of Internal Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Chien Tsai
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ruei-Ming Chen
- TMU Research Center of Cancer Translational Medicine, Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei City, Taiwan
| | - Yi-Chien Liu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Neurology, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Kuo-Cheng Lu
- Department of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yao-Liang Chen
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung City, Taiwan
| | - Ting-Wen Shen
- Department of Medical Imaging and Intervention, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jiun-Jie Wang
- Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan City, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung City, Taiwan
- Department of Chemical Engineering, Ming-Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
8
|
Cox JG, Cole JH, Kempton MJ, Williams SCR, de Groot M. Volume and distribution of white matter hyperintensities in rheumatoid arthritis and ulcerative colitis patients. Sci Rep 2024; 14:32010. [PMID: 39738366 PMCID: PMC11685908 DOI: 10.1038/s41598-024-83559-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Brain white matter disruptions have been implicated in contributing to fatigue, brain fog and other central symptoms commonly reported in inflammatory diseases. In this study, we included 252 RA patients with 756 age and sex matched controls and 240 UC patients with 720 age and sex matched controls using the UK Biobank imaging dataset. We looked for differences in total volume of white matter hyperintensities (WMH) between patients compared to controls. Then, using voxelwise analysis, we explored the spatial distribution of these white matter hyperintensities and differences in these between patients and controls and between disease groups. A significantly higher volume of WMH was observed in both the RA (p = 1.9 × 10-8, β = - 0.36, 95% CI = - 0.48, - 0.23) and UC (p = 0.003, β = - 0.18 95% CI = - 0.31, - 0.06) patients compared to their respective control groups. Voxelwise analysis revealed only a small cluster of RA associated WMH compared to controls. These results indicate an increased risk of white matter hyperintensities in patients with RA and UC. These findings help quantify the effect of inflammation from autoimmune diseases on cerebrovascular health and white matter integrity.
Collapse
Affiliation(s)
- Jennifer G Cox
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - James H Cole
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Matthew J Kempton
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Steven C R Williams
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marius de Groot
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Groover Consulting, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Alagiakrishnan K, Halverson T, Ahmed A, Frishman WH, Aronow WS. Hypertension and Cognitive Disorders. Cardiol Rev 2024:00045415-990000000-00385. [PMID: 39714291 DOI: 10.1097/crd.0000000000000825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Systemic hypertension is possibly the most important modifiable risk factor for the development of cognitive decline, both for mild cognitive impairment (MCI) and dementia. For effective blood pressure (BP) control, it requires proper assessment, using brachial, central, and ambulatory measurements, and monitoring with a focus on different BP parameters. Different BP parameters like pulse pressure, mean arterial pressure, BP variability, and circadian parameters, like nondippers and early morning surge, should be considered in the evaluation for the risk of cognitive decline due to hypertension in middle age and older adults. Chronic hypertension causes vascular remodeling in the brain and leads to brain failure or cognitive decline. Achieving specific BP goals can improve clinical outcomes and possibly slow down cognitive decline for patients with comorbid hypertension and cognitive impairment.
Collapse
Affiliation(s)
| | - Tyler Halverson
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ali Ahmed
- Department of Medicine, Washington, DC VA Medical Center, George Washington University School of Medicine, and Georgetown University School of Medicine, Washington, DC
| | | | - Wilbert S Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
10
|
Trang H, Hartung TJ, Chen Q, Hetzer S, Chien C, Sperber PS, Schmitz-Hübsch T, Asseyer S, Rust R, Mewes D, Anderhalten L, Sy M, Brandt AU, Finke C, Paul F. A quantitative multi-parameter mapping protocol standardized for clinical research in multiple sclerosis. Sci Rep 2024; 14:30481. [PMID: 39681586 DOI: 10.1038/s41598-024-80274-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Quantitative magnetic resonance imaging (qMRI) involves mapping microstructure in standardized units sensitive to histological properties and supplements conventional MRI, which relies on contrast weighted images where intensities have no biophysical meaning. While measuring tissue properties such as myelin, iron or water content is desired in a disease context, qMRI changes may typically reflect mixed influences from aging or pre-clinical degeneration. We used a fast multi-parameter mapping (MPM) protocol for clinical routine at 3T to reconstruct whole-brain quantitative maps of magnetization transfer saturation (MT), proton density (PD), longitudinal (R1), and transverse relaxation rate (R2*) with 1.6 mm isotropic resolution. We report reference MPM values from a healthy population with age and gender distributions typical of multiple sclerosis in whole brain white matter (WM), T2-weighted WM hyperintensities, cortical grey matter and deep grey matter regions and present post-processing optimizations including integration of lesions and normalization of PD maps against cerebrospinal fluid (CSF) for standardized research in multiple sclerosis (MS) and potentially also in related disorders. PD maps were affected by WM abnormalities in MS using WM calibration. The results acknowledge the impact of non-linear age effects on MPM and suggest using CSF calibration for future clinical application in MS.
Collapse
Affiliation(s)
- Henri Trang
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tim J Hartung
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Qianlan Chen
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudia Chien
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pia S Sperber
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Susanna Asseyer
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rebekka Rust
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institut Für Med. Immunologie, Immundefektambulanz, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Darius Mewes
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Biomedical Innovation Academy, Berlin, Germany
| | - Lina Anderhalten
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sy
- Department of Neurology, University of California Irvine, 208 Sprague Hall, Mail Code 4032, Irvine, CA, 92697, USA
| | - Alexander U Brandt
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurology, University of California Irvine, 208 Sprague Hall, Mail Code 4032, Irvine, CA, 92697, USA
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Friedemann Paul
- Experimental and Clinical Research Center, a Cooperation Between Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité - Universitätsmedizin Berlin, Geschäftsführung, Charitéplatz 1, 10117, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- NCRC - Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Mayer C, Walther C, Borof K, Nägele FL, Petersen M, Schell M, Gerloff C, Kühn S, Heydecke G, Beikler T, Cheng B, Thomalla G, Aarabi G. Association between periodontal disease and microstructural brain alterations in the Hamburg City Health Study. J Clin Periodontol 2024; 51:1598-1609. [PMID: 37263624 PMCID: PMC11651723 DOI: 10.1111/jcpe.13828] [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: 12/15/2022] [Revised: 04/24/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
AIM The aim of the PAROBRAIN study was to examine the association of periodontal health with microstructural white matter integrity and cerebral small vessel disease (CSVD) in the Hamburg City Health Study, a large population-based cohort with dental examination and brain magnetic resonance imaging (MRI). MATERIALS AND METHODS Periodontal health was determined by measuring clinical attachment loss (CAL) and plaque index. Additionally, the decayed/missing/filled teeth (DMFT) index was quantified. 3D-FLAIR and 3D-T1-weighted images were used for white matter hyperintensity (WMH) segmentation. Diffusion-weighted MRI was used to quantify peak width of skeletonized mean diffusivity (PSMD). RESULTS Data from 2030 participants were included in the analysis. Median age was 65 years, with 43% female participants. After adjusting for age and sex, an increase in WMH load was significantly associated with more CAL, higher plaque index and higher DMFT index. PSMD was significantly associated with the plaque index and DMFT. Additional adjustment for education and cardiovascular risk factors revealed a significant association of PSMD with plaque index (p < .001) and DMFT (p < .01), whereas effects of WMH load were attenuated (p > .05). CONCLUSIONS These findings suggest an adverse effect of periodontal health on CSVD and white matter integrity. Further research is necessary to examine whether early treatment of periodontal disease can prevent microstructural brain damage.
Collapse
Affiliation(s)
- Carola Mayer
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Carolin Walther
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of Prosthetic Dentistry, Center for Dental and Oral MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Katrin Borof
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Felix L. Nägele
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Marvin Petersen
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Maximilian Schell
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Gerloff
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Simone Kühn
- Department of PsychiatryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Guido Heydecke
- Department of Prosthetic Dentistry, Center for Dental and Oral MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bastian Cheng
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Götz Thomalla
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ghazal Aarabi
- Department of Periodontics, Preventive and Restorative DentistryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of Prosthetic Dentistry, Center for Dental and Oral MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
12
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Fang YT, Liao SF, Chen PL, Yeh TS, Chen CI, Piravej K, Wu CC, Chiu WT, Lam C. Risk of Traumatic Intracranial Hemorrhage After Stroke: A Nationwide Population-Based Cohort Study in Taiwan. J Am Heart Assoc 2024; 13:e035725. [PMID: 39291491 DOI: 10.1161/jaha.124.035725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Stroke and traumatic intracranial hemorrhage (tICH) are major causes of disability worldwide, with stroke exerting significant negative effects on the brain, potentially elevating tICH risk. In this study, we investigated tICH risk in stroke survivors. METHODS AND RESULTS Using relevant data (2017-2019) from Taiwan's National Health Insurance Research Database, we conducted a population-based retrospective cohort study. Patients were categorized into stroke and nonstroke groups, and tICH risk was compared using a Cox proportional-hazards model. Among 164 628 patients with stroke, 1004 experienced tICH. Patients with stroke had a higher tICH risk than nonstroke counterparts (adjusted hazard ratio [HR], 3.49 [95% CI, 3.17-3.84]). Subgroup analysis by stroke type revealed higher tICH risk in hemorrhagic stroke survivors compared with ischemic stroke survivors (HR, 5.64 [95% CI, 4.97-6.39] versus 2.87 [95% CI, 2.58-3.18], respectively). Older patients (≥45 years) with stroke had a higher tICH risk compared with their younger counterparts (<45 years), in contrast to younger patients without stroke (HR, 7.89 [95% CI, 6.41-9.70] versus 4.44 [95% CI, 2.99-6.59], respectively). Dementia and Parkinson disease emerged as significant tICH risk factors (HR, 1.69 [95% CI, 1.44-2.00] versus 2.17 [95% CI, 1.71-2.75], respectively). In the stroke group, the highest tICH incidence density occurred 3 months after stroke, particularly in patients aged >65 years. CONCLUSIONS Stroke survivors, particularly those with hemorrhagic stroke and those aged ≥45 years, face elevated tICH risk. Interventions targeting the high-risk period are vital, with fall injuries potentially contributing to tICH incidence.
Collapse
Affiliation(s)
- Yun-Ting Fang
- School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Taipei Veterans General Hospital Taipei Taiwan
| | - Shu-Fen Liao
- Department of Medical Research, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- School of Public Health, College of Public Health Taipei Medical University Taipei Taiwan
| | - Ping-Ling Chen
- Graduate Institute of Injury Prevention and Control, College of Public Health Taipei Medical University Taipei Taiwan
| | - Tian-Shin Yeh
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Department of Physical Medicine and Rehabilitation, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Epidemiology and Nutrition, Harvard T. H. Chan School of Public Health Harvard University Boston MA
- Department of Physical Medicine and Rehabilitation National Taiwan University Hospital Taipei Taiwan
- Department of Physical Medicine and Rehabilitation, College of Medicine National Taiwan University Taipei Taiwan
| | - Chin-I Chen
- Department of Neurology, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Neurology, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| | - Krisna Piravej
- Department of Rehabilitation Medicine, Faculty of Medicine Chulalongkorn University Bangkok Thailand
- Department of Chula Neuroscience Center King Chulalongkorn Memorial Hospital Bangkok Thailand
| | - Chia-Chieh Wu
- Emergency Department, Wan Fang Hospital Taipei Medical University Taipei Taiwan
| | - Wen-Ta Chiu
- Graduate Institute of Injury Prevention and Control, College of Public Health Taipei Medical University Taipei Taiwan
- Department of Neurosurgery, Shuang Ho Hospital Taipei Medical University New Taipei Taiwan
- AHMC Health System Alhambra CA
| | - Carlos Lam
- Emergency Department, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Emergency, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| |
Collapse
|
14
|
Ishikawa J, Toba A, Futami S, Saito Y, Tamura Y, Araki A, Harada K. Association of pulse pressure and mean blood pressure to frailty, sarcopenia, and cognitive dysfunction in elderly outpatients with history of hypertension. Hypertens Res 2024; 47:2029-2040. [PMID: 38671218 DOI: 10.1038/s41440-024-01684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
This study aimed to evaluate the relationship between pulse pressure (PP) and sarcopenia, frailty, and cognitive function in elderly patients with hypertension. We evaluated 435 elderly patients with a history of hypertension who visited the frail outpatient clinic between July 2015 and October 2021. Data at the 1-, 2-, and 3-year follow-ups were available for 222, 177, and 164 patients, respectively. Sarcopenia, frailty, and cognitive function, including Mini-Mental State Examination (MMSE) scores, were evaluated. The patients' mean age was 79.2 ± 6.3 years (male, 34.9%). PP and mean blood pressure (BP) were 60.1 ± 13.6 mmHg and 94.1 ± 13.0 mmHg, respectively. At baseline, lower PP was associated with probable dementia (MMSE score ≤23 points) (OR = 0.960 per 1 mmHg increase; 95% CI, 0.933-0.989; P = 0.006) in the model adjusted for conventional confounding factors and comorbidities, whereas higher PP was associated with low handgrip strength (OR = 1.018 per 1 mmHg increase; 95% CI, 1.001-1.036; P = 0.041). In multivariate-adjusted logistic regression analysis of patients with preserved handgrip strength at baseline, reductions in PP (OR = 0.844; 95% CI, 0.731-0.974; P = 0.020) and mean BP (OR = 0.861; 95% CI, 0.758-0.979; P = 0.022) were significantly associated with the incidence of low handgrip strength at 3 years. In conclusion, a higher PP induced by increased arterial stiffness was associated with lower handgrip strength, whereas a lower PP was associated with probable dementia. Reduced PP was associated with decreased handgrip strength after three years.
Collapse
Affiliation(s)
- Joji Ishikawa
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| | - Ayumi Toba
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Shutaro Futami
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshihiro Saito
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kazumasa Harada
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
15
|
Cipolli GC, Ribeiro IC, Yasuda CL, Balthazar MLF, Fattori A, Yassuda MS. Frailty and brain changes in older adults without cognitive impairment: A scoping review. Arch Gerontol Geriatr 2024; 123:105395. [PMID: 38492289 DOI: 10.1016/j.archger.2024.105395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/11/2024] [Accepted: 03/02/2024] [Indexed: 03/18/2024]
Abstract
Little is known about changes in the brain associated with frailty, in particular, which brain areas could be related to frailty in older people without cognitive impairment. This scoping review mapped evidence on functional and/or structural brain changes in frail older adults without cognitive impairment. The methodology proposed by the JBI® was used in this study. The search in PubMed, PubMed PMC, BVS/BIREME, EBSCOHOST, Scopus, Web of Science, Embase, and PROQUEST was conducted up to January 2023. Studies included following the population, concepts, context and the screening and data extraction were performed by two independent reviewers. A total of 9,912 records were identified, 5,676 were duplicates and were excluded. The remaining articles were screened; 31 were read in full and 17 articles were included. The results showed that lesions in white matter hyperintensities, reduced volume of the hippocampus, cerebellum, middle frontal gyrus, low gray matter volume, cortical atrophy, decreased connectivity of the supplementary motor area, presence of amyloid-beta peptide (aβ) in the anterior and posterior putamen and precuneus regions were more frequently observed in frail older adults, compared with non-frail individuals. Studies have suggested that such findings may be of neurodegenerative or cerebrovascular origin. The identification of these brain alterations in frail older adults through neuroimaging studies contributes to our understanding of the underlying mechanisms of frailty. Such findings may have implications for the early detection of frailty and implementation of intervention strategies.
Collapse
Affiliation(s)
- Gabriela Cabett Cipolli
- Postgraduate Program in Gerontology, College of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Isadora Cristina Ribeiro
- Postgraduate Program in Medical Pathophysiology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Clarissa Lin Yasuda
- Faculty of Medical Sciences, Department of Neurology, State University of Campinas, Campinas, Brazil
| | | | - André Fattori
- Postgraduate Program in Gerontology, College of Medical Sciences, State University of Campinas, Campinas, Brazil; Faculty of Medical Sciences, Department of Internal Medicine, State University of Campinas, Campinas, Brazil
| | - Mônica Sanches Yassuda
- Postgraduate Program in Gerontology, College of Medical Sciences, State University of Campinas, Campinas, Brazil; Postgraduate Program in Gerontology, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
16
|
Pradeep A, Raghavan S, Przybelski SA, Preboske GM, Schwarz CG, Lowe VJ, Knopman DS, Petersen RC, Jack CR, Graff-Radford J, Cogswell PM, Vemuri P. Can white matter hyperintensities based Fazekas visual assessment scales inform about Alzheimer's disease pathology in the population? Alzheimers Res Ther 2024; 16:157. [PMID: 38987827 PMCID: PMC11234605 DOI: 10.1186/s13195-024-01525-5] [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: 03/05/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND White matter hyperintensities (WMH) are considered hallmark features of cerebral small vessel disease and have recently been linked to Alzheimer's disease (AD) pathology. Their distinct spatial distributions, namely periventricular versus deep WMH, may differ by underlying age-related and pathobiological processes contributing to cognitive decline. We aimed to identify the spatial patterns of WMH using the 4-scale Fazekas visual assessment and explore their differential association with age, vascular health, AD imaging markers, namely amyloid and tau burden, and cognition. Because our study consisted of scans from GE and Siemens scanners with different resolutions, we also investigated inter-scanner reproducibility and combinability of WMH measurements on imaging. METHODS We identified 1144 participants from the Mayo Clinic Study of Aging consisting of a population-based sample from Olmsted County, Minnesota with available structural magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). WMH distribution patterns were assessed on FLAIR-MRI, both 2D axial and 3D, using Fazekas ratings of periventricular and deep WMH severity. We compared the association of periventricular and deep WMH scales with vascular risk factors, amyloid-PET, and tau-PET standardized uptake value ratio, automated WMH volume, and cognition using Pearson partial correlation after adjusting for age. We also evaluated vendor compatibility and reproducibility of the Fazekas scales using intraclass correlations (ICC). RESULTS Periventricular and deep WMH measurements showed similar correlations with age, cardiometabolic conditions score (vascular risk), and cognition, (p < 0.001). Both periventricular WMH and deep WMH showed weak associations with amyloidosis (R = 0.07, p = < 0.001), and none with tau burden. We found substantial agreement between data from the two scanners for Fazekas measurements (ICC = 0.82 and 0.74). The automated WMH volume had high discriminating power for identifying participants with Fazekas ≥ 2 (area under curve = 0.97) and showed poor correlation with amyloid and tau PET markers similar to the visual grading. CONCLUSION Our study investigated risk factors underlying WMH spatial patterns and their impact on global cognition, with no discernible differences between periventricular and deep WMH. We observed minimal impact of amyloidosis on WMH severity. These findings, coupled with enhanced inter-scanner reproducibility of WMH data, suggest the combinability of inter-scanner data assessed by harmonized protocols in the context of vascular contributions to cognitive impairment and dementia biomarker research.
Collapse
Affiliation(s)
| | - Sheelakumari Raghavan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gregory M Preboske
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Christopher G Schwarz
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Petrice M Cogswell
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
17
|
Hannan J, Busby N, Roth R, Wilmskoetter J, Newman-Norlund R, Rorden C, Bonilha L, Fridriksson J. Under pressure: the interplay of hypertension and white matter hyperintensities with cognition in chronic stroke aphasia. Brain Commun 2024; 6:fcae200. [PMID: 38894950 PMCID: PMC11184349 DOI: 10.1093/braincomms/fcae200] [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: 09/06/2023] [Revised: 05/08/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
While converging research suggests that increased white matter hyperintensity load is associated with poorer cognition, and the presence of hypertension is associated with increased white matter hyperintensity load, the relationship among hypertension, cognition and white matter hyperintensities is not well understood. We sought to determine the effect of white matter hyperintensity burden on the relationship between hypertension and cognition in individuals with post-stroke aphasia, with the hypothesis that white matter hyperintensity load moderates the relationship between history of hypertension and cognitive function. Health history, Fazekas scores for white matter hyperintensities and Wechsler Adult Intelligence Scale Matrix Reasoning subtest scores for 79 people with aphasia collected as part of the Predicting Outcomes of Language Rehabilitation study at the Center for the Study of Aphasia Recovery at the University of South Carolina and the Medical University of South Carolina were analysed retrospectively. We found that participants with a history of hypertension had increased deep white matter hyperintensity severity (P < 0.001), but not periventricular white matter hyperintensity severity (P = 0.116). Moderation analysis revealed that deep white matter hyperintensity load moderates the relationship between high blood pressure and Wechsler Adult Intelligence Scale scores when controlling for age, education, aphasia severity and lesion volume. The interaction is significant, showing that a history of high blood pressure and severe deep white matter hyperintensities together are associated with poorer Matrix Reasoning scores. The overall model explains 41.85% of the overall variation in Matrix Reasoning score in this group of participants. These findings underscore the importance of considering cardiovascular risk factors in aphasia treatment, specifically hypertension and its relationship to brain health in post-stroke cognitive function.
Collapse
Affiliation(s)
- Jade Hannan
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, SC 29208, USA
| | - Natalie Busby
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, SC 29208, USA
| | - Rebecca Roth
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
| | - Janina Wilmskoetter
- Department of Health and Rehabilitation Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Chris Rorden
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA
| | - Leonardo Bonilha
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Julius Fridriksson
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
18
|
Chen F, Cao LH, Ma FY, Zeng LL, He JR. Development and validation of a predictive model for severe white matter hyperintensity with obesity. Front Aging Neurosci 2024; 16:1404756. [PMID: 38887608 PMCID: PMC11180876 DOI: 10.3389/fnagi.2024.1404756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Purpose The purpose of the present study was to identify predictors of severe white matter hyperintensity (WMH) with obesity (SWO), and to build a prediction model for screening obese people with severe WMH without Nuclear Magnetic Resonance Imaging (MRI) examination. Patients subjects and methods From September 2020 to October 2021, 650 patients with WMH were recruited consecutively. The subjects were divided into two groups, SWO group and non-SWO group. Univariate and Logistic regression analysis were was applied to explore the potential predictors of SWO. The Youden index method was adopted to determine the best cut-off value in the establishment of the prediction model of SWO. Each parameter had two options, low and high. The score table of the prediction model and nomogram based on the logistic regression were constructed. Of the 650 subjects, 487 subjects (75%) were randomly assigned to the training group and 163 subjects (25%) to the validation group. By resampling the area under the curve (AUC) of the subject's operating characteristics and calibration curves 1,000 times, nomogram performance was verified. A decision curve analysis (DCA) was used to evaluate the nomogram's clinical usefulness. By resampling the area under the curve (AUC) of the subject's operating characteristics and calibration curves 1,000 times, nomogram performance was verified. A decision curve analysis (DCA) was used to evaluate the nomogram's clinical usefulness. Results Logistic regression demonstrated that hypertension, uric acid (UA), complement 3 (C3) and Interleukin 8 (IL-8) were independent risk factors for SWO. Hypertension, UA, C3, IL-8, folic acid (FA), fasting C-peptide (FCP) and eosinophil could be used to predict the occurrence of SWO in the prediction models, with a good diagnostic performance, Areas Under Curves (AUC) of Total score was 0.823 (95% CI: 0.760-0.885, p < 0.001), sensitivity of 60.0%, specificity of 91.4%. In the development group, the nomogram's AUC (C statistic) was 0.829 (95% CI: 0.760-0.899), while in the validation group, it was 0.835 (95% CI: 0.696, 0.975). In both the development and validation groups, the calibration curves following 1,000 bootstraps showed a satisfactory fit between the observed and predicted probabilities. DCA showed that the nomogram had great clinical utility. Conclusion Hypertension, UA, C3, IL-8, FA, FCP and eosinophil models had the potential to predict the incidence of SWO. When the total score of the model exceeded 9 points, the risk of SWO would increase significantly, and the nomogram enabled visualization of the patient's WMH risk. The application prospect of our models mainly lied in the convenient screening of SWO without MRI examination in order to detect SWO and control the WMH hazards early.
Collapse
Affiliation(s)
- Fu Chen
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Medicine, Yinhang Community Health Centre, Shanghai, China
| | - Lin-Hao Cao
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei-Yue Ma
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Li Zeng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Rong He
- Department of Neurology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Huang C, Zhang W, Shen Z, Li M, Yin J, Tang Y, Zhou X, Zhu X, Sun Z. The association between alpha diversity of gut microbiota, neuroimaging markers and cognitive function in cerebral small vessel disease. Brain Res 2024; 1827:148757. [PMID: 38215865 DOI: 10.1016/j.brainres.2024.148757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/11/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
There is increasing recognition of gut microbial dysbiosis in cerebral small vessel disease (CSVD). The altered diversity in a single ecosystem - alpha diversity index of gut microbiota has attracted wide attention. Our study aims to determine whether the alpha diversity index differs among healthy control (HC), CSVD with and without cognitive impairment. Moreover, we investigate the correlation between the alpha diversity index, neuroimaging markers, and cognitive function. We recruited 40 HC, 43 CSVD patients without cognitive impairment (CSVD-NCI), and 35 CSVD patients with mild cognitive impairment (CSVD-MCI). Clinical and neuropsychological assessments, MRI scanning, and gut microbiota analysis were performed on all participants. The alpha diversity indexes Chao1 and Shannon were calculated to evaluate community richness and diversity in a sample, respectively. Individual neuroimaging markers of CSVD and the CSVD burden score were also evaluated. A significantly lower level of Chao 1 rather than the Shannon index was observed in the CSVD subgroups than in the HC group. The level of the Chao 1 index was negatively correlated with both CMB counts, a neuroimaging characteristic of CSVD, and CSVD burden score in patients with CSVD. Additionally, the Chao 1 index has been associated with general cognitive function, information processing speed, and language function in patients with CSVD. Remarkably, the increased CSVD burden score mediated the effects of decreased levels of Chao 1 on information processing speed and language function. Hence, the alterations in species richness may be associated with CSVD-related cognitive impairment and mediated by CSVD neuroimaging markers.
Collapse
Affiliation(s)
- Chaojuan Huang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wei Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Zhu Shen
- Department of Radiology, North District of the First Affiliated Hospital of Anhui Medical University, Hefei 230011, China; Center of Medical Imaging, Anhui Public Health Clinical Center, Hefei 230011, China
| | - Mingxu Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jiabin Yin
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yating Tang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiaoqun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
20
|
Pradeep A, Raghavan S, Przybelski SA, Preboske G, Schwarz CG, Lowe VJ, Knopman DS, Petersen RC, Jack CR, Graff-Radford J, Cogswell PM, Vemuri P. Can white matter hyperintensities based Fazekas visual assessment scales inform about Alzheimer's disease pathology in the population? RESEARCH SQUARE 2024:rs.3.rs-4017874. [PMID: 38558965 PMCID: PMC10980106 DOI: 10.21203/rs.3.rs-4017874/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: 04/04/2024]
Abstract
Background White matter hyperintensities (WMH) are considered hallmark features of cerebral small vessel disease and have recently been linked to Alzheimer's disease pathology. Their distinct spatial distributions, namely periventricular versus deep WMH, may differ by underlying age-related and pathobiological processes contributing to cognitive decline. We aimed to identify the spatial patterns of WMH using the 4-scale Fazekas visual assessment and explore their differential association with age, vascular health, Alzheimer's imaging markers, namely amyloid and tau burden, and cognition. Because our study consisted of scans from GE and Siemens scanners with different resolutions, we also investigated inter-scanner reproducibility and combinability of WMH measurements on imaging. Methods We identified 1144 participants from the Mayo Clinic Study of Aging consisting of older adults from Olmsted County, Minnesota with available structural magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). WMH distribution patterns were assessed on FLAIR-MRI, both 2D axial and 3D, using Fazekas ratings of periventricular and deep WMH severity. We compared the association of periventricular and deep WMH scales with vascular risk factors, amyloid-PET and tau-PET standardized uptake value ratio, WMH volume, and cognition using Pearson partial correlation after adjusting for age. We also evaluated vendor compatibility and reproducibility of the Fazekas scales using intraclass correlations (ICC). Results Periventricular and deep WMH measurements showed similar correlations with age, cardiometabolic conditions score (vascular risk), and cognition, (p < 0.001). Both periventricular WMH and deep WMH showed weak associations with amyloidosis (R = 0.07, p = < 0.001), and none with tau burden. We found substantial agreement between data from the two scanners for Fazekas measurements (ICC = 0.78). The automated WMH volume had high discriminating power for identifying participants with Fazekas ≥ 2 (area under curve = 0.97). Conclusion Our study investigates risk factors underlying WMH spatial patterns and their impact on global cognition, with no discernible differences between periventricular and deep WMH. We observed minimal impact of amyloidosis on WMH severity. These findings, coupled with enhanced inter-scanner reproducibility of WMH data, suggest the combinability of inter-scanner data assessed by harmonized protocols in the context of vascular contributions to cognitive impairment and dementia biomarker research.
Collapse
|
21
|
Li T, Qin R, Li C, Li L, Wang X, Wang L. Diffusion kurtosis imaging of brain white matter alteration in patients with coronary artery disease based on the TBSS method. Front Aging Neurosci 2024; 16:1301826. [PMID: 38425783 PMCID: PMC10901985 DOI: 10.3389/fnagi.2024.1301826] [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: 09/25/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Object The aim of our study was to examine the alterations in microstructure in patients with coronary artery disease (CAD) and cognitive impairment (CI) using diffusion kurtosis imaging (DKI). Additionally, we aimed to investigate the potential correlation between DKI parameters and cognitive function. Materials and methods A total of 28 CAD patients and 30 healthy controls (HC) were prospectively enrolled in our study. All participants underwent routine and diffusion sequences of head imaging. DKE software was utilized to generate various diffusion kurtosis imaging parameters (DKI), including kurtosis fractional anisotropy (KFA), mean kurtosis (MK), axial kurtosis (AK), radial kurtosis (RK), fractional anisotropy (FA), and mean diffusivity (MD). Nonparametric tests were conducted using tract-based spatial statistics (TBSS) to compare the parameter values between the two groups. The parameter values of the significantly different fiber tracts were extracted and correlated with the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scores. Results Compared to the HC group, patients with coronary artery disease exhibited significant reductions in FA values in the bilateral Superior corona radiata, bilateral Anterior corona radiata, bilateral Posterior corona radiata, corpus callosum, left Posterior thalamic radiation, right Posterior limb of internal capsule, Anterior limb of internal capsule, and Cerebral peduncle, as well as in the left Superior longitudinal fasciculus. Additionally, KFA values decreased in the bilateral Anterior corona radiata, bilateral Anterior limb of internal capsule, and Genu of the corpus callosum. The MK values decreased in the right Posterior corona radiata, Retrolenticular part of the internal capsule, Posterior thalamic radiation (including optic radiation), Superior longitudinal fasciculus, and left Posterior thalamic radiation (including optic radiation). Moreover, the RK values decreased in the bilateral Retrolenticular part of the internal capsule, right Posterior thalamic radiation (including optic radiation), and Superior longitudinal fasciculus, as well as in the left Superior longitudinal fasciculus and Posterior thalamic radiation (including optic radiation) (p < 0.01, TFCE corrected), while no significant differences were observed in other parameter values (p > 0.01, TFCE corrected). The FA values of the right posterior limb of the internal capsule (r = 0.610, p = 0.001) and the right cerebral peduncle (r = 0.622, p < 0.001) were positively correlated with MMSE scores. Additionally, a significant correlation between kurtosis and diffusion coefficient parameters (FA and KFA) was observed. Conclusion CAD patients showed radial shrinkage and complexity of brain white matter microstructure. Whole-brain white matter analysis based on TBSS DKI can objectively reflect the characteristics of white matter damage in CAD patients, providing a basis for the auxiliary diagnosis of CAD with CI.
Collapse
Affiliation(s)
- Tong Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Qin
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cuicui Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Li Wang
- Department of Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
22
|
Yang Z, Wen J, Abdulkadir A, Cui Y, Erus G, Mamourian E, Melhem R, Srinivasan D, Govindarajan ST, Chen J, Habes M, Masters CL, Maruff P, Fripp J, Ferrucci L, Albert MS, Johnson SC, Morris JC, LaMontagne P, Marcus DS, Benzinger TLS, Wolk DA, Shen L, Bao J, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Gene-SGAN: discovering disease subtypes with imaging and genetic signatures via multi-view weakly-supervised deep clustering. Nat Commun 2024; 15:354. [PMID: 38191573 PMCID: PMC10774282 DOI: 10.1038/s41467-023-44271-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Disease heterogeneity has been a critical challenge for precision diagnosis and treatment, especially in neurologic and neuropsychiatric diseases. Many diseases can display multiple distinct brain phenotypes across individuals, potentially reflecting disease subtypes that can be captured using MRI and machine learning methods. However, biological interpretability and treatment relevance are limited if the derived subtypes are not associated with genetic drivers or susceptibility factors. Herein, we describe Gene-SGAN - a multi-view, weakly-supervised deep clustering method - which dissects disease heterogeneity by jointly considering phenotypic and genetic data, thereby conferring genetic correlations to the disease subtypes and associated endophenotypic signatures. We first validate the generalizability, interpretability, and robustness of Gene-SGAN in semi-synthetic experiments. We then demonstrate its application to real multi-site datasets from 28,858 individuals, deriving subtypes of Alzheimer's disease and brain endophenotypes associated with hypertension, from MRI and single nucleotide polymorphism data. Derived brain phenotypes displayed significant differences in neuroanatomical patterns, genetic determinants, biological and clinical biomarkers, indicating potentially distinct underlying neuropathologic processes, genetic drivers, and susceptibility factors. Overall, Gene-SGAN is broadly applicable to disease subtyping and endophenotype discovery, and is herein tested on disease-related, genetically-associated neuroimaging phenotypes.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center, San Antonio, TX, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Tachibana A, Iga JI, Tatewaki Y, Thyreau B, Chen H, Ozaki T, Yoshida T, Yoshino Y, Shimizu H, Mori T, Furuta Y, Shibata M, Ohara T, Hata J, Taki Y, Nakaji S, Maeda T, Ono K, Mimura M, Nakashima K, Takebayashi M, Ninomiya T, Ueno SI. Late-Life High Blood Pressure and Enlarged Perivascular Spaces in the Putaminal Regions of Community-Dwelling Japanese Older Persons. J Geriatr Psychiatry Neurol 2024; 37:61-72. [PMID: 37537887 DOI: 10.1177/08919887231195235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND Enlarged perivascular spaces (EPVS) of the brain may be involved in dementia, such as Alzheimer's disease and cerebral small vessel disease (CSVD). Hypertension has been reported to be a risk factor for dementia and CSVD, but the association between blood pressure (BP) and perivascular spaces is still unclear. The aim of this study was to determine the association between BP and EPVS volumes and to examine the interactions of relevant factors. METHODS A total of 9296 community-dwelling subjects aged ≥65 years participated in a brain magnetic resonance imaging and health status screening examination. Perivascular volume was measured using a software package based on deep learning that was developed in-house. The associations between BP and EPVS volumes were examined by analysis of covariance and multiple regression analysis. RESULTS Mean EPVS volumes increased significantly with rising systolic and diastolic BP levels (P for trend = .003, P for trend<.001, respectively). In addition, mean EPVS volumes increased significantly for every 1-mmHg-increment in systolic and diastolic BPs (both P values <.001). These significant associations were still observed in the sensitivity analysis after excluding subjects with dementia. CONCLUSIONS The present data suggest that higher systolic and diastolic BP levels are associated with greater EPVS volumes in cognitively normal older people.
Collapse
Affiliation(s)
- Ayumi Tachibana
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Yasuko Tatewaki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Benjamin Thyreau
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hongkun Chen
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Tomoki Ozaki
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Taku Yoshida
- Department of Neuropsychiatry, Zaidan Niihama Hospital, Ehime, Japan
| | - Yuta Yoshino
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | | | - Takaaki Mori
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Yoshihiko Furuta
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Taki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Shigeyuki Nakaji
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki, Japan
| | - Tetsuya Maeda
- Division of Neurology and Gerontology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | | | - Kenji Nakashima
- National Hospital Organization, Matsue Medical Center, Shimane, Japan
| | - Minoru Takebayashi
- Department of Neuropsychiatry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shu-Ichi Ueno
- Department of Neuropsychiatry, Neuroscience, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| |
Collapse
|
24
|
Tang C, Ma Y, Lei X, Ding Y, Yang S, He D. Hypertension linked to Alzheimer's disease via stroke: Mendelian randomization. Sci Rep 2023; 13:21606. [PMID: 38062190 PMCID: PMC10703897 DOI: 10.1038/s41598-023-49087-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
This study aimed to investigate the relationship between hypertension and Alzheimer's disease (AD) and demonstrate the key role of stroke in this relationship using mediating Mendelian randomization. AD, a neurodegenerative disease characterized by memory loss, cognitive impairment, and behavioral abnormalities, severely affects the quality of life of patients. Hypertension is an important risk factor for AD. However, the precise mechanism underlying this relationship is unclear. To investigate the relationship between hypertension and AD, we used a mediated Mendelian randomization method and screened for mediating variables between hypertension and AD by setting instrumental variables. The results of the mediated analysis showed that stroke, as a mediating variable, plays an important role in the causal relationship between hypertension and AD. Specifically, the mediated indirect effect value for stroke obtained using multivariate mediated MR analysis was 54.9%. This implies that approximately 55% of the risk of AD owing to hypertension can be attributed to stroke. The results suggest that the increased risk of AD owing to hypertension is mediated through stroke. The finding not only sheds light on the relationship between hypertension and AD but also indicates novel methods for the prevention and treatment of AD. By identifying the critical role of stroke in the link between hypertension and AD, this study provides insights into potential interventions that could mitigate the impact of hypertension on AD. This could help develop personalized treatments and help improve the quality of life of patients with AD who suffer from hypertension.
Collapse
Affiliation(s)
- Chao Tang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Yayu Ma
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Xiaoyang Lei
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Yaqi Ding
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Sushuang Yang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China
| | - Dian He
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
25
|
Lee S, Rieu Z, Kim RE, Lee M, Yen K, Yong J, Kim D. Automatic segmentation of white matter hyperintensities in T2-FLAIR with AQUA: A comparative validation study against conventional methods. Brain Res Bull 2023; 205:110825. [PMID: 38000477 DOI: 10.1016/j.brainresbull.2023.110825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
White matter hyperintensities (WMHs) are lesions in the white matter of the brain that are associated with cognitive decline and an increased risk of dementia. The manual segmentation of WMHs is highly time-consuming and prone to intra- and inter-variability. Therefore, automatic segmentation approaches are gaining attention as a more efficient and objective means to detect and monitor WMHs. In this study, we propose AQUA, a deep learning model designed for fully automatic segmentation of WMHs from T2-FLAIR scans, which improves upon our previous study for small lesion detection and incorporating a multicenter approach. AQUA implements a two-dimensional U-Net architecture and uses patch-based training. Additionally, the network was modified to include Bottleneck Attention Module on each convolutional block of both the encoder and decoder to enhance performance for small-sized WMH. We evaluated the performance and robustness of AQUA by comparing it with five well-known supervised and unsupervised methods for automatic segmentation of WMHs (LGA, LPA, SLS, UBO, and BIANCA). To accomplish this, we tested these six methods on the MICCAI 2017 WMH Segmentation Challenge dataset, which contains MRI images from 170 elderly participants with WMHs of presumed vascular origin, and assessed their robustness across multiple sites and scanner types. The results showed that AQUA achieved superior performance in terms of spatial (Dice = 0.72) and volumetric (logAVD = 0.10) agreement with the manual segmentation compared to the other methods. While the recall and F1-score were moderate at 0.49 and 0.59, respectively, they improved to 0.75 and 0.82 when excluding small lesions (≤ 6 voxels). Remarkably, despite being trained on a different dataset with different ethnic backgrounds, lesion loads, and scanners, AQUA's results were comparable to the top 10 ranked methods of the MICCAI challenge. The findings suggest that AQUA is effective and practical for automatic segmentation of WMHs from T2-FLAIR scans, which could help identify individuals at risk of cognitive decline and dementia and allow for early intervention and management.
Collapse
Affiliation(s)
- Soojin Lee
- Research Institute, NEUROPHET Inc., Seoul, South Korea; Pacific Parkinson's Research Centre, The University of British Columbia, Vancouver, Canada.
| | - ZunHyan Rieu
- Research Institute, NEUROPHET Inc., Seoul, South Korea
| | - Regina Ey Kim
- Research Institute, NEUROPHET Inc., Seoul, South Korea
| | - Minho Lee
- Research Institute, NEUROPHET Inc., Seoul, South Korea
| | - Kevin Yen
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Junghyun Yong
- Research Institute, NEUROPHET Inc., Seoul, South Korea
| | - Donghyeon Kim
- Research Institute, NEUROPHET Inc., Seoul, South Korea
| |
Collapse
|
26
|
Grainger SA, Henry JD, Alister M, Bourdaniotis XE, Mead J, Bailey TG, Coombes JS, Vear N. Cardiorespiratory Fitness and Muscular Strength Do Not Predict Social Cognitive Capacity in Older Age. J Gerontol B Psychol Sci Soc Sci 2023; 78:1824-1833. [PMID: 37480568 PMCID: PMC10645310 DOI: 10.1093/geronb/gbad101] [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: 02/20/2023] [Indexed: 07/24/2023] Open
Abstract
OBJECTIVES Social cognitive function often declines in older age but the mechanisms underlying these declines are not completely clear. Cardiorespiratory fitness (CRF) and muscular strength are positively associated with broader cognitive function in older adults, yet surprisingly, no study has examined whether a similar relationship exists between CRF or muscular strength and social cognition in older age. METHODS We assessed whether higher CRF and muscular strength were associated with enhanced social cognitive function in a sample of fifty older adults (Mage = 70.08, standard deviation = 3.93). Participants completed a gold-standard cardiopulmonary exercise test to assess CRF, an isometric handgrip strength test to index muscular strength, and validated measures of social cognition to index emotion perception and theory of mind (ToM). RESULTS The results showed that CRF and muscular strength did not explain any unique variance in older adults' social cognitive performance. Bayesian analyses confirmed that the evidence for the null hypothesis was moderate for all tested relationships, except for the relationship between CRF and cognitive ToM where the evidence for the null was anecdotal. DISCUSSION This study has provided the first evidence to suggest that CRF and muscular strength-two important modifiable lifestyle factors-are not associated with social cognition in healthy older adults. However, replication studies are now needed to cross-validate these findings and to clarify whether any moderating variables may be important for understanding the relationship between fitness and social cognition in older age.
Collapse
Affiliation(s)
- Sarah A Grainger
- School of Psychology, University of Queensland, St Lucia, Queensland, Australia
| | - Julie D Henry
- School of Psychology, University of Queensland, St Lucia, Queensland, Australia
| | - Manikya Alister
- School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Jessica Mead
- School of Psychology, University of Queensland, St Lucia, Queensland, Australia
| | - Tom G Bailey
- School of Human Movement and Nutrition Sciences, University of Queensland, St Lucia, Queensland, Australia
- School of Nursing, Midwifery and Social Work, University of Queensland, St Lucia, Queensland, Australia
| | - Jeff S Coombes
- School of Human Movement and Nutrition Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Natalie Vear
- School of Nursing, Midwifery and Social Work, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
27
|
Liu S, Wang M, Gu D, Li Y, Zhang X, Li H, Ji C, Nie X, Liu J. Optimal systolic and diastolic blood pressure threshold that associated with lower risk of white matter hyperintensity progression. Front Aging Neurosci 2023; 15:1254463. [PMID: 37927340 PMCID: PMC10620971 DOI: 10.3389/fnagi.2023.1254463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Background The optimal control thresholds for systolic blood pressure (SBP) and diastolic blood pressure (DBP) in patients with white matter hyperintensity (WMH) are still unclear. Method A longitudinal retrospective study of patients with brain magnetic resonance imaging (MRI) scans with intervals of more than 3 years was conducted. Blood pressure records during hospitalization and from outpatient visits between baseline and the last MRI scan were collected. The outcome was the change in total WMH from baseline to the final visit. Results Among the 965 patients with MRI scans, 457 patients with detailed longitudinal blood pressure records were ultimately included and classified into the WMH absent group (n = 121), mild WMH group (n = 126), and moderate to severe WMH group (n = 210). Both baseline and longitudinal mean SBP, DBP, and SBP SD were significantly associated with WMH severity (p < 0.05). An average SBP of 130-140 mmHg [vs. <130 mmHg, aOR, 1.80, (95% CI, 1.05-3.07), p = 0.03] was associated with a higher risk of WMH progression. DBP ≥ 90 mmHg [vs. <80 mmHg, OR, 1.81, (95% CI, 0.88-3.74), p = 0.02, aOR, 1.54, (95% CI, 0.66-3.53), p = 0.32] was associated with a higher risk of WMH progression, but was not after adjusted for other covariates. Longitudinal BP variability was not significantly associated with WMH progression. Conclusion Both SBP and DBP had a stronger relationship with the severity of WMH. A target mean SBP of <130 mmHg and mean DBP of <80 mmHg was associated with a lower risk of WMH progression.
Collapse
Affiliation(s)
- Sibo Liu
- Intensive Care Unit, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Mengxing Wang
- China National Clinical Research Center for Neurological Diseases Beijing China, Beijing, China
| | - De’an Gu
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, China
| | - Yanzhao Li
- Department of Neurosurgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xin Zhang
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Hang Li
- Department of Geriatrics, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China
| | - Chenhua Ji
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| | - Ximing Nie
- Neurocritical Care Unit, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinjie Liu
- Department of General Medicine, Dalian Municipal Central Hospital Affiliated Dalian University of Technology, Dalian, China
| |
Collapse
|
28
|
Nakazawa T, Ohara T, Hirabayashi N, Furuta Y, Hata J, Shibata M, Honda T, Kitazono T, Nakao T, Ninomiya T. Association of white matter lesions and brain atrophy with the development of dementia in a community: the Hisayama Study. Psychiatry Clin Neurosci 2023; 77:330-337. [PMID: 36700514 PMCID: PMC11488640 DOI: 10.1111/pcn.13533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/05/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
AIM To investigate the association of white matter lesions volume (WMLV) levels with dementia risk and the association between dementia risk and the combined measures of WMLV and either total brain atrophy or dementia-related gray matter atrophy in a general older population. METHODS One thousand one hundred fifty-eight Japanese dementia-free community-residents aged ≥65 years who underwent brain magnetic resonance imaging were followed for 5.0 years. WMLV were segmented using the Lesion Segmentation Toolbox. Total brain volume (TBV) and regional gray matter volume were estimated by voxel-based morphometry. The WMLV-to-intracranial brain volume ratio (WMLV/ICV) was calculated, and its association with dementia risk was estimated using Cox proportional hazard models. Total brain atrophy, defined as the TBV-to-ICV ratio (TBV/ICV), and dementia-related regional brain atrophy defined based on our previous report were calculated. The association between dementia risk and the combined measures of WMLV/ICV and either total brain atrophy or the number of atrophied regions was also tested. RESULTS During the follow-up, 113 participants developed dementia. The risks of dementia increased significantly with higher WMLV/ICV levels. In addition, dementia risk increased additively both in participants with higher WMLV/ICV levels and lower TBV/ICV levels and in those with higher WMLV/ICV levels and a higher number of dementia-related brain regional atrophy. CONCLUSION The risk of dementia increased significantly with higher WMLV/ICV levels. An additive increment in dementia risk was observed with higher WMLV/ICV levels and lower TBV/ICV levels or a higher number of dementia-related brain regional atrophy, suggesting the importance of prevention or control of cardiovascular risk factors.
Collapse
Grants
- JP22dk0207053 Japan Agency for Medical Research and Development
- JPMH20FA1002 the Health and Labour Sciences Research Grants of the Ministry of Health, Labour and Welfare of Japan
- JP19K07890 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP20K10503 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP20K11020 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP21H03200 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP21K07522 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP21K10448 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP21K11725 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP22K07421 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- JP22K17396 the Ministry of Education, Culture, Sports, Science and Technology of Japan (JSPS KAKENHI)
- Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Taro Nakazawa
- Department of Neuropsychiatry, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Naoki Hirabayashi
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Psychosomatic Medicine, Graduate School of Medical ScienceKyushu UniversityFukuokaJapan
| | - Yoshihiko Furuta
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Medicine and Clinical Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Medicine and Clinical Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Center for Cohort Studies, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Psychosomatic Medicine, Graduate School of Medical ScienceKyushu UniversityFukuokaJapan
- Center for Cohort Studies, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takanori Honda
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Center for Cohort Studies, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Center for Cohort Studies, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
29
|
Zhu Z, Vanderschelden B, Lee SJ, Blackwill H, Shafie M, Soun JE, Chow D, Chang P, Stradling D, Qian T, Yu W. Methamphetamine use increases the risk of cerebral small vessel disease in young patients with acute ischemic stroke. Sci Rep 2023; 13:8494. [PMID: 37231082 DOI: 10.1038/s41598-023-35788-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/24/2023] [Indexed: 05/27/2023] Open
Abstract
Methamphetamine use causes spikes in blood pressure. Chronic hypertension is a major risk factor for cerebral small vessel disease (cSVD). The aim of this study is to investigate whether methamphetamine use increases the risk of cSVD. Consecutive patients with acute ischemic stroke at our medical center were screened for methamphetamine use and evidence of cSVD on MRI of the brain. Methamphetamine use was identified by self-reported history and/or positive urine drug screen. Propensity score matching was used to select non-methamphetamine controls. Sensitivity analysis was performed to assess the effect of methamphetamine use on cSVD. Among 1369 eligible patients, 61 (4.5%) were identified to have a history of methamphetamine use and/or positive urine drug screen. Compared with the non-methamphetamine group (n = 1306), the patients with methamphetamine abuse were significantly younger (54.5 ± 9.7 vs. 70.5 ± 12.4, p < 0.001), male (78.7% vs. 54.0%, p < 0.001) and White (78.7% vs. 50.4%, p < 0.001). Sensitivity analysis showed that methamphetamine use was associated with increased white matter hyperintensities, lacunes, and total burden of cSVD. The association was independent of age, sex, concomitant cocaine use, hyperlipidemia, acute hypertension, and stroke severity. Our findings suggest that methamphetamine use increases the risk of cSVD in young patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Zhu Zhu
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
| | - Benjamin Vanderschelden
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
| | - Sook Joung Lee
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
- Department of Physical Medicine and Rehabilitation, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Haley Blackwill
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
| | - Mohammad Shafie
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
| | - Jennifer E Soun
- Department of Radiological Science, University of California, Irvine, CA, USA
| | - Daniel Chow
- Department of Radiological Science, University of California, Irvine, CA, USA
| | - Peter Chang
- Department of Radiological Science, University of California, Irvine, CA, USA
| | - Dana Stradling
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA
| | - Tianchen Qian
- Department of Statistics, University of California, Irvine, CA, USA
| | - Wengui Yu
- Department of Neurology, University of California Irvine, 200 S. Manchester Ave. Suite 206, Orange, CA, 92868, USA.
| |
Collapse
|
30
|
Tomoto T, Tarumi T, Zhang R. Central arterial stiffness, brain white matter hyperintensity and total brain volume across the adult lifespan. J Hypertens 2023; 41:819-829. [PMID: 36883450 PMCID: PMC10079586 DOI: 10.1097/hjh.0000000000003404] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
OBJECTIVES Mounting evidence suggests that central arterial stiffening is associated with brain ageing in older adults. The purpose of this study was to determine the associations of age with carotid arterial stiffness and carotid-femoral pulse wave velocity (cfPWV), both measurements of central arterial stiffness, the relationship between age-related arterial stiffness, brain white matter hyperintensity (WMH) and total brain volume (TBV), and whether effects of central arterial stiffness on WMH volume and TBV are mediated by pulsatile cerebral blood flow (CBF). METHODS One hundred and seventy-eight healthy adults (21-80 years) underwent measurements of central arterial stiffness using tonometry and ultrasonography, WMH and TBV via MRI, and pulsatile CBF at the middle cerebral artery via transcranial Doppler. RESULTS Advanced age was associated with increases in both carotid arterial stiffness and cfPWV, increases in WMH volume and decreases in TBV (all P < 0.01). Multiple linear regression analysis showed that carotid β-stiffness was positively associated with WMH volume (B = 0.015, P = 0.017) and cfPWV negatively with TBV (B = -0.558, P < 0.001) after adjustment for age, sex and arterial pressure. Pulsatile CBF mediates the associations between carotid β-stiffness and WMH (95% confidence interval: 0.0001-0.0079). CONCLUSION These findings suggest that age-related central arterial stiffness is associated with increased WMH volume and decreased TBV, which is likely mediated by increased arterial pulsation.
Collapse
Affiliation(s)
- Tsubasa Tomoto
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology
| | - Takashi Tarumi
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
31
|
Li Z, Wang W, Sang F, Zhang Z, Li X. White matter changes underlie hypertension-related cognitive decline in older adults. Neuroimage Clin 2023; 38:103389. [PMID: 37004321 PMCID: PMC10102561 DOI: 10.1016/j.nicl.2023.103389] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/18/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
Hypertension has been well recognized as a risk factor for cognitive impairment and dementia. Although the underlying mechanisms of hypertension-affected cognitive deterioration are not fully understood, white matter changes (WMCs) seem to play an important role. WMCs include low microstructural integrity and subsequent white matter macrostructural lesions, which are common on brain imaging in hypertensive patients and are critical for multiple cognitive domains. This article provides an overview of the impact of hypertension on white matter microstructural and macrostructural changes and its link to cognitive dysfunction. Hypertension may induce microstructural changes in white matter, especially for the long-range fibers such as anterior thalamic radiation (ATR) and inferior fronto-occipital fasciculus (IFOF), and then macrostructural abnormalities affecting different lobes, especially the periventricular area. Different regions' WMCs would further exert different effects to specific cognitive domains and accelerate brain aging. As a modifiable risk factor, hypertension might provide a new perspective for alleviating and delaying cognitive impairment.
Collapse
Affiliation(s)
- Zilin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Aging Brain Rejuvenation Initiative Centre, Beijing Normal University, Beijing 100875, China
| | - Wenxiao Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Aging Brain Rejuvenation Initiative Centre, Beijing Normal University, Beijing 100875, China
| | - Feng Sang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Aging Brain Rejuvenation Initiative Centre, Beijing Normal University, Beijing 100875, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Aging Brain Rejuvenation Initiative Centre, Beijing Normal University, Beijing 100875, China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Aging Brain Rejuvenation Initiative Centre, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
32
|
Abellaneda-Pérez K, Cattaneo G, Cabello-Toscano M, Solana-Sánchez J, Mulet-Pons L, Vaqué-Alcázar L, Perellón-Alfonso R, Solé-Padullés C, Bargalló N, Tormos JM, Pascual-Leone A, Bartrés-Faz D. Purpose in life promotes resilience to age-related brain burden in middle-aged adults. Alzheimers Res Ther 2023; 15:49. [PMID: 36915148 PMCID: PMC10009845 DOI: 10.1186/s13195-023-01198-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Disease-modifying agents to counteract cognitive impairment in older age remain elusive. Hence, identifying modifiable factors promoting resilience, as the capacity of the brain to maintain cognition and function with aging and disease, is paramount. In Alzheimer's disease (AD), education and occupation are typical cognitive reserve proxies. However, the importance of psychological factors is being increasingly recognized, as their operating biological mechanisms are elucidated. Purpose in life (PiL), one of the pillars of psychological well-being, has previously been found to reduce the deleterious effects of AD-related pathological changes on cognition. However, whether PiL operates as a resilience factor in middle-aged individuals and what are the underlying neural mechanisms remain unknown. METHODS Data was obtained from 624 middle-aged adults (mean age 53.71 ± 6.9; 303 women) from the Barcelona Brain Health Initiative cohort. Individuals with lower (LP; N = 146) and higher (HP; N = 100) PiL rates, according to the division of this variable into quintiles, were compared in terms of cognitive status, a measure reflecting brain burden (white matter lesions; WMLs), and resting-state functional connectivity, examining system segregation (SyS) parameters using 14 common brain circuits. RESULTS Neuropsychological status and WMLs burden did not differ between the PiL groups. However, in the LP group, greater WMLs entailed a negative impact on executive functions. Subjects in the HP group showed lower SyS of the dorsal default-mode network (dDMN), indicating lesser segregation of this network from other brain circuits. Specifically, HP individuals had greater inter-network connectivity between specific dDMN nodes, including the frontal cortex, the hippocampal formation, the midcingulate region, and the rest of the brain. Greater functional connectivity in some of these nodes positively correlated with cognitive performance. CONCLUSION Expanding previous findings on AD pathology and advanced age, the present results suggest that higher rates of PiL may promote resilience against brain changes already observable in middle age. Furthermore, having a purposeful life implies larger functional integration of the dDMN, which may potentially reflect greater brain reserve associated to better cognitive function.
Collapse
Affiliation(s)
- Kilian Abellaneda-Pérez
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain. .,Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain. .,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Barcelona, Spain.
| | - Gabriele Cattaneo
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - María Cabello-Toscano
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain
| | - Javier Solana-Sánchez
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Lídia Mulet-Pons
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ruben Perellón-Alfonso
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina Solé-Padullés
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Núria Bargalló
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain.,Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain.,Magnetic Resonance Image Core Facility (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Josep M Tormos
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.,Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Barcelona, Spain.,Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Alvaro Pascual-Leone
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain.,Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - David Bartrés-Faz
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143, 08036, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Barcelona, Spain.
| |
Collapse
|
33
|
Singh G, Chanda A. Development and Mechanical Characterization of Artificial Surrogates for Brain Tissues. BIOMEDICAL ENGINEERING ADVANCES 2023. [DOI: 10.1016/j.bea.2023.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
|
34
|
Rashid T, Li K, Toledo JB, Nasrallah I, Pajewski NM, Dolui S, Detre J, Wolk DA, Liu H, Heckbert SR, Bryan RN, Williamson J, Davatzikos C, Seshadri S, Launer LJ, Habes M. Association of Intensive vs Standard Blood Pressure Control With Regional Changes in Cerebral Small Vessel Disease Biomarkers: Post Hoc Secondary Analysis of the SPRINT MIND Randomized Clinical Trial. JAMA Netw Open 2023; 6:e231055. [PMID: 36857053 PMCID: PMC9978954 DOI: 10.1001/jamanetworkopen.2023.1055] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
IMPORTANCE Little is known about the associations of strict blood pressure (BP) control with microstructural changes in small vessel disease markers. OBJECTIVE To investigate the regional associations of intensive vs standard BP control with small vessel disease biomarkers, such as white matter lesions (WMLs), fractional anisotropy (FA), mean diffusivity (MD), and cerebral blood flow (CBF). DESIGN, SETTING, AND PARTICIPANTS The Systolic Blood Pressure Intervention Trial (SPRINT) is a multicenter randomized clinical trial that compared intensive systolic BP (SBP) control (SBP target <120 mm Hg) vs standard control (SBP target <140 mm Hg) among participants aged 50 years or older with hypertension and without diabetes or a history of stroke. The study began randomization on November 8, 2010, and stopped July 1, 2016, with a follow-up duration of approximately 4 years. A total of 670 and 458 participants completed brain magnetic resonance imaging at baseline and follow-up, respectively, and comprise the cohort for this post hoc analysis. Statistical analyses for this post hoc analysis were performed between August 2020 and October 2022. INTERVENTIONS At baseline, 355 participants received intensive SBP treatment and 315 participants received standard SBP treatment. MAIN OUTCOMES AND MEASURES The main outcomes were regional changes in WMLs, FA, MD (in white matter regions of interest), and CBF (in gray matter regions of interest). RESULTS At baseline, 355 participants (mean [SD] age, 67.7 [8.0] years; 200 men [56.3%]) received intensive BP treatment and 315 participants (mean [SD] age, 67.0 [8.4] years; 199 men [63.2%]) received standard BP treatment. Intensive treatment was associated with smaller mean increases in WML volume compared with standard treatment (644.5 mm3 vs 1258.1 mm3). The smaller mean increases were observed specifically in the deep white matter regions of the left anterior corona radiata (intensive treatment, 30.3 mm3 [95% CI, 16.0-44.5 mm3]; standard treatment, 80.5 mm3 [95% CI, 53.8-107.2 mm3]), left tapetum (intensive treatment, 11.8 mm3 [95% CI, 4.4-19.2 mm3]; standard treatment, 27.2 mm3 [95% CI, 19.4-35.0 mm3]), left superior fronto-occipital fasciculus (intensive treatment, 3.2 mm3 [95% CI, 0.7-5.8 mm3]; standard treatment, 9.4 mm3 [95% CI, 5.5-13.4 mm3]), left posterior corona radiata (intensive treatment, 26.0 mm3 [95% CI, 12.9-39.1 mm3]; standard treatment, 52.3 mm3 [95% CI, 34.8-69.8 mm3]), left splenium of the corpus callosum (intensive treatment, 45.4 mm3 [95% CI, 25.1-65.7 mm3]; standard treatment, 83.0 mm3 [95% CI, 58.7-107.2 mm3]), left posterior thalamic radiation (intensive treatment, 53.0 mm3 [95% CI, 29.8-76.2 mm3]; standard treatment, 106.9 mm3 [95% CI, 73.4-140.3 mm3]), and right posterior thalamic radiation (intensive treatment, 49.5 mm3 [95% CI, 24.3-74.7 mm3]; standard treatment, 102.6 mm3 [95% CI, 71.0-134.2 mm3]). CONCLUSIONS AND RELEVANCE This study suggests that intensive BP treatment, compared with standard treatment, was associated with a slower increase of WMLs, improved diffusion tensor imaging, and FA and CBF changes in several brain regions that represent vulnerable areas that may benefit from more strict BP control. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01206062.
Collapse
Affiliation(s)
- Tanweer Rashid
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
| | - Karl Li
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
| | - Jon B. Toledo
- Department of Neurology, University of Florida, Gainesville
- Department of Neurology, Houston Methodist Hospital, Houston, Texas
| | - Ilya Nasrallah
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Nicholas M. Pajewski
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sudipto Dolui
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - John Detre
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Hangfan Liu
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | | | - R. Nick Bryan
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - Jeff Williamson
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Christos Davatzikos
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Sudha Seshadri
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
| | - Lenore J. Launer
- Intramural Research Program, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| |
Collapse
|
35
|
Ben-Shabat M, Awad-Igbaria Y, Sela S, Gross B, Yagil Y, Yagil C, Palzur E. Predisposition to cortical neurodegenerative changes in brains of hypertension prone rats. J Transl Med 2023; 21:51. [PMID: 36707861 PMCID: PMC9881299 DOI: 10.1186/s12967-023-03916-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Substantial evidence suggests that hypertension is a significant risk factor for cognitive decline. However, it is unclear whether the genetic predisposition to hypertension is also associated with cellular dysfunction that promotes neurodegeneration. METHODS Changes in blood pressure were evaluated following dietary salt-loading or administration of a regular diet in Sabra Normotensive (SBN/y) and Sabra Hypertension-prone rats (SBH/y). We performed quantitative RT-PCR and immunofluorescence staining in brain cortical tissues before salt loading and 6 and 9 months after salt loading. To examine the expression of brain cortical proteins involved in the gene regulation (Histone Deacetylase-HDAC2; Histone Acetyltransferase 1-HAT1), stress response (Activating Transcription Factor 4-ATF4; Eukaryotic Initiation Factor 2- eIF2α), autophagy (Autophagy related 4A cysteine peptidase- Atg4a; light-chain 3-LC3A/B; mammalian target of rapamycin complex 1- mTORC1) and apoptosis (caspase-3). RESULTS Prior to salt loading, SBH/y compared to SBN/y expressed a significantly higher level of cortical HAT1 (protein), Caspase-3 (mRNA/protein), LC3A, and ATF4 (mRNA), lower levels of ATG4A (mRNA/protein), LC3A/B, HDAC2 (protein), as well as a lower density of cortical neurons. Following dietary salt loading, SBH/y but not SBN/y developed high blood pressure. In hypertensive SBH/y, there was significant upregulation of cortical HAT1 (protein), Caspase-3 (protein), and eIF2α ~ P (protein) and downregulation of HDAC2 (protein) and mTORC1 (mRNA), and cortical neuronal loss. CONCLUSIONS The present findings suggest that genetic predisposition to hypertension is associated in the brain cortex with disruption in autophagy, gene regulation, an abnormal response to cellular stress, and a high level of cortical apoptosis, and could therefore exacerbate cellular dysfunction and thereby promote neurodegeneration.
Collapse
Affiliation(s)
- Moti Ben-Shabat
- grid.415839.2Research Institute of Galilee Medical Center, Nahariya, Israel ,grid.22098.310000 0004 1937 0503Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel ,grid.415839.2Neurology Department, Galilee Medical Center, Nahariya, Israel
| | - Yaseen Awad-Igbaria
- grid.415839.2Research Institute of Galilee Medical Center, Nahariya, Israel ,grid.22098.310000 0004 1937 0503Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Shifra Sela
- grid.415839.2Research Institute of Galilee Medical Center, Nahariya, Israel ,grid.415839.2Neurology Department, Galilee Medical Center, Nahariya, Israel
| | - Bella Gross
- grid.415839.2Research Institute of Galilee Medical Center, Nahariya, Israel ,grid.415839.2Neurology Department, Galilee Medical Center, Nahariya, Israel
| | - Yoram Yagil
- Laboratory for Molecular Medicine, Barzilai University Medical Center, Ashkelon, Israel ,grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Chana Yagil
- Laboratory for Molecular Medicine, Barzilai University Medical Center, Ashkelon, Israel ,grid.7489.20000 0004 1937 0511Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Eilam Palzur
- grid.415839.2Research Institute of Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
36
|
Gao F, Sun J, Yao M, Song Y, Yi H, Yang M, Ni Q, Kong J, Yuan H, Sun B, Wang Y. SERS "hot spot" enhance-array assay for misfolded SOD1 correlated with white matter lesions and aging. Anal Chim Acta 2023; 1238:340163. [PMID: 36464456 DOI: 10.1016/j.aca.2022.340163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 12/15/2022]
Abstract
Misfolding of superoxide dismutase-1 (SOD1) has been correlated with many neurodegenerative diseases, such as Amyotrophic lateral sclerosis's and Alzheimer's among others. However, it is unclear whether misfolded SOD1 plays a role in another neurodegenerative disease of white matter lesions (WMLs). In this study, a sensitive and specific method based on SERS technique was proposed for quantitative detection of misfolded SOD1 content in WMLs. To fabricate the double antibodysandwich substrates for SERS detection, gold nanostars modified with capture antibody were immobilized on glass substrates to prepare active SERS substrates, and then SERS probes conjugated with a Raman reporter and a specific target antibody were coupled with active SERS substrates. This SERS substrates had been employed for quantitative detection of misfolded SOD1 levels in WMLs and exhibited excellent stability, reliability, and accuracy. Moreover, experimental results indicated that the level of misfolded SOD1 increased with the increase in age and the degree of WMLs. Hence, misfolded SOD1 may be a potential blood marker for WMLs and aging. Meanwhile, SERS-based gold nanostars have great clinical application potential in the screening, diagnosis and treatment of WMLs.
Collapse
Affiliation(s)
- Feng Gao
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Jingyi Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Minmin Yao
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Yanan Song
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China; Medical College of Qingdao University, Qingdao, 266021, China
| | - Hui Yi
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Mingfeng Yang
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Qingbin Ni
- Postdoctoral Workstation, Taian Central Hospital, Taian, 271000, Shandong, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, Canada
| | - Hui Yuan
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China.
| | - Baoliang Sun
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China.
| | - Ying Wang
- Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China.
| |
Collapse
|
37
|
Lee S, Bijsterbosch JD, Almagro FA, Elliott L, McCarthy P, Taschler B, Sala-Llonch R, Beckmann CF, Duff EP, Smith SM, Douaud G. Amplitudes of resting-state functional networks - investigation into their correlates and biophysical properties. Neuroimage 2023; 265:119779. [PMID: 36462729 PMCID: PMC10933815 DOI: 10.1016/j.neuroimage.2022.119779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
Resting-state fMRI studies have shown that multiple functional networks, which consist of distributed brain regions that share synchronised spontaneous activity, co-exist in the brain. As these resting-state networks (RSNs) have been thought to reflect the brain's intrinsic functional organization, intersubject variability in the networks' spontaneous fluctuations may be associated with individuals' clinical, physiological, cognitive, and genetic traits. Here, we investigated resting-state fMRI data along with extensive clinical, lifestyle, and genetic data collected from 37,842 UK Biobank participants, with the object of elucidating intersubject variability in the fluctuation amplitudes of RSNs. Functional properties of the RSN amplitudes were first examined by analyzing correlations with the well-established between-network functional connectivity. It was found that a network amplitude is highly correlated with the mean strength of the functional connectivity that the network has with the other networks. Intersubject clustering analysis showed the amplitudes are most strongly correlated with age, cardiovascular factors, body composition, blood cell counts, lung function, and sex, with some differences in the correlation strengths between sensory and cognitive RSNs. Genome-wide association studies (GWASs) of RSN amplitudes identified several significant genetic variants reported in previous GWASs for their implications in sleep duration. We provide insight into key factors determining RSN amplitudes and demonstrate that intersubject variability of the amplitudes primarily originates from differences in temporal synchrony between functionally linked brain regions, rather than differences in the magnitude of raw voxelwise BOLD signal changes. This finding additionally revealed intriguing differences between sensory and cognitive RSNs with respect to sex effects on temporal synchrony and provided evidence suggesting that synchronous coactivations of functionally linked brain regions, and magnitudes of BOLD signal changes, may be related to different genetic mechanisms. These results underscore that intersubject variability of the amplitudes in health and disease need to be interpreted largely as a measure of the sum of within-network temporal synchrony and amplitudes of BOLD signals, with a dominant contribution from the former.
Collapse
Affiliation(s)
- Soojin Lee
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Pacific Parkinson's Research Institute, University of British Columbia, Canada.
| | - Janine D Bijsterbosch
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Mallinckrodt Institute of Radiology, Washington University Medical School, Washington University in St Louis, USA
| | - Fidel Alfaro Almagro
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Lloyd Elliott
- Department of Statistics and Actuarial Science, Simon Fraser University (SFU), Canada
| | - Paul McCarthy
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Bernd Taschler
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Roser Sala-Llonch
- Department of Biomedicine, Institute of Neurosciences, University of Barcelona, Spain
| | - Christian F Beckmann
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Eugene P Duff
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Department of Brain Sciences, Imperial College London, UK Dementia Research Institute, London UK
| | - Stephen M Smith
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Gwenaëlle Douaud
- Centre for Functional MRI of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| |
Collapse
|
38
|
Timmermans EJ, Leeuwis AE, Bots ML, van Alphen JL, Biessels GJ, Brunner-La Rocca HP, Kappelle LJ, van Rossum AC, van Osch MJP, Vaartjes I. Neighbourhood walkability in relation to cognitive functioning in patients with disorders along the heart-brain axis. Health Place 2023; 79:102956. [PMID: 36525834 DOI: 10.1016/j.healthplace.2022.102956] [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: 09/12/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
This study examined associations of neighbourhood walkability with cognitive functioning (i.e., global cognition, memory, language, attention-psychomotor speed, and executive functioning) in participants without or with either heart failure, carotid occlusive disease, or vascular cognitive impairment. Neighbourhood walkability at baseline was positively associated with global cognition and attention-psychomotor speed. These associations were stronger in patients with vascular cognitive impairment. Individuals who live in residential areas with higher walkability levels were less likely to have impairments in language and executive functioning at two-year follow-up. These findings highlight the importance of the built environment for cognitive functioning in healthy and vulnerable groups.
Collapse
Affiliation(s)
- Erik J Timmermans
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Anna E Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Juliette L van Alphen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - L Jaap Kappelle
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Albert C van Rossum
- Department of Cardiology, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Matthias J P van Osch
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ilonca Vaartjes
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
39
|
Sung Kim J, Bin Bae J, Won Han J, Jong Oh D, Wan Suh S, Hyoung Kim J, Woong Kim K. Association of estimated white matter hyperintensity age with cognition in elderly with controlled hypertension. Neuroimage Clin 2023; 37:103323. [PMID: 36638599 PMCID: PMC9860510 DOI: 10.1016/j.nicl.2023.103323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Hypertension is associated with white matter hyperintensity (WMH) and cognitive impairment. Further, WMH is associated with cognitive impairment including executive, attention and visuospatial functions. The aim of this study was to investigate the effects of controlled hypertension (cHT) and previously developed concept, 'WMH age' on cognitive function and the mediating role of WMH in the effect of cHT on cognitive impairment. METHODS We enrolled 855 Koreans without dementia aged 60 years or older, 326 of whom completed 2-year follow-up assessment. We measured their blood pressure thrice in a sitting position using an automated blood pressure monitoring device. We estimated 'WMH age' of every participant using previously developed WMH probability map of healthy older Koreans. We analyzed the mediating effect of WMH age in the association of cHT and cognitive function using the PROCESS Macro model. RESULTS Old WMH age was associated with a faster decline in the Mini-Mental Status Examination (MMSE; p =.003), Consortium to Establish a Registry for Alzheimer's Disease total score (CERAD-TS; p =.003), and Frontal Assessment Battery (FAB; p =.007). Old WMH age showed an approximately-six times higher risk of incident mild cognitive impairment (OR = 6.47, 95 % CI = 1.37 - 9.50, p =.024) compared to young or normal WMH age over the 2-year follow-up period in the cHT group. WMH age mediated the effects of cHT on the MMSE, CERAD-TS, and FAB scores at baseline and two-year follow-up period. CONCLUSIONS WMH mediates the adverse effect of hypertension on cognitive function. Elders with cHT who have older WMH age may be at a higher risk of cognitive decline.
Collapse
Affiliation(s)
- Jun Sung Kim
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggido, South Korea
| | - Jong Bin Bae
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggido, South Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggido, South Korea
| | - Dae Jong Oh
- Workplace Mental Health Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seung Wan Suh
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Jae Hyoung Kim
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggido, South Korea
| | - Ki Woong Kim
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggido, South Korea; Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, Seoul, South Korea; Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea.
| |
Collapse
|
40
|
Xiao G, Kumar R, Komuro Y, Burguet J, Kakarla V, Azizkhanian I, Sheth SA, Williams CK, Zhang XR, Macknicki M, Brumm A, Kawaguchi R, Mai P, Kaneko N, Vinters HV, Carmichael ST, Havton LA, DeCarli C, Hinman JD. IL-17/CXCL5 signaling within the oligovascular niche mediates human and mouse white matter injury. Cell Rep 2022; 41:111848. [PMID: 36543124 PMCID: PMC10026849 DOI: 10.1016/j.celrep.2022.111848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 10/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease and brain white matter injury are worsened by cardiovascular risk factors including obesity. Molecular pathways in cerebral endothelial cells activated by chronic cerebrovascular risk factors alter cell-cell signaling, blocking endogenous and post-ischemic white matter repair. Using cell-specific translating ribosome affinity purification (RiboTag) in white matter endothelia and oligodendrocyte progenitor cells (OPCs), we identify a coordinated interleukin-chemokine signaling cascade within the oligovascular niche of subcortical white matter that is triggered by diet-induced obesity (DIO). DIO induces interleukin-17B (IL-17B) signaling that acts on the cerebral endothelia through IL-17Rb to increase both circulating and local endothelial expression of CXCL5. In white matter endothelia, CXCL5 promotes the association of OPCs with the vasculature and triggers OPC gene expression programs regulating cell migration through chemokine signaling. Targeted blockade of IL-17B reduced vessel-associated OPCs by reducing endothelial CXCL5 expression. In multiple human cohorts, blood levels of CXCL5 function as a diagnostic and prognostic biomarker of vascular cognitive impairment.
Collapse
Affiliation(s)
- Guanxi Xiao
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rosie Kumar
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yutaro Komuro
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jasmine Burguet
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Visesha Kakarla
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ida Azizkhanian
- New York Medical College, School of Medicine, Valhalla, NY, USA
| | - Sunil A Sheth
- Department of Neurology, UT Health McGovern School of Medicine, Houston, TX, USA
| | - Christopher K Williams
- Department of Neuropathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinhai R Zhang
- Department of Neuropathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michal Macknicki
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew Brumm
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Phu Mai
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Naoki Kaneko
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Harry V Vinters
- Department of Neuropathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leif A Havton
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Choi J, Kim JY, Kwon HJ, Choi HJ, Kim SH, Kim S, Lee J, Park JE. Association of cerebral white matter hyperintensities with coronary artery calcium in a healthy population: a cross-sectional study. Sci Rep 2022; 12:21562. [PMID: 36513747 PMCID: PMC9747792 DOI: 10.1038/s41598-022-25654-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
In brain magnetic resonance imaging (MRI), white matter hyperintensity (WMH) is a commonly encountered finding and is known to reflect cerebral small vessel disease. The aim of our study was to investigate the association of coronary artery calcium (CAC) with WMH and elucidate the relationship between WMH and atherosclerotic risk factors in a large-scale healthy population. This retrospective study included 1337 individuals who underwent brain MRI and CAC scoring computed tomography at healthcare centers affiliated with a tertiary hospital. Cerebral WMH was defined as Fazekas score greater than 2 on brain MRI. Intracranial artery stenosis (ICAS) was also assessed and determined to be present when stenosis was more than 50% on angiography. The associations of risk factors, CAC score, and ICAS with cerebral WMH were assessed by multivariable regression analysis. In multivariable analysis, categories of higher CAC scores showed increased associations with both periventricular and deep WMHs in a dose-dependent relationship. The presence of ICAS was also significantly related to cerebral WMH, and among the clinical variables, age and hypertension were independent risk factors. In conclusion, CAC showed a significant association with cerebral WMH in a healthy population, which might provide evidence for referring to the CAC score to identify individuals with risk of cerebral WMH.
Collapse
Affiliation(s)
- Jinyoung Choi
- grid.264381.a0000 0001 2181 989XDepartment of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Jung Youn Kim
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Heon-Ju Kwon
- grid.264381.a0000 0001 2181 989XDepartment of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Hye Jeong Choi
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Sang Heum Kim
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Sinae Kim
- grid.264381.a0000 0001 2181 989XDivision of Biostatistics, Department of R&D Management, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Jungbin Lee
- grid.412678.e0000 0004 0634 1623Department of Radiology, Soonchunhyang University Bucheon Hospital, Bucheon, Gyeonggi-Do 14584 Republic of Korea
| | - Ji Eun Park
- grid.413967.e0000 0001 0842 2126Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505 Republic of Korea
| |
Collapse
|
42
|
Lespinasse J, Chêne G, Mangin J, Dubois B, Blanc F, Paquet C, Hanon O, Planche V, Gabelle A, Ceccaldi M, Annweiler C, Krolak‐Salmon P, Godefroy O, Wallon D, Sauvée M, Bergeret S, Chupin M, Proust‐Lima C, Dufouil C, for the MEMENTO study group. Associations among hypertension, dementia biomarkers, and cognition: The MEMENTO cohort. Alzheimers Dement 2022. [PMID: 36464896 DOI: 10.1002/alz.12866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Approximately 40% of dementia cases could be delayed or prevented acting on modifiable risk factors including hypertension. However, the mechanisms underlying the hypertension-dementia association are still poorly understood. METHODS We conducted a cross-sectional analysis in 2048 patients from the MEMENTO cohort, a French multicenter clinic-based study of outpatients with either isolated cognitive complaints or mild cognitive impairment. Exposure to hypertension was defined as a combination of high blood pressure (BP) status and antihypertensive treatment intake. Pathway associations were examined through structural equation modeling integrating extensive collection of neuroimaging biomarkers and clinical data. RESULTS Participants treated with high BP had significantly lower cognition compared to the others. This association was mediated by higher neurodegeneration and higher white matter hyperintensities load but not by Alzheimer's disease (AD) biomarkers. DISCUSSION These results highlight the importance of controlling hypertension for prevention of cognitive decline and offer new insights on mechanisms underlying the hypertension-dementia association. HIGHLIGHTS Paths of hypertension-cognition association were assessed by structural equation models. The hypertension-cognition association is not mediated by Alzheimer's disease biomarkers. The hypertension-cognition association is mediated by neurodegeneration and leukoaraiosis. Lower cognition was limited to participants treated with uncontrolled blood pressure. Blood pressure control could contribute to promote healthier brain aging.
Collapse
Affiliation(s)
- Jérémie Lespinasse
- Inserm Research Center « Bordeaux Population Health », Bordeaux School of Public Health, CIC 1401‐EC Bordeaux University Bordeaux France
- Pôle de santé publique Centre Hospitalier Universitaire (CHU) de Bordeaux Bordeaux France
| | - Geneviève Chêne
- Inserm Research Center « Bordeaux Population Health », Bordeaux School of Public Health, CIC 1401‐EC Bordeaux University Bordeaux France
- Pôle de santé publique Centre Hospitalier Universitaire (CHU) de Bordeaux Bordeaux France
| | - Jean‐Francois Mangin
- CATI, US52‐UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP Paris France
- Université Paris‐Saclay, CEA, CNRS, Neurospin, UMR9027 Baobab Gif‐sur‐Yvette France
| | - Bruno Dubois
- Sorbonne Université, CNRS, INSERM Laboratoire d'Imagerie Biomédicale Paris France
- Sorbonne‐Université, Service des maladies cognitives et comportementales et Institut de la mémoire et de la maladie d'Alzheimer (IM2A) Hôpital de la Salpêtrière Paris AP‐PH France
| | - Frederic Blanc
- Univ. Strasbourg, CNRS, ICube laboratory, UMR 7357, Fédération de Médecine Translationnelle de Strasbourg, Centre Mémoire de Ressources et de Recherches Departement de Gériatrie Strasbourg France
| | - Claire Paquet
- Univ. Paris, Inserm U1144, GHU APHP Nord Lariboisière Fernand‐Widal Paris France
| | - Olivier Hanon
- Univ. de Paris, EA 4468, Service de Gériatrie, AP‐HP Hôpital Broca Paris France
| | - Vincent Planche
- Univ. Bordeaux, CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches Pôle de Neurosciences Cliniques, CHU de Bordeaux Bordeaux France
| | - Audrey Gabelle
- Univ. Montpellier, i‐site MUSE, Inserm U1061, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Département de Neurologie, CHU de Montpellier Montpellier France
| | - Mathieu Ceccaldi
- Univ. Aix Marseille, Inserm UMR 1106, Institut de Neurosciences des Systèmes, Centre Mémoire de Ressources et de Recherches Département de Neurologie et de Neuropsychologie, AP‐HM Marseille France
| | - Cedric Annweiler
- Univ. Angers, UPRES EA 4638, Centre Mémoire de Ressources et de Recherches, Département de Gériatrie, CHU d'Angers Angers France
| | - Pierre Krolak‐Salmon
- Univ. Lyon, Inserm U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, Centre Mémoire Ressource et Recherche de Lyon (CMRR), Hôpital des Charpennes Hospices Civils de Lyon Lyon France
| | - Olivier Godefroy
- Neurology Departement and Functional Neurosciences Lab. (UR UPJV 4559) Amiens University Hospital Amiens France
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Neurology and CNR‐MAJ, Normandy Center for Genomic and Personalized Medicine CIC‐CRB1404 Rouen France
| | - Mathilde Sauvée
- CMRR Grenoble Arc Alpin CHU Grenoble Grenoble France
- Laboratoire de Psychologie et NeuroCognition: LPNC CNRS 5105 Université Grenoble Alpes Grenoble France
| | - Sébastien Bergeret
- Département de Médecine NucléaireAP‐HP, Hôpital Pitié‐Salpêtrière ParisFrance
| | - Marie Chupin
- CATI, US52‐UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP Paris France
| | - Cécile Proust‐Lima
- Inserm Research Center « Bordeaux Population Health », Bordeaux School of Public Health, CIC 1401‐EC Bordeaux University Bordeaux France
| | - Carole Dufouil
- Inserm Research Center « Bordeaux Population Health », Bordeaux School of Public Health, CIC 1401‐EC Bordeaux University Bordeaux France
- Pôle de santé publique Centre Hospitalier Universitaire (CHU) de Bordeaux Bordeaux France
| | | |
Collapse
|
43
|
Hu Y, Wu Y, Su H, Tu J, Zeng L, Lei J, Xia L. Exploring the relationship between brain white matter change and higher degree of invisible hand tremor with computer technology. Technol Health Care 2022; 31:921-931. [PMID: 36442160 DOI: 10.3233/thc-220361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND: At present, the clinical diagnosis of white matter change (WMC) patients depends on cranial magnetic resonance imaging (MRI) technology. This diagnostic method is costly and does not allow for large-scale screening, leading to delays in the patient’s condition due to inability to receive timely diagnosis. OBJECTIVE: To evaluate whether the burden of WMC is associated with the degree of invisible hand tremor in humans. METHODS: Previous studies have shown that tremor is associated with WMC, however, tremor does not always have imaging of WMC. Therefore, to confirm that the appearance of WMC causes tremor, which are sometimes invisible to the naked eye, we achieved an optical-based computer-aided diagnostic device by detecting the invisible hand tremor, and we proposed a calculation method of WMC volume by using the characteristics of MRI images. RESULTS: Statistical analysis results further clarified the relationship between WMC and tremor, and our devices are validated for the detection of tremors with WMC. CONCLUSIONS: The burden of WMC volume is positive factor for degree of invisible hand tremor in the participants without visible hand tremor. Detection technology provides a more convenient and low-cost evaluating method before MRI for tremor diseases.
Collapse
Affiliation(s)
- Yang Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Hai Su
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jianglong Tu
- Department of Nephrology Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Luchuan Zeng
- School of Software, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Lei
- School of Software, Nanchang University, Nanchang, Jiangxi, China
| | - Linglin Xia
- School of Software, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
44
|
HUNG STANLEYHUGHWA, KHLIF MOHAMEDSALAH, KRAMER SHARON, WERDEN EMILIO, BIRD LAURAJ, CAMPBELL BRUCECV, BRODTMANN AMY. Poststroke White Matter Hyperintensities and Physical Activity: A CANVAS Study Exploratory Analysis. Med Sci Sports Exerc 2022; 54:1401-1409. [DOI: 10.1249/mss.0000000000002946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
45
|
Mayer C, Nägele FL, Petersen M, Frey BM, Hanning U, Pasternak O, Petersen E, Gerloff C, Thomalla G, Cheng B. Free-water diffusion MRI detects structural alterations surrounding white matter hyperintensities in the early stage of cerebral small vessel disease. J Cereb Blood Flow Metab 2022; 42:1707-1718. [PMID: 35410517 PMCID: PMC9441727 DOI: 10.1177/0271678x221093579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [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
In cerebral small vessel disease (CSVD), both white matter hyperintensities (WMH) of presumed vascular origin and the normal-appearing white matter (NAWM) contain microstructural brain alterations on diffusion-weighted MRI (DWI). Contamination of DWI-derived metrics by extracellular free-water can be corrected with free-water (FW) imaging. We investigated the alterations in FW and FW-corrected fractional anisotropy (FA-t) in WMH and surrounding tissue and their association with cerebrovascular risk factors. We analysed 1,000 MRI datasets from the Hamburg City Health Study. DWI was used to generate FW and FA-t maps. WMH masks were segmented on FLAIR and T1-weighted MRI and dilated repeatedly to create 8 NAWM masks representing increasing distance from WMH. Linear models were applied to compare FW and FA-t across WMH and NAWM masks and in association with cerebrovascular risk. Median age was 64 ± 14 years. FW and FA-t were altered 8 mm and 12 mm beyond WMH, respectively. Smoking was significantly associated with FW in NAWM (p = 0.008) and FA-t in WMH (p = 0.008) and in NAWM (p = 0.003) while diabetes and hypertension were not. Further research is necessary to examine whether FW and FA-t alterations in NAWM are predictors for developing WMH.
Collapse
Affiliation(s)
- Carola Mayer
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix L Nägele
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marvin Petersen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benedikt M Frey
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uta Hanning
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Elina Petersen
- Clinical for Cardiology, University Heart and Vascular Center, Germany.,Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Ueda E, Hirabayashi N, Ohara T, Hata J, Honda T, Fujiwara K, Furuta Y, Shibata M, Hashimoto S, Nakamura S, Nakazawa T, Nakao T, Kitazono T, Ninomiya T, Sonoda KH. Association of Inner Retinal Thickness with Prevalent Dementia and Brain Atrophy in a General Older Population. OPHTHALMOLOGY SCIENCE 2022; 2:100157. [PMID: 36249677 PMCID: PMC9559916 DOI: 10.1016/j.xops.2022.100157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/28/2022] [Accepted: 04/07/2022] [Indexed: 10/27/2022]
|
47
|
Daily blood pressure profile and blood-brain barrier permeability in patients with cerebral small vessel disease. Sci Rep 2022; 12:7723. [PMID: 35545641 PMCID: PMC9095696 DOI: 10.1038/s41598-022-11172-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Cerebral small vessel disease (CSVD) plays an important role in cognitive impairment, stroke, disability, and death. Hypertension is the main risk factor for CSVD. The use of antihypertensive therapy has not resulted in the expected decrease in CSVD complications, which may be related to the underestimation of significance of daily blood pressure profile for blood–brain barrier (BBB) permeability. 53 patients with CSVD of varying severity (mean age 60.08 ± 6.8 years, 69.8% women, subjects with treated long-standing hypertension vs. normotensive subjects − 84.8% vs. 15.2%) and 17 healthy volunteers underwent ambulatory blood pressure monitoring (ABPM) and MRI, including T1-weighted dynamic contrast-enhanced magnetic resonance imaging for assessing BBB permeability. Most of ABPM parameters in CSVD patients did not differ from controls, but were associated with the severity of white matter hyperintensity (WMH) and the total CSVD score. BBB permeability in normal-appearing white matter (NAWM) and grey matter (GM) was significantly higher in CSVD patients, and the severity of BBB permeability remained similar in patients with different stages of WMH. Among BBB permeability parameters, the area under the curve, corresponding to an increase in the contrast transit time in NAWM, had the greatest number of correlations with deviations of ABPM parameters. BBB permeability in CSVD is a universal mechanism of NAWM and GM damage associated with a slight increase in ABPM parameters. It is obvious that the treatment of hypertension in patients with not severe WMH should be more aggressive and carried out under the control of ABPM.
Collapse
|
48
|
Wartolowska KA, Webb AJ. White matter damage due to pulsatile versus steady blood pressure differs by vascular territory: A cross-sectional analysis of the UK Biobank cohort study. J Cereb Blood Flow Metab 2022; 42:802-810. [PMID: 34775867 PMCID: PMC9014677 DOI: 10.1177/0271678x211058803] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Small vessel disease is associated with age, mean blood pressure (MAP) and blood pressure pulsatility (PP). We used data from the UK Biobank cohort study to determine the relative importance of MAP versus PP driving white matter injury within individual white matter tracts, particularly in the anterior and posterior vascular territory. The associations between blood pressure and diffusion indices in 27 major tracts were analysed using unadjusted and fully-adjusted general linear models and mixed-effect linear models. Blood pressure and neuroimaging data were available for 37,041 participants (mean age 64+/-7.5 years, 53% female). In unadjusted analyses, MAP and PP were similarly associated with diffusion indices in the anterior circulation. In the posterior circulation, the associations were weaker, particularly for MAP. In fully-adjusted analyses, MAP remained associated with all diffusion indices in the anterior circulation, independently of age. In the posterior circulation, the effect of MAP became protective. PP remained associated with greater mean diffusivity and extracellular free water diffusion in the anterior circulation and all diffusion indices in the posterior circulation. There was a significant interaction between PP and age. This implies discordant mechanisms for chronic white matter injury in different brain regions and potentially in the associated stroke risks.
Collapse
Affiliation(s)
- Karolina A Wartolowska
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Alastair Js Webb
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| |
Collapse
|
49
|
Olasehinde TA, AKomolafe SF, Oladapo IF, Oyeleye SI. Effect of diet supplemented with African Star Apple Fruit Pulp on purinergic, cholinergic and monoaminergic enzymes, TNF-α expression and redox imbalance in the brain of hypertensive rats. Nutr Neurosci 2022; 26:496-510. [PMID: 35470775 DOI: 10.1080/1028415x.2022.2062925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE This study examined whether diet supplemented with African star apple fruit pulp (FP) can mitigate the effect of high blood pressure on brain neurochemicals, histopathology and expression of genes linked with neuroinflammation. METHODS Rats were administered with cyclosporine (25 mg/kg.bw) to induce hypertension and were fed with or without FP supplemented diet. Purinergic (Nucleoside triphosphate diphosphohydrolases [NTPdase] and adenosine deaminase [ADA]) cholinergic (acetylcholinesterase [AChE]) and monoaminergic (monoamine oxidase-B) enzymes were assessed in treated and untreated hypertensive rats' brains. Oxidative stress biomarkers (catalase, glutathione-S-transferase, thiols, reactive oxygen species [ROS] and malondialdehyde [MDA]), as well as AChE, tumour necrosis factor and receptor (TNF-α and TNF-α-R) expression, were also determined. RESULTS FP supplemented diet significantly reduced NTPdase and ADA activities and increased Na+/K+-ATPase activities in hypertensive rats' brains compared to the untreated group. Furthermore, FP reduced acetylcholinesterase and monoamine oxidase-B activities compared to the hypertensive group. Redox imbalance was observed in hypertensive rats with inhibition of antioxidant enzymes and high levels of ROS and MDA. However, FP supplemented diet improved antioxidant enzymes, reduced ROS and MDA production in the brain of hypertensive rats. High blood pressure also triggered upregulation of AChE, TNF-α and TNF-α-R while feeding with FP supplemented diet downregulated the genes. CONCLUSION This study demonstrates the neuroprotective role of FP supplemented diet against alterations in neurochemicals associated with Alzheimer's disease, oxidative stress-induced neuronal damage and expression of genes linked with neuroinflammation. Moreover, studies on animal behaviour and human subjects are required to confirm these beneficial effects.
Collapse
Affiliation(s)
- Tosin A Olasehinde
- Nutrition and Toxicology Division, Food Technology Department, Federal Institute of Industrial Research, Lagos, Nigeria.,Department of Biochemistry and Microbiology, University of Fort Hare Alice South Africa, Alice, South Africa
| | - Seun F AKomolafe
- Department of Biochemistry, Ekiti State University, Ado-Ekiti, Nigeria
| | - Iyabo F Oladapo
- Department of Basic Medical Science, College of Health Science and Technology, Ijero Ekiti, Nigeria
| | - Sunday I Oyeleye
- Department of Biomedical Technology, Federal University of Technology, Akure, Ondo State.,Functional Food and Nutraceutical Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State
| |
Collapse
|
50
|
Canavan M, O'Donnell MJ. Hypertension and Cognitive Impairment: A Review of Mechanisms and Key Concepts. Front Neurol 2022; 13:821135. [PMID: 35185772 PMCID: PMC8855211 DOI: 10.3389/fneur.2022.821135] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/07/2022] [Indexed: 11/26/2022] Open
Abstract
Cognitive impairment, and dementia, are major contributors to global burden of death and disability, with projected increases in prevalence in all regions of the world, but most marked increases in low and middle-income countries. Hypertension is a risk factor for both Vascular Cognitive Impairment and Alzheimer's disease, the two most common causes of dementia, collectively accounting for 85% of cases. Key end-organ pathological mechanisms, for which hypertension is proposed to be causative, include acute and covert cerebral ischemia and hemorrhage, accelerated brain atrophy, cerebral microvascular rarefaction and endothelial dysfunction, disruption of blood-brain barrier and neuroinflammation that affects amyloid pathologies. In addition to the direct-effect of hypertension on brain structure and microvasculature, hypertension is a risk factor for other diseases associated with an increased risk of dementia, most notably chronic kidney disease and heart failure. Population-level targets to reduce the incidence of dementia are a public health priority. Meta-analyses of blood pressure lowering trials report a significant reduction in the risk of dementia, but the relative (7–11%) and absolute risk reductions (0.4% over 4 years) are modest. However, given the high lifetime prevalence of both conditions, such relative risk reduction would translate into important population-level reductions in dementia globally with effective screening and control of hypertension. Optimal blood pressure target, especially in older adults with orthostatic hypotension, and antihypertensive agent(s) are uncertain. In this review article, we will detail the observational and interventional evidence linking hypertension with cognitive impairment, summarizing the mechanisms through which hypertension causes cognitive decline.
Collapse
Affiliation(s)
- Michelle Canavan
- Health Research Board (HRB), Clinical Research Facility, National University of Ireland, Galway, Ireland
- Galway University Hospital, Galway, Ireland
- *Correspondence: Michelle Canavan
| | - Martin J. O'Donnell
- Health Research Board (HRB), Clinical Research Facility, National University of Ireland, Galway, Ireland
- Galway University Hospital, Galway, Ireland
| |
Collapse
|