1
|
Zhang W, He J, Wang Y, Jin H, Wang R. Scientific status analysis of exercise benefits for vascular cognitive impairment: Evidence of neuroinflammation. J Neuroimmunol 2025; 402:578574. [PMID: 40086400 DOI: 10.1016/j.jneuroim.2025.578574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/07/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
Vascular cognitive impairment (VCI) is a syndrome characterized by cognitive decline resulting from insufficient perfusion to the entire brain or specific brain regions. The lack of a clear understanding of the mechanisms linking cerebrovascular disease to cognitive impairment has impeded the development of targeted treatments for VCI. Increasing evidence indicates that exercise may offer significant benefits for patients with VCI. This study explores how neuroinflammatory mechanisms mediate the effects of exercise on VCI, focusing on the broader biological processes involved. Exercise plays a crucial role in mitigating vascular risk factors, reducing oxidative stress, and promoting neurogenesis. Furthermore, exercise influences neuroinflammatory mediators and central immune cells via various signaling pathways. Different types and intensities of exercise, including resistance and endurance training, have been shown to differentially modulate neuroinflammation during the progression of VCI. This paper summarizes the current mechanisms of action and proposes exercise interventions targeting neuroinflammatory pathways, along with biomarker studies, to enhance our understanding of VCI pathogenesis and inform clinical practice. A more in-depth understanding of the inflammatory mechanisms underlying VCI may facilitate the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Wei Zhang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing He
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuxin Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - He Jin
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China; Beijing Institute of Major Brain Diseases, Beijing, China.
| |
Collapse
|
2
|
Patai R, Patel K, Csik B, Gulej R, Nagaraja RY, Nagy D, Chandragiri SS, Shanmugarama S, Kordestan KV, Nagykaldi M, Ekambaram S, Ungvari A, Yabluchanskiy A, Tarantini S, Benyo Z, Csiszar A, Ungvari Z, Nyul-Toth A. Aging, mitochondrial dysfunction, and cerebral microhemorrhages: a preclinical evaluation of SS-31 (elamipretide) and development of a high-throughput machine learning-driven imaging pipeline for cerebromicrovascular protection therapeutic screening. GeroScience 2025:10.1007/s11357-025-01634-5. [PMID: 40169521 DOI: 10.1007/s11357-025-01634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) contribute to vascular cognitive impairment and dementia (VCID), with aging and hypertension being key risk factors. Mitochondrial oxidative stress is a hallmark of cerebrovascular aging, leading to endothelial dysfunction. This study tests the hypothesis that increased mitochondrial oxidative stress contributes to age-related CMH susceptibility and evaluates the mitochondrial-targeted antioxidative peptide SS-31 (elamipretide) as a potential protective agent in an aged, hypertensive mouse model. Concurrently, we developed a high-throughput, machine learning-driven imaging pipeline to enhance CMH quantification and facilitate the screening of anti-aging vasoprotective interventions. To detect CMHs, brain sections were labeled with diaminobenzidine (DAB) and digitized using a slide scanner-based imaging platform. We developed multiple quantification tools, including color space transformation for enhanced contrast separation and a supervised machine-learning approach utilizing a random forest algorithm to generate whole-brain 3D reconstructions and precisely localize CMHs. We optimized a semi-automated detection method integrating color space transformation and machine learning, benchmarking it against traditional manual counting and color deconvolution-based approaches. While SS-31 treatment did not significantly mitigate hypertension-induced CMH burden in aged mice, our high-throughput imaging pipeline provided a reliable, scalable, and unbiased approach to CMH detection, reducing processing time while improving accuracy. This methodological advancement paves the way for future preclinical studies evaluating therapeutic strategies for cerebrovascular protection in aging. Our findings underscore the need for multi-targeted interventions to mitigate CMH-related neurovascular impairments and prevent VCID.
Collapse
Affiliation(s)
- Roland Patai
- 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
| | - Krish Patel
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Boglarka Csik
- 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
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, 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
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Raghavendra Y Nagaraja
- 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
| | - Dorina Nagy
- 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
| | - Siva Sai Chandragiri
- 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
| | - Santny Shanmugarama
- 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
| | - Kiana Vali Kordestan
- 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
| | - Mark Nagykaldi
- 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
| | - Shoba Ekambaram
- 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
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, 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
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 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
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Benyo
- International Training Program in Geroscience, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, Hungarian Research Network, Semmelweis University (HUN-REN-SU), Budapest, Hungary
| | - 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
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Institute of Translational Medicine, 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
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam Nyul-Toth
- 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
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Yen HY, Lin YH, Wang YF, Fuh JL, Wang SJ, Chen HS, Chiang SC, Li SR, Lin MH, Chen TJ, Hwang SJ, Chang HT. The association between metabolic syndrome components and cognitive function in community-dwelling middle-aged and older adults: the first wave result of a cohort study. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:94. [PMID: 40158120 PMCID: PMC11954201 DOI: 10.1186/s41043-025-00824-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/11/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The components of metabolic syndrome (MetS) have previously been demonstrated to be contributors to cognitive decline in older adults as individual factors, but not collectively as a syndrome. This study investigated whether adults ≥ 50 years old who meet the criteria for MetS were more likely to develop impaired cognition than those without MetS. METHODS Adults aged 50 years or older without significant cognitive impairment who received outpatient care at Taipei Veterans General Hospital were recruited. Waist circumference, blood tests for Mets components, and high-sensitivity C-reactive protein (hsCRP) were measured. Demographics, health condition, cognitive function (by Montreal Cognitive Assessment Taiwanese version, MoCA-T, and AD-8), depression symptoms (by Geriatric Depression Scale-15) and functional status (by Barthel's Index, and Lawton & Brody instrumental activities of daily living, IADL) were evaluated. Associations between MetS and cognitive function were analyzed by multivariate logistic regression. RESULTS Data of 567 participants were analyzed. The prevalence of MetS of the study population was 34.2%. MetS status was not significantly correlated to cognitive decline as indicated by Montreal Cognitive Assessment Taiwan version (p = 0.13) and AD-8 (p = 0.42). Mild abdominal obesity decreased the risk of developing impaired cognition in women (adjusted OR = 0.62, 95% CI = 0.42, 0.93, p = 0.02) but not in men (adjusted OR = 0.84, 95% CI = 0.46, 1.53, p = 0.58). CONCLUSIONS MetS is not a significant contributory factor to cognitive decline in community-dwelling middle-aged and older adults. An optimal waist circumference in community-dwelling older women is protective against the development of mild dementia.
Collapse
Affiliation(s)
- Hsin-Yeong Yen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Lin
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Feng Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jong-Ling Fuh
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shuu-Jiun Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Harn-Shen Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Chiung Chiang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sih-Rong Li
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Hwai Lin
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital Hsinchu Branch, Hsinchu County, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shinn-Jang Hwang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Family Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Hsiao-Ting Chang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
Ungvari A, Nyúl-Tóth Á, Patai R, Csik B, Gulej R, Nagy D, Shanmugarama S, Benyó Z, Kiss T, Ungvari Z, Csiszar A. Cerebromicrovascular senescence in vascular cognitive impairment: does accelerated microvascular aging accompany atherosclerosis? GeroScience 2025:10.1007/s11357-025-01621-w. [PMID: 40113668 DOI: 10.1007/s11357-025-01621-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Vascular cognitive impairment (VCI) is a leading cause of age-related cognitive decline, driven by cerebrovascular dysfunction and cerebral small vessel disease (CSVD). Emerging evidence suggests that cerebromicrovascular endothelial senescence plays an important role in the pathogenesis of VCI by promoting cerebral blood flow dysregulation, neurovascular uncoupling, blood-brain barrier (BBB) disruption, and the development of cerebral microhemorrhages (CMHs). This review explores the concept of cerebromicrovascular senescence as a continuum of vascular aging, linking macrovascular atherosclerosis with microvascular dysfunction. It examines the mechanisms by which endothelial senescence drives neurovascular pathology and highlights the impact of cardiovascular risk factors in accelerating these processes. We examine preclinical and clinical studies that provide compelling evidence that atherosclerosis-induced microvascular senescence exacerbates cognitive impairment. In particular, findings suggest that targeting senescent endothelial cells through senolytic therapy can restore cerebrovascular function and improve cognitive outcomes in experimental models of atherosclerosis. Given the growing recognition of microvascular senescence as a therapeutic target, further research is warranted to explore novel interventions such as senolytics, anti-inflammatory agents, and metabolic modulators. The development of circulating biomarkers of vascular senescence (e.g., senescence-associated secretory phenotype [SASP] components and endothelial-derived extracellular vesicles) could enable early detection and risk stratification in individuals at high risk for VCI. Additionally, lifestyle modifications, including the Mediterranean diet, hold promise for delaying endothelial senescence and mitigating cognitive decline. In conclusion, cerebromicrovascular senescence is a key mechanistic link between atherosclerosis and cognitive impairment. Addressing microvascular aging as a modifiable risk factor through targeted interventions offers a promising strategy for reducing the burden of VCI and preserving cognitive function in aging populations.
Collapse
Affiliation(s)
- Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, 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
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- Institute of Preventive Medicine and Public Health, 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
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Dorina Nagy
- 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
| | - Santny Shanmugarama
- 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
| | - Tamas Kiss
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
- Pediatric Center, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- 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
| | - 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
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Singh Solorzano C, Festari C, Mirabelli P, Mombelli E, Coppola L, Luongo D, Naviglio D, Soricelli A, Quattrini G, Salvatore M, Pievani M, Cattaneo A, Frisoni GB, Marizzoni M. Association between cognitive functioning and microbiota-gut-brain axis mediators in a memory clinic population. Front Cell Neurosci 2025; 19:1550333. [PMID: 40144018 PMCID: PMC11936893 DOI: 10.3389/fncel.2025.1550333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
IntroductionA growing body of evidence recognises the role of signaling molecule of the microbiota-gut-brain axis (MGBA) in cognitive impairment (CI), but data on the link with alterations in specific cognitive domains are limited. We compared the functioning in several cognitive domains (i.e., memory, visuo-constructional, executive, and language) among cognitively unimpaired (CU) subjects, patients with CI due to Alzheimer’s disease (CI-AD) and not due to AD (CI-NAD). Then, we investigated the association of these cognitive domains with the gut microbiota (GM), MGBA mediators, and neurodegeneration-related markers.Materials and methodsThe study included 34 CI-AD, 38 CI-NAD, and 13 CU. Memory, visuo-constructional, executive, and language domains were assessed using composite measures. Faecal GM composition was inferred using 16S rRNA gene sequencing. MGBA mediators included the blood quantification of bacterial products (lipolysaccharide, LPS), cell adhesion molecules indicative of endothelial damage, vascular changes or overexpressed in response to infections, and pro- and anti-inflammatory cytokines. Neurodegeneration-related markers included plasma phosphorylated tau (p-tau181), neurofilament light chain (NfL), and glial fibrillary protein (GFAP).ResultsThe CI-NAD and CI-AD groups had significantly lower scores than the CU group for all cognitive domains (p < 0.043). Associations of MGBA modulators with cognitive functioning included pro-inflammatory cytokines, markers of endothelial dysfunction or overexpressed in response to infection in both groups of patients (|ρ| > 0.33, ps < 0.042). In the CU and CI-AD pooled group, lower cognitive functioning was specifically associated with higher abundance of Dialister and Clostridia_UCG-014, higher levels of LPS and with all neurodegeneration markers (|ρ| > 0.32, p < 0.048 for all). In the CU and CI-NAD pooled group, lower cognitive performance was associated with lower abundance of Acetonema, higher abundance of Bifidobacterium, [Eubacterium]_coprostanoligenes_group and Collinsella, and higher levels of vascular changes (|ρ| > 0.30, p < 0.049).DiscussionThese results support the hypothesis that gut dysbiosis and MGBA mediators may have distinct effects on cognitive functioning and different mechanisms of action depending on the disease.
Collapse
Affiliation(s)
- Claudio Singh Solorzano
- Laboratory of Neuroimaging and Alzheimer’s Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Cristina Festari
- Laboratory of Neuroimaging and Alzheimer’s Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Peppino Mirabelli
- AORN Santobono-Pausilipon, UOS Laboratori di Ricerca e Biobanca, Naples, Italy
| | - Elisa Mombelli
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Delia Luongo
- Istituto Di Biostrutture E Bioimmagini (I.B.B.) - CNR, Naples, Italy
| | - Daniele Naviglio
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Andrea Soricelli
- IRCCS SYNLAB SDN, Naples, Italy
- Department of Medical, Movement and Wellbeing Sciences, University of Naples Parthenope, Naples, Italy
| | - Giulia Quattrini
- Laboratory of Neuroimaging and Alzheimer’s Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Michela Pievani
- Laboratory of Neuroimaging and Alzheimer’s Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Giovanni B. Frisoni
- Memory Centre, Division of Geriatrics and Rehabilitation, University Hospitals of Geneva, Geneva, Switzerland
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
6
|
Breidenbach BM, Driscoll I, Glittenberg MP, Paulsen AJ, Fernandes-Taylor S, Naren T, Roberts GS, Brach TL, Jarchow MM, Symanski LE, Gaul AY, Lose SR, Rivera-Rivera LA, Johnson SC, Asthana S, Christian BT, Cook DB, Wieben O, Okonkwo OC. Cardiorespiratory Fitness Modifies the Relationship Between Arterial Stiffness and Cerebral Blood Flow Independent of Physical Activity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323254. [PMID: 40093262 PMCID: PMC11908333 DOI: 10.1101/2025.03.03.25323254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Central arterial stiffness and cerebral blood flow (CBF) are inversely related. Poor cardiorespiratory fitness (CRF) and low physical activity (PA) are related to both higher arterial stiffness and lower CBF. The present study examined (i) whether CRF or PA moderate the relationship between arterial stiffness and CBF and (ii) whether the intensity or the type of PA need to be considered. METHODS Participants (N=78, Mean AGE =64.2±6.14, 72% female) from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center were categorized into low, average and high fitness groups based on maximal graded exercise treadmill test performance. PA was assessed using the CHAMPS questionnaire. Based on hours/week, participants were classified as meeting the recommended 2.5 hours of moderate intensity PA per week (PA Rec Met). Weekly hours of moderate and low intensity PA were calculated as activities of > 3 or < 3 metabolic equivalents, respectively. Activity type was categorized as exercise-, sports/leisure- and work-related. Arterial stiffness was measured as aortic pulse wave velocity (aoPWV) by 2D phase contrast MRI. CBF was assessed by 4D flow MRI in the internal carotid arteries (ICAs), cavernous ICAs, middle cerebral arteries (MCAs), and via two composite measures of total and global flow. RESULTS The association between aoPWV and CBF differed by fitness levels, with a negative relationship in the low fitness group and positive relationships in the average and high fitness groups (all P s<0.05). Significant moderating effects on the relationships between aoPWV and CBF were also observed for PA Rec Met (all P s<0.05), moderate intensity ( P =0.05) and exercise-related (all P s<0.02) PA. DISCUSSION Average or high fitness, meeting the PA guidelines, and more specifically, moderate intensity and exercise-related PA seem to attenuate the negative relationship between aoPWV on CBF.
Collapse
|
7
|
de Rezende VL, de Aguiar da Costa M, Martins CD, Mathias K, Gonçalves CL, Barichello T, Petronilho F. Systemic Rejuvenating Interventions: Perspectives on Neuroinflammation and Blood-Brain Barrier Integrity. Neurochem Res 2025; 50:112. [PMID: 40035979 DOI: 10.1007/s11064-025-04361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
The aging process results in structural, functional, and immunological changes in the brain, which contribute to cognitive decline and increase vulnerability to neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and stroke-related complications. Aging leads to cognitive changes and also affect executive functions. Additionally, it causes neurogenic and neurochemical alterations, such as a decline in dopamine and acetylcholine levels, which also impact cognitive performance. The chronic inflammation caused by aging contributes to the impairment of the blood-brain barrier (BBB), contributing to the infiltration of immune cells and exacerbating neuronal damage. Therefore, rejuvenating therapies such as heterochronic parabiosis, cerebrospinal fluid (CSF) administration, plasma, platelet-rich plasma (PRP), and stem cell therapy have shown potential to reverse these changes, offering new perspectives in the treatment of age-related neurological diseases. This review focuses on highlighting the effects of rejuvenating interventions on neuroinflammation and the BBB.
Collapse
Affiliation(s)
- Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Carla Damasio Martins
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
- Laboratory of Immunoparasitology, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, Mcgovern Medical School, The University of Texas Health Science Center at Houston (Uthealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil.
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
8
|
Bravi B, Verga C, Palladini M, Poletti S, Buticchi C, Stefania S, Stefano D, Colombo C, Comai S, Benedetti F. Effects of kynurenine pathway metabolites on choroid plexus volume, hemodynamic response, and spontaneous neural activity: A new mechanism for disrupted neurovascular communication and impaired cognition in mood disorders. Brain Behav Immun 2025; 125:414-427. [PMID: 39909168 DOI: 10.1016/j.bbi.2025.01.025] [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: 10/15/2024] [Revised: 01/07/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025] Open
Abstract
Major Depressive Disorder (MDD) and Bipolar Disorder (BD) involve alterations of immune-inflammatory setpoints that activate the kynurenine pathway (KP), affecting serotoninergic and glutamatergic neurotransmission through indoleamine-2,3-dioxygenase (IDO) activity. This process produces metabolites like Kynurenine (Kyn), 3-Hydroxykynurenine (3-HK), Quinolinic acid (QuinA), and Kynurenic acid (KynA), these last two acting as agonist and antagonist at glutamatergic N-methyl-D-aspartate receptors (NMDARs), respectively. NMDARs, expressed in the choroid plexus (ChP) and arteriolar smooth muscle cells, regulate blood-brain-barrier permeability and cerebral artery dilation, suggesting that KP may influence neurovascular coupling, aligning blood flow with neural energy demand. KP's role in modulating vascular tone supports this hypothesis. Altered fractional amplitude of low-frequency fluctuations (fALFF) and disrupted default mode network (DMN) activity in mood disorders are linked to cognitive deficits possibly through neurovascular uncoupling like in neurological diseases. This makes fALFF and hemodynamic response function (HRF) potential indicators of these changes. We investigated KP associations with ChP volumes, functional-MRI at rest measures like spontaneous neural activity (fALFF) and hemodynamic response function (HRF) parameters within the default mode network (DMN), and cognitive performance in 42 MDD and 36 BD inpatients experiencing a depressive episode. Results revealed that lower QuinA/KynA ratios and higher KynA levels predict larger ChP volumes. Higher KYN and 3-HK levels, along with lower KynA levels, were associated with increased DMN fALFF and shorter time-to-peak (TTP) in HRF, suggesting altered neurovascular coupling. Mediation analyses indicated that KP metabolites influenced cognitive performance through their effects on resting state measures, affecting global cognitive functioning score, verbal fluency, and psychomotor coordination. These findings suggest that KP metabolites modulate brain function and structure via NMDAR-mediated pathways and vascular-based mechanisms, offering insights into the cognitive impairments observed in mood disorders and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Beatrice Bravi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Chiara Verga
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Mariagrazia Palladini
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Camilla Buticchi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sut Stefania
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Dall'Acqua Stefano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milan, Italy; Mood Disorder Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
9
|
Lambert M, Miquel G, Villeneuve L, Thorin-Trescases N, Thorin E. The senolytic ABT-263 improves cognitive functions in middle-aged male, but not female, atherosclerotic LDLr -/-;hApoB 100+/+ mice. GeroScience 2025:10.1007/s11357-025-01563-3. [PMID: 39982668 DOI: 10.1007/s11357-025-01563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Accumulation of cerebral senescent cells may compromise the continuum between vascular and neuronal function, leading to damage and cognitive decline. Elimination of senescent cells might therefore preserve vascular and neuronal functions. To test this hypothesis, we used male and female atherosclerotic LDLr-/-;hApoB100+/+ mice (ATX-mice), a model of vascular cognitive impairment (VCI), treated with the senolytic ABT-263 for 3 months (3- to 6-month or 9- to 12-month old). In young male ATX mice, prevention with ABT-263 improved spatial retention memory, in association with a higher endothelial sensitivity to shear stress and a higher hippocampal CD31+ endothelial cell density, lower activation of both astrocytes and glial cells. In young females, ABT-263 tended to improve delayed memory; however, atherosclerotic plaque was magnified by ABT-263, endothelial function was unaffected, hippocampal astrocyte activation increased and expression of CD31+ cells decreased. Hence, unlike in males, ABT-263 appears deleterious in young ATX females. In middle-aged males, the curative treatment improved the learning process and memory. Although no change in endothelial function was observed, the benefits of ABT-263 were associated with a decreased expression of several inflammaging markers, a higher density of CD31+ cells and a lower activation of glial cells. In middle-aged females, ABT-263 induced a surge of inflammaging markers, associated with a slower learning process. Altogether, our data demonstrate that ABT-263 differentially affects VCI, improving cognition in male while being deleterious in female ATX mice. More studies are needed to understand the mechanisms at the basis of the sexual dimorphic effects of the senolytic ABT-263.
Collapse
Affiliation(s)
- Mélanie Lambert
- Faculty of Medicine, Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada.
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada.
| | - Géraldine Miquel
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
| | | | - Eric Thorin
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
- Faculty of Medicine, Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Patai R, Csik B, Nyul-Toth A, Gulej R, Vali Kordestan K, Chandragiri SS, Shanmugarama S, Tarantini S, Mukli P, Ungvari A, Yabluchanskiy A, Ungvari Z, Csiszar A. Persisting blood-brain barrier disruption following cisplatin treatment in a mouse model of chemotherapy-associated cognitive impairment. GeroScience 2025:10.1007/s11357-025-01569-x. [PMID: 39982666 DOI: 10.1007/s11357-025-01569-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Chemotherapy-related cognitive impairment, commonly referred to as "chemobrain," significantly affects cancer survivors' quality of life, yet its underlying mechanisms remain unclear. Most chemotherapeutic agents cannot cross the blood-brain barrier (BBB), yet they cause central nervous system side effects, suggesting alternative pathways of toxicity. Given that these drugs interact with the cerebrovascular endothelium at their highest concentrations, it is logical to hypothesize that endothelial damage contributes to these effects. Our recent studies demonstrated that paclitaxel-induced cognitive impairment in a mouse model results in a partial BBB disruption and subsequent neuroinflammation, mediated by chemotherapy-induced endothelial senescence. In this pilot study, we used two-photon microscopy to assess BBB permeability in mice receiving a clinically relevant cisplatin regimen, evaluating the leakage of fluorescent dextran tracers of varying molecular weights. Two months post-treatment, cisplatin-treated mice exhibited significantly increased BBB permeability to smaller molecular tracers (40 kDa, 3 kDa, and 0.3 kDa) compared to controls, indicating sustained BBB disruption. These results align with our findings for paclitaxel and suggest that chemotherapy-induced endothelial damage and senescence play a central role in cognitive impairments. Interventions targeting endothelial health could mitigate these long-term effects, improving cognitive outcomes for cancer survivors.
Collapse
Affiliation(s)
- Roland Patai
- 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, 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
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Kiana Vali Kordestan
- 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
| | - Siva Sai Chandragiri
- 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
| | - Santny Shanmugarama
- 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
| | - 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, 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
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
11
|
Patai R, Kiss T, Gulej R, Nyul-Toth A, Csik B, Chandragiri SS, Shanmugarama S, Tarantini S, Ungvari A, Pacher P, Mukli P, Yabluchanskiy A, Csiszar A, Ungvari Z. Transcriptomic profiling of senescence effects on blood–brain barrier-related gene expression in brain capillary endothelial cells in a mouse model of paclitaxel-induced chemobrain. GeroScience 2025. [DOI: 10.1007/s11357-025-01561-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/07/2025] [Indexed: 03/04/2025] Open
Abstract
Abstract
Chemotherapy-induced cognitive impairment (CICI), commonly referred to as “chemobrain,” is a frequent and debilitating side effect experienced by cancer survivors treated with paclitaxel (PTX). Preclinical models have shown that PTX promotes cerebromicrovascular endothelial cell senescence, leading to chronic blood–brain barrier (BBB) disruption and neuroinflammation. Conversely, the elimination of senescent cells through senolytic therapies has been shown to restore BBB integrity, reduce neuroinflammation, and alleviate PTX-induced cognitive impairment. In this study, we tested the hypothesis that PTX-induced endothelial senescence alters gene expression patterns associated with BBB integrity. To investigate this, we analyzed a scRNA-seq dataset from the brains of mice treated with a clinically relevant PTX regimen alongside vehicle-treated control mice. We identified capillary endothelial cells by their distinct transcriptomic profiles and matched these profiles to known transcriptomic markers of cellular senescence. Our analysis confirmed that PTX induces senescence in capillary endothelial cells and revealed significant transcriptional alterations linked to impaired BBB function. In senescent endothelial cells, gene set enrichment analysis (GSEA) highlighted downregulated pathways associated with cell junction assembly and upregulated pathways involved in extracellular matrix remodeling and inflammatory signaling, including Vitronectin (VTN) and Pleiotrophin (PTN) pathways. Additionally, cell–cell communication analysis revealed reduced Junctional Adhesion Molecule (JAM) signaling, further implicating senescence in BBB disruption. These findings highlight endothelial senescence as a driver of BBB dysfunction through transcriptional changes and altered intercellular signaling. The enrichment of VTN and PTN pathways in the senescent state indicates a shift toward vascular remodeling and inflammation, exacerbating microvascular fragility and BBB disruption. Supported by prior experimental findings, this study suggests that targeting endothelial senescence and its downstream effects could mitigate PTX-induced BBB dysfunction and associated cognitive impairments. These results advance our understanding of CICI pathogenesis and provide a foundation for developing therapeutic strategies aimed at preserving vascular integrity.
Collapse
|
12
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
13
|
Freeberg KA, McCarty NP, Chonchol M, Seals DR, Craighead DH. Oxidative stress suppresses internal carotid artery dilation to hypercapnia in healthy older adults. J Appl Physiol (1985) 2025; 138:536-545. [PMID: 39819040 DOI: 10.1152/japplphysiol.00322.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/23/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
Cerebrovascular disease and dementia risk increases with age, and lifetime risk is greater in women. Cerebrovascular dysfunction likely precedes cerebrovascular disease and dementia but the mechanisms are incompletely understood. We hypothesized that oxidative stress mediates cerebrovascular dysfunction with human aging. Internal carotid artery dilation (ICACO2 dilation) and middle cerebral artery cerebrovascular reactivity (MCA CVRCO2) in response to hypercapnia (5% CO2) were measured in 20 young [10 F/10 M; age 23 ± 3 yr (means ± SD)] and 21 older (11 F/10 M; age 69 ± 9 yr) adults during intravenous infusions of saline (control) and vitamin C (acutely reduced oxidative stress condition). ICACO2 dilation increased in response to vitamin C infusion in older adults (saline = 4.3 ± 2.4%; vitamin C = 6.7 ± 3.3%) but was unchanged in young adults (saline = 6.1 ± 2.7%; vitamin C = 5.5 ± 1.9%) (group × condition: P = 0.004). MCA CVRCO2 was not different in response to vitamin C in either group (group × condition: P = 0.341). However, when separated by sex, older female participants exhibited increased MCA CVRCO2 with vitamin C (saline = 0.85 ± 0.79 cm/s/mmHg; vitamin C = 1.33 ± 1.01 cm/s/mmHg) compared with older male participants (saline = 1.21 ± 0.57 cm/s/mmHg; vitamin C = 0.99 ± 0.47 cm/s/mmHg) (sex × condition: P = 0.011). Oxidative stress selectively impairs cerebrovascular function in older adults in an artery- and sex-specific manner.NEW & NOTEWORTHY This study is the first to report oxidative stress-mediated suppression of cerebrovascular reactivity to hypercapnia in the internal carotid artery in older compared with young adults. Overall, these in vivo findings identify oxidative stress as an important pathophysiological contributor to cerebrovascular aging in humans, highlighting the need to identify novel interventions that can reduce oxidative stress in the aging population.
Collapse
Affiliation(s)
- Kaitlin A Freeberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Narissa P McCarty
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Daniel H Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
14
|
Hooijmans MT, Jeneson JA, Jørstad HT, Bakermans AJ. Exercise MR of Skeletal Muscles, the Heart, and the Brain. J Magn Reson Imaging 2025; 61:535-560. [PMID: 38726984 PMCID: PMC11706321 DOI: 10.1002/jmri.29445] [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: 02/21/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 01/11/2025] Open
Abstract
Magnetic resonance (MR) imaging (MRI) is routinely used to evaluate organ morphology and pathology in the human body at rest or in combination with pharmacological stress as an exercise surrogate. With MR during actual physical exercise, we can assess functional characteristics of tissues and organs under real-life stress conditions. This is particularly relevant in patients with limited exercise capacity or exercise intolerance, and where complaints typically present only during physical activity, such as in neuromuscular disorders, inherited metabolic diseases, and heart failure. This review describes practical and physiological aspects of exercise MR of skeletal muscles, the heart, and the brain. The acute effects of physical exercise on these organs are addressed in the light of various dynamic quantitative MR readouts, including phosphorus-31 MR spectroscopy (31P-MRS) of tissue energy metabolism, phase-contrast MRI of blood flow and muscle contraction, real-time cine MRI of cardiac performance, and arterial spin labeling MRI of muscle and brain perfusion. Exercise MR will help advancing our understanding of underlying mechanisms that contribute to exercise intolerance, which often proceed structural and anatomical changes in disease. Its potential to detect disease-driven alterations in organ function, perfusion, and metabolism under physiological stress renders exercise MR stress testing a powerful noninvasive imaging modality to aid in disease diagnosis and risk stratification. Although not yet integrated in most clinical workflows, and while some applications still require thorough validation, exercise MR has established itself as a comprehensive and versatile modality for characterizing physiology in health and disease in a noninvasive and quantitative way. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Melissa T. Hooijmans
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Human Movement Sciences, Faculty of Behavioral and Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Jeroen A.L. Jeneson
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Center for Child Development and Exercise, Wilhelmina Children's Hospital/Division of Child HealthUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Harald T. Jørstad
- Department of CardiologyAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
| | - Adrianus J. Bakermans
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
15
|
Cao X, Peng H, Hu Z, Xu C, Ning M, Zhou M, Mi Y, Yu P, Fazekas-Pongor V, Major D, Ungvari Z, Fekete M, Lehoczki A, Guo Y. Exploring the global impact of obesity and diet on dementia burden: the role of national policies and sex differences. GeroScience 2025; 47:1345-1360. [PMID: 39612068 PMCID: PMC11872863 DOI: 10.1007/s11357-024-01457-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: 10/10/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024] Open
Abstract
Obesity is a significant modifiable risk factor for dementia. This study aims to quantify the global impact of obesity on dementia burden and examine how national strategies for managing overweight/obesity and dietary factors influence dementia prevalence and mortality, with a focus on sex-specific differences. We used data from the Global Burden of Disease (GBD) and World Health Organization (WHO) to evaluate the association between obesity age-standardized prevalence rate (ASPR) and dementia age-standardized mortality rate (ASMR) and ASPR across 161 countries. A two-step multivariate analysis adjusted for socioeconomic and lifestyle factors was performed. Temporal trends in dementia were analyzed based on the presence of national obesity management strategies and varying dietary scores. A 1% increase in national obesity prevalence was associated with a 0.36% increase in dementia mortality (OR: 1.0036; 95% CI: 1.0028-1.0045) in males and 0.12% in females (OR: 1.0012; 95% CI: 1.0007-1.0018). A 1% increase in national obesity ASPR was associated with an increase in ASPR of dementia by 0.26% for males (OR: 1.0026, 95% CI: 1.0024-1.0028) and 0.05% for females (OR: 1.0005, 95% CI: 1.0004-1.0006). Males exhibited a higher susceptibility to obesity-related dementia. Countries with national obesity management strategies showed a significantly greater reduction in dementia mortality, particularly among females (P = 0.025). Higher dietary scores were associated with a more significant decrease in dementia prevalence across both sexes. Rising obesity prevalence is linked to increased dementia burden globally, with males being more vulnerable to this relationship. National management of overweight/obesity and healthier dietary habits may help mitigate the dementia burden, emphasizing the need for integrated public health interventions.
Collapse
Affiliation(s)
- Xueshan Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huiyuan Peng
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziyi Hu
- Mingde Innovation Class, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chang Xu
- Mingde Innovation Class, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Monan Ning
- Mingde Innovation Class, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mengge Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yuanqi Mi
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Peixin Yu
- School of Arts and Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Vince Fazekas-Pongor
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - David Major
- Institute of Preventive Medicine and Public Health, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Yang Guo
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
16
|
Gáspár Z, Szabó BG, Ceglédi A, Lakatos B. Human herpesvirus reactivation and its potential role in the pathogenesis of post-acute sequelae of SARS-CoV-2 infection. GeroScience 2025; 47:167-187. [PMID: 39207648 PMCID: PMC11872864 DOI: 10.1007/s11357-024-01323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
The emergence of SARS-CoV-2 has precipitated a global pandemic with substantial long-term health implications, including the condition known as post-acute sequelae of SARS-CoV-2 infection (PASC), commonly referred to as Long COVID. PASC is marked by persistent symptoms such as fatigue, neurological issues, and autonomic dysfunction that persist for months beyond the acute phase of COVID-19. This review examines the potential role of herpesvirus reactivation, specifically Epstein-Barr virus (EBV) and cytomegalovirus (CMV), in the pathogenesis of PASC. Elevated antibody titers and specific T cell responses suggest recent herpesvirus reactivation in some PASC patients, although viremia is not consistently detected. SARS-CoV-2 exhibits endothelial trophism, directly affecting the vascular endothelium and contributing to microvascular pathologies. These pathologies are significant in PASC, where microvascular dysfunction may underlie various chronic symptoms. Similarly, herpesviruses like CMV also exhibit endothelial trophism, which may exacerbate endothelial damage when reactivated. Evidence suggests that EBV and CMV reactivation could indirectly contribute to the immune dysregulation, immunosenescence, and autoimmune responses observed in PASC. Additionally, EBV may play a role in the genesis of neurological symptoms through creating mitochondrial dysfunction, though direct confirmation remains elusive. The reviewed evidence suggests that while herpesviruses may not play a direct role in the pathogenesis of PASC, their potential indirect effects, especially in the context of endothelial involvement, warrant further investigation.
Collapse
Affiliation(s)
- Zsófia Gáspár
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Bálint Gergely Szabó
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary.
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
| | - Andrea Ceglédi
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Botond Lakatos
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| |
Collapse
|
17
|
Ungvari A, Gulej R, Patai R, Papp Z, Toth A, Szabó AÁ, Podesser BK, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Maier AB, Csiszar A, Ungvari Z. Sex-specific mechanisms in vascular aging: exploring cellular and molecular pathways in the pathogenesis of age-related cardiovascular and cerebrovascular diseases. GeroScience 2025; 47:301-337. [PMID: 39754010 PMCID: PMC11872871 DOI: 10.1007/s11357-024-01489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/17/2024] [Indexed: 03/04/2025] Open
Abstract
Aging remains the foremost risk factor for cardiovascular and cerebrovascular diseases, surpassing traditional factors in epidemiological significance. This review elucidates the cellular and molecular mechanisms underlying vascular aging, with an emphasis on sex differences that influence disease progression and clinical outcomes in older adults. We discuss the convergence of aging processes at the macro- and microvascular levels and their contributions to the pathogenesis of vascular diseases. Critical analysis of both preclinical and clinical studies reveals significant sex-specific variations in these mechanisms, which could be pivotal in understanding the disparity in disease morbidity and mortality between sexes. The review highlights key molecular pathways, including oxidative stress, inflammation, and autophagy, and their differential roles in the vascular aging of males and females. We argue that recognizing these sex-specific differences is crucial for developing targeted therapeutic strategies aimed at preventing and managing age-related vascular pathologies. The implications for personalized medicine and potential areas for future research are also explored, emphasizing the need for a nuanced approach to the study and treatment of vascular aging.
Collapse
Affiliation(s)
- Anna Ungvari
- Institute of Preventive Medicine and Public Health, 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
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Á Szabó
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, 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
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK, USA
- Reynolds Section of Geriatrics and Palliative Medicine, Department of Medicine, University of Oklahoma Health Sciences, Oklahoma City, OK, 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
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- @AgeSingapore, Healthy Longevity Program, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
18
|
Qiu Y, Cheng L, Xiong Y, Liu Z, Shen C, Wang L, Lu Y, Wei S, Zhang L, Yang SB, Zhang X. Advances in the Study of Necroptosis in Vascular Dementia: Focus on Blood-Brain Barrier and Neuroinflammation. CNS Neurosci Ther 2025; 31:e70224. [PMID: 39915907 PMCID: PMC11802338 DOI: 10.1111/cns.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Vascular dementia (VaD) includes a group of brain disorders that are characterized by cerebrovascular pathology.Neuroinflammation, disruption of the blood-brain barrier (BBB) permeability, white matter lesions, and neuronal loss are all significant pathological manifestations of VaD and play a key role in disease progression. Necroptosis, also known asprogrammed necrosis, is a mode of programmed cell death distinct from apoptosis and is closely associated with ischemic injury and neurodegenerative diseases. Recent studies have shown that necroptosis in VaD exacerbates BBB destruction, activates neuroinflammation, promotes neuronal loss, and severely affects VaD prognosis. RESULTS AND CONCLUSIONS In this review, we outline the significant roles of necroptosis and its molecular mechanisms in the pathological process of VaD, with a particular focus on the role of necroptosis in modulating neuroinflammation and exacerbating the disruption of BBB permeability in VaD, and elaborate on the molecular regulatory mechanisms and the centrally involved cells of necroptosis mediated by tumor necrosis factor-α in neuroinflammation in VaD. We also analyze the possibility and specific strategy that targeting necroptosis would help inhibit neuroinflammation and BBB destruction in VaD. With a focus on necroptosis, this study delved into its impact on the pathological changes and prognosis of VaD to provide new treatment ideas.
Collapse
Affiliation(s)
- Yuemin Qiu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Lin Cheng
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Yinyi Xiong
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of RehabilitationAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Ziying Liu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Chunxiao Shen
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Liangliang Wang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Yujia Lu
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Shufei Wei
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Lushun Zhang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Seung Bum Yang
- Department of Medical Non‐Commissioned OfficerWonkwang Health Science UniversityIksanRepublic of Korea
| | - Xiaorong Zhang
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Department of PathologyJiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
19
|
Fu W, Ma Z, Mei B, Yan Y, Peng J, Hou J, Yang Z, Zhang Y, Li Z, Gong X. Predicting progression of cerebral small vessel disease: relevance of carotid perivascular fat density based on computed tomography angiography. Quant Imaging Med Surg 2025; 15:1582-1598. [PMID: 39995701 PMCID: PMC11847194 DOI: 10.21037/qims-24-1512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/31/2024] [Indexed: 02/26/2025]
Abstract
Background Symptomatic carotid lesions surrounded by perivascular fat have been found to be associated with the presence of cerebral small vessel disease (CSVD). In this study, we investigated the possible relationship of perivascular fat density (PFD) with CSVD and its progression, independent of the presence of carotid stenosis. Methods This study retrospectively evaluated consecutive patients without carotid stenosis who underwent carotid computed tomography angiography (CTA), computed tomography perfusion (CTP), and two brain magnetic resonance imaging (MRI) scans at Zhejiang Provincial People's Hospital (hospital I) from January 2019 to March 2024. Patients were categorized into three groups: without CSVD (n=34), with CSVD without progression (n=83), and with CSVD progression (n=146) according to MRI markers of CSVD. Additionally, 65 patients (including 22 with CSVD without progression and 43 with CSVD progression) were collected from Hangzhou Traditional Chinese Medicine Hospital (hospital II) for external validation. PFD was quantified using a dedicated software. The association between perfusion status on CTP and CSVD was assessed. The associations of PFD and imaging markers with the progression of CSVD were also analyzed. Six models based on PFD, significant clinical factors, and radiomic signatures were developed and validated to predict the CSVD progression. Results PFD values were positively associated with lacunes, cerebral microbleeds (CMBs), and white matter hyperintensities (WMH) (all P<0.05). In addition, patients with CSVD progression had higher PFD than those without [-51.38±7.35 vs. -57.19±7.31 Hounsfield unit (HU); P<0.001]. Multivariate analysis indicated that diabetes, coronary artery disease, PFD, and radiomic signatures were independent predictors of CSVD progression. Moreover, the hybrid model showed enhanced performance and yielded the highest area under the receiver operating characteristic curve (AUC) of the receiver operator characteristic curve [training: AUC =0.818, 95% confidence interval (CI): 0.758-0.876; internal validation: AUC =0.805, 95% CI: 0.690-0.908; external validation: AUC =0.807, 95% CI: 0.676-0.921]. Conclusions This study showed that, in participants without carotid stenosis, PFD was predictive of CSVD progression, suggesting the possible involvement of the inflammation present in perivascular fat in the pathogenesis of CSVD.
Collapse
Affiliation(s)
- Wanyun Fu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Zhangman Ma
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bozhe Mei
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College), Hangzhou, China
| | - Yuting Yan
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jiaxuan Peng
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jie Hou
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | | | - Yongsheng Zhang
- Department of Radiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiping Li
- Department of Radiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangyang Gong
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
- Institute of Artificial Intelligence and Remote Imaging, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
20
|
Stankovics L, Ungvari A, Fekete M, Nyul-Toth A, Mukli P, Patai R, Csik B, Gulej R, Conley S, Csiszar A, Toth P. The vasoprotective role of IGF-1 signaling in the cerebral microcirculation: prevention of cerebral microhemorrhages in aging. GeroScience 2025; 47:445-455. [PMID: 39271571 PMCID: PMC11872839 DOI: 10.1007/s11357-024-01343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Aging is closely associated with various cerebrovascular pathologies that significantly impact brain function, with cerebral small vessel disease (CSVD) being a major contributor to cognitive decline in the elderly. Consequences of CSVD include cerebral microhemorrhages (CMH), which are small intracerebral bleeds resulting from the rupture of microvessels. CMHs are prevalent in aging populations, affecting approximately 50% of individuals over 80, and are linked to increased risks of vascular cognitive impairment and dementia (VCID). Hypertension is a primary risk factor for CMHs. Vascular smooth muscle cells (VSMCs) adapt to hypertension by undergoing hypertrophy and producing extracellular matrix (ECM) components, which reinforce vessel walls. Myogenic autoregulation, which involves pressure-induced constriction, helps prevent excessive pressure from damaging the vulnerable microvasculature. However, aging impairs these adaptive mechanisms, weakening vessel walls and increasing susceptibility to damage. Insulin-like Growth Factor 1 (IGF-1) is crucial for vascular health, promoting VSMC hypertrophy, ECM production, and maintaining normal myogenic protection. IGF-1 also prevents microvascular senescence, reduces reactive oxygen species (ROS) production, and regulates matrix metalloproteinase (MMP) activity, which is vital for ECM remodeling and stabilization. IGF-1 deficiency, common in aging, compromises these protective mechanisms, increasing the risk of CMHs. This review explores the vasoprotective role of IGF-1 signaling in the cerebral microcirculation and its implications for preventing hypertension-induced CMHs in aging. Understanding and addressing the decline in IGF-1 signaling with age are crucial for maintaining cerebrovascular health and preventing hypertension-related vascular injuries in the aging population.
Collapse
Affiliation(s)
- Levente Stankovics
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Boglarka Csik
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Department of Cell Biology, 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
| | - Peter Toth
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
21
|
Fekete M, Lehoczki A, Szappanos Á, Toth A, Mahdi M, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Ungvari Z. Cerebromicrovascular mechanisms contributing to long COVID: implications for neurocognitive health. GeroScience 2025; 47:745-779. [PMID: 39777702 PMCID: PMC11872997 DOI: 10.1007/s11357-024-01487-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long COVID (also known as post-acute sequelae of SARS-CoV-2 infection [PASC] or post-COVID syndrome) is characterized by persistent symptoms that extend beyond the acute phase of SARS-CoV-2 infection, affecting approximately 10% to over 30% of those infected. It presents a significant clinical challenge, notably due to pronounced neurocognitive symptoms such as brain fog. The mechanisms underlying these effects are multifactorial, with mounting evidence pointing to a central role of cerebromicrovascular dysfunction. This review investigates key pathophysiological mechanisms contributing to cerebrovascular dysfunction in long COVID and their impacts on brain health. We discuss how endothelial tropism of SARS-CoV-2 and direct vascular infection trigger endothelial dysfunction, impaired neurovascular coupling, and blood-brain barrier disruption, resulting in compromised cerebral perfusion. Furthermore, the infection appears to induce mitochondrial dysfunction, enhancing oxidative stress and inflammation within cerebral endothelial cells. Autoantibody formation following infection also potentially exacerbates neurovascular injury, contributing to chronic vascular inflammation and ongoing blood-brain barrier compromise. These factors collectively contribute to the emergence of white matter hyperintensities, promote amyloid pathology, and may accelerate neurodegenerative processes, including Alzheimer's disease. This review also emphasizes the critical role of advanced imaging techniques in assessing cerebromicrovascular health and the need for targeted interventions to address these cerebrovascular complications. A deeper understanding of the cerebrovascular mechanisms of long COVID is essential to advance targeted treatments and mitigate its long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| | - Ágnes Szappanos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032, Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, 4031, Debrecen, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 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
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
22
|
Alok K, White TG, Li C. Bioelectronic medicine in modulation of cortical spreading depolarization and beyond. Neural Regen Res 2025; 20:481-482. [PMID: 38819057 PMCID: PMC11317948 DOI: 10.4103/nrr.nrr-d-23-02059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/08/2024] [Accepted: 03/19/2024] [Indexed: 06/01/2024] Open
Affiliation(s)
- Khaled Alok
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Timothy G. White
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Chunyan Li
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| |
Collapse
|
23
|
Negri S, Reyff Z, Troyano-Rodriguez E, Milan M, Ihuoma J, Tavakol S, Shi H, Patai R, Jiang R, Mohon J, Boma-Iyaye J, Ungvari Z, Csiszar A, Yabluchanskiy A, Moccia F, Tarantini S. Endothelial Colony-Forming Cells (ECFCs) in cerebrovascular aging: Focus on the pathogenesis of Vascular Cognitive Impairment and Dementia (VCID), and treatment prospects. Ageing Res Rev 2025; 104:102672. [PMID: 39884362 DOI: 10.1016/j.arr.2025.102672] [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: 11/01/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Endothelial colony-forming cells (ECFCs), a unique endothelial progenitor subset, are essential for vascular integrity and repair, providing significant regenerative potential. Recent studies highlight their role in cerebrovascular aging, particularly in the pathogenesis of vascular cognitive impairment and dementia (VCID). Aging disrupts ECFC functionality through mechanisms such as oxidative stress, chronic inflammation, and cellular senescence, leading to compromised vascular repair and reduced neurovascular resilience. ECFCs influence key cerebrovascular processes, including neurovascular coupling (NVC), blood-brain barrier (BBB) integrity, and vascular regeneration, which are critical for cognitive health. Age-related decline in ECFC quantity and functionality contributes to vascular rarefaction, diminished cerebral blood flow (CBF), and BBB permeability-processes that collectively exacerbate cognitive decline. This review delves into the multifaceted role of ECFCs in cerebrovascular aging and underscores their potential as therapeutic targets in addressing age-related vascular dysfunctions, presenting new directions for mitigating the effects of aging on brain health.
Collapse
Affiliation(s)
- Sharon Negri
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zeke Reyff
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eva Troyano-Rodriguez
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madison Milan
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer Ihuoma
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sherwin Tavakol
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Helen Shi
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Raymond Jiang
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Casady School, Oklahoma City, OK, USA
| | - Jonah Mohon
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Jed Boma-Iyaye
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Francesco Moccia
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso 86100, Italy
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
24
|
Fekete M, Liotta EM, Molnar T, Fülöp GA, Lehoczki A. The role of atrial fibrillation in vascular cognitive impairment and dementia: epidemiology, pathophysiology, and preventive strategies. GeroScience 2025; 47:287-300. [PMID: 39138793 PMCID: PMC11872872 DOI: 10.1007/s11357-024-01290-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
The aging population in Europe faces a substantial burden from dementia, with vascular cognitive impairment and dementia (VCID) being a preventable cause. Atrial fibrillation (AF), a common cardiac arrhythmia, increases the risk of VCID through mechanisms such as thromboembolism, cerebral hypoperfusion, and inflammation. This review explores the epidemiology, pathophysiology, and preventive strategies for AF-related VCID. Epidemiological data indicate that AF prevalence rises with age, affecting up to 12% of individuals over 80. Neuroimaging studies reveal chronic brain changes in AF patients, including strokes, lacunar strokes, white matter hyperintensities (WMHs), and cerebral microbleeds (CMHs), while cognitive assessments show impairments in memory, executive function, and attention. The COVID-19 pandemic has exacerbated the underdiagnosis of AF, leading to an increase in undiagnosed strokes and cognitive impairment. Many elderly individuals did not seek medical care due to fear of exposure, resulting in delayed diagnoses. Additionally, reduced family supervision during the pandemic contributed to missed opportunities for early detection of AF and related complications. Emerging evidence suggests that long COVID may also elevate the risk of AF, further complicating the management of this condition. This review underscores the importance of early detection and comprehensive management of AF to mitigate cognitive decline. Preventive measures, including public awareness campaigns, patient education, and the use of smart devices for early detection, are crucial. Anticoagulation therapy, rate and rhythm control, and addressing comorbid conditions are essential therapeutic strategies. Recognizing and addressing the cardiovascular and cognitive impacts of AF, especially in the context of the COVID-19 pandemic, is essential for advancing public health.
Collapse
Affiliation(s)
- Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Eric M Liotta
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| | - Gábor A Fülöp
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
25
|
Godos J, Giampieri F, Frias-Toral E, Zambrano-Villacres R, Vistorte AOR, Yélamos Torres V, Battino M, Galvano F, Castellano S, Grosso G. Nut Consumption Is Associated with Cognitive Status in Southern Italian Adults. Nutrients 2025; 17:521. [PMID: 39940379 PMCID: PMC11820291 DOI: 10.3390/nu17030521] [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: 12/28/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Nut consumption has been considered a potential protective factor against cognitive decline. The aim of this study was to test whether higher total and specific nut intake was associated with better cognitive status in a sample of older Italian adults. METHODS A cross-sectional analysis on 883 older adults (>50 y) was conducted. A 110-item food frequency questionnaire was used to collect information on the consumption of various types of nuts. The Short Portable Mental Status Questionnaire was used to assess cognitive status. Multivariate logistic regression analyses were performed to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between nut intake and cognitive status after adjusting for potential confounding factors. RESULTS The median intake of total nuts was 11.7 g/day and served as a cut-off to categorize low and high consumers (mean intake 4.3 g/day vs. 39.7 g/day, respectively). Higher total nut intake was significantly associated with a lower prevalence of impaired cognitive status among older individuals (OR = 0.35, CI 95%: 0.15, 0.84) after adjusting for potential confounding factors. Notably, this association remained significant after additional adjustment for adherence to the Mediterranean dietary pattern as an indicator of diet quality, (OR = 0.32, CI 95%: 0.13, 0.77). No significant associations were found between cognitive status and specific types of nuts. CONCLUSIONS Habitual nut intake is associated with better cognitive status in older adults.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Francesca Giampieri
- Department of Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
| | - Evelyn Frias-Toral
- School of Medicine, Universidad Católica de Santiago de Guayaquil, Av. Pdte. Carlos Julio Arosemena Tola, Guayaquil 090615, Ecuador
| | | | - Angel Olider Rojas Vistorte
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidad Internacional Iberoamericana, Arecibo, PR 00613, USA
- Universidad Internacional do Cuanza, Cuito EN250, Bié, Angola
| | - Vanessa Yélamos Torres
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidad Internacional Iberoamericana, Campeche 24560, México
- Universidad de La Romana, La Romana 22000, Dominican Republic
| | - Maurizio Battino
- Department of Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang 212013, China
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| |
Collapse
|
26
|
Bhattarai P, Yilmaz E, Cakir EÖ, Korkmaz HY, Lee AJ, Ma Y, Celikkaya H, Cosacak MI, Haage V, Wang X, Nelson N, Lin W, Zhang Y, Nuriel T, Jülich D, Iş Ö, Holley SA, de Jager P, Fisher E, Tubbesing K, Teich AF, Bertucci T, Temple S, Ertekin-Taner N, Vardarajan BN, Mayeux R, Kizil C. APOE- ε4-induced Fibronectin at the blood-brain barrier is a conserved pathological mediator of disrupted astrocyte-endothelia interaction in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634732. [PMID: 39975303 PMCID: PMC11838230 DOI: 10.1101/2025.01.24.634732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Blood-brain barrier (BBB) dysfunction is a key feature of Alzheimer's disease (AD), particularly in individuals carrying the APOE-ε4 allele. This dysfunction worsens neuroinflammation and hinders the removal of toxic proteins, such as amyloid-beta (Aβ42), from the brain. In post-mortem brain tissues and in animal models, we previously reported that fibronectin accumulates at the BBB predominantly in APOE-ε4 carriers. Furthermore, we found a loss-of-function variant in the fibronectin 1 ( FN1 ) gene significantly reduces aggregated fibronectin levels and decreases AD risk among APOE-ε4 carriers. Yet, the molecular mechanisms downstream of fibronectin at the BBB remain unclear. The extracellular matrix (ECM) plays a crucial role in maintaining BBB homeostasis and orchestrating the interactions between BBB cell types, including endothelia and astrocytes. Understanding the mechanisms affecting the ECM and BBB cell types will be critical for developing effective therapies against AD, especially among APOE-ε4 carriers. Here, we demonstrate that APOE-ε4 , Aβ42, and inflammation drive the induction of FN1 expression in several models including zebrafish, mice, iPSC-derived human 3D astrocyte and 3D cerebrovascular cell cultures, and in human brains. Fibronectin accumulation disrupts astroglial-endothelial interactions and the signalling cascade between vascular endothelial growth factor (VEGF), heparin-binding epidermal growth factor (HBEGF) and Insulin-like growth factor 1 (IGF1). This accumulation of fibronectin in APOE-ε4- associated AD potentiates BBB dysfunction, which strongly implicates reducing fibronectin deposition as a potential therapeutic target for AD. Graphical abstract Accessibility text This image illustrates the effects of different APOE isoforms (ApoE-ε3 and ApoE-ε4) on blood-brain barrier (BBB) integrity, focusing on the molecular interactions between astrocytes and endothelial cells. This figure emphasizes the detrimental effects of ApoE-ε4 on BBB integrity via fibronectin accumulation and altered signaling pathways. The top section provides a schematic overview of the blood-brain barrier, highlighting astrocytes, endothelial cells, and their interface. The left panel represents the ApoE-ε3 condition: Normal fibronectin (FN1) levels support healthy interactions between astrocytes and endothelial cells. Growth factors, including VEGFA, HBEGF, and IGF1, maintain BBB integrity through their respective receptors (VEGFR and EGFR). Green arrows indicate activation of these signaling pathways. The right panel depicts the ApoE-ε4 condition: Elevated fibronectin (FN1) disrupts astrocyte-endothelium interactions. FN1 binds integrins and activates focal adhesion kinase (FAK), inhibiting VEGFA, which is required for endothelial HBEGF that in turn activates IGF1 signaling. Red symbols indicate inhibition of HBEGF, VEGFA, and IGF1 pathways, leading to BBB dysfunction. Highlights APOE-ε4 drives fibronectin deposition in Alzheimer's, disrupting astrocyte-endothelia interactions. APOE-ε4 and fibronectin co-localize, forming aggregates at blood-brain barrier (BBB). Fibronectin alters the signaling between VEGF, IGF1, and HBEGF impairing BBB function. Reducing fibronectin restores BBB integrity and offsets APOE-ε4 pathology.
Collapse
|
27
|
Fekete M, Varga P, Ungvari Z, Fekete JT, Buda A, Szappanos Á, Lehoczki A, Mózes N, Grosso G, Godos J, Menyhart O, Munkácsy G, Tarantini S, Yabluchanskiy A, Ungvari A, Győrffy B. The role of the Mediterranean diet in reducing the risk of cognitive impairement, dementia, and Alzheimer's disease: a meta-analysis. GeroScience 2025:10.1007/s11357-024-01488-3. [PMID: 39797935 DOI: 10.1007/s11357-024-01488-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
Age-related cognitive impairment and dementia pose a significant global health, social, and economic challenge. While Alzheimer's disease (AD) has historically been viewed as the leading cause of dementia, recent evidence reveals the considerable impact of vascular cognitive impairment and dementia (VCID), which now accounts for nearly half of all dementia cases. The Mediterranean diet-characterized by high consumption of fruits, vegetables, whole grains, fish, and olive oil-has been widely recognized for its cardiovascular benefits and may also reduce the risk of cognitive decline and dementia. To investigate the protective effects of the Mediterranean diet on cognitive health, we conducted a systematic literature review using PubMed, Web of Science, and Google Scholar, focusing on studies published between 2000 and 2024. The studies included in the meta-nalysis examined the adherence to the Mediterranean diet and the incidence of dementia and AD. We applied a random-effects model to calculate pooled hazard ratios (HRs) with 95% confidence intervals (CIs) and assessed heterogeneity through I-square statistics. Forest plots, funnel plots, and Z-score plots were used to visualize study outcomes. Of the 324 full-text records reviewed, 23 studies met the inclusion criteria. The combined HR for cognitive impairment among those adhering to the Mediterranean diet was 0.82 (95% CI 0.75-0.89); for dementia, the HR was 0.89 (95% CI 0.83-0.95); and for AD, the HR was 0.70 (95% CI 0.60-0.82), indicating substantial protective effects. Significant heterogeneity was observed across studies, though Z-score plots suggested sufficient sample sizes to support reliable conclusions for each condition. In conclusion, this meta-analysis confirms that adherence to the Mediterranean diet is associated with an 11-30% reduction in the risk of age-related cognitive disorders, including cognitive impairment, dementia, and AD. These findings underscore the Mediterranean diet's potential as a central element in neuroprotective public health strategies to mitigate the global impact of cognitive decline and dementia and to promote healthier cognitive aging.
Collapse
Affiliation(s)
- Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Péter Varga
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - János Tibor Fekete
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Annamaria Buda
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Ágnes Szappanos
- Department of Vascular and Endovascular Surgery, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Noémi Mózes
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Otilia Menyhart
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Gyöngyi Munkácsy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, 7624, Pecs, Hungary
| |
Collapse
|
28
|
Ungvari Z, Fekete M, Varga P, Fekete JT, Buda A, Szappanos Á, Lehoczki A, Mózes N, Grosso G, Menyhart O, Munkácsy G, Tarantini S, Yabluchanskiy A, Ungvari A, Győrffy B. Impact of adherence to the Mediterranean diet on stroke risk. GeroScience 2025:10.1007/s11357-024-01491-8. [PMID: 39777701 DOI: 10.1007/s11357-024-01491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Stroke is a leading cause of morbidity and mortality worldwide, and dietary patterns have emerged as a significant modifiable factor in stroke prevention. The Mediterranean diet, characterized by high intake of fruits, vegetables, whole grains, nuts, olive oil, and fish, has been widely recognized for its cardiovascular benefits. However, its specific impact on stroke risk requires further elucidation. We conducted a comprehensive meta-analysis of 30 studies, including both cohort and case-control designs, to evaluate the relationship between adherence to the Mediterranean diet and the risk of stroke. A systematic search was performed across multiple databases, and a random-effects model was used to estimate pooled hazard ratios (HRs) with 95% confidence intervals (CIs). Heterogeneity was assessed using the I2 statistic, and publication bias was examined through funnel plots and Egger's regression test. Additionally, trial sequential analysis was conducted to determine the adequacy of the sample size. The meta-analysis revealed a significant reduction in stroke risk among individuals adhering to the Mediterranean diet, with a pooled HR of 0.88 (95% CI: 0.84-0.91). Notably, a significant heterogeneity was detected (I2 = 34%). The Z-score plot from trial sequential analysis confirmed that the sample sizes were sufficient to draw definitive conclusions. However, a potential publication bias was identified. The case-control studies confirmed a highly significant effect (HR = 0.54, 95% CI: 0.4-0.73). The funnel plots in both settings hinted at the presence of a potential publication bias, supported by a significant Egger's test. Our findings provide robust evidence supporting the protective effect of the Mediterranean diet against stroke. Despite the presence of some heterogeneity and potential publication bias, the cumulative evidence suggests that promoting the Mediterranean diet could serve as an effective public health strategy for stroke prevention. Further research is recommended to explore the underlying mechanisms and to assess the diet's impact across diverse populations.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Péter Varga
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - János Tibor Fekete
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Annamaria Buda
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Ágnes Szappanos
- Department of Vascular and Endovascular Surgery, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Noémi Mózes
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Otilia Menyhart
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Gyöngyi Munkácsy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, 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
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, 7624, Pecs, Hungary
| |
Collapse
|
29
|
Downs TL, Whiteside EJ, Denham J, Mills DE, Bliss ES. Exercise as a Therapeutic Strategy to Improve Cerebrovascular Function and Cognition in Breast Cancer Survivors: A Scoping Review. J Clin Med 2024; 13:7841. [PMID: 39768764 PMCID: PMC11678850 DOI: 10.3390/jcm13247841] [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: 11/05/2024] [Revised: 12/07/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Breast cancer is the most diagnosed cancer globally. While the breast cancer prevalence continues to rise, so too do patient survival rates, thus resulting in a large survivor population. Up to 75% of this population report experiencing cancer-related cognitive impairment during their cancer journey, thus reducing their quality of survivorship. This review systematically evaluates the effect of physical activity and exercise training on cerebrovascular function and cognition in breast cancer survivors. Cross-sectional, intervention or observational studies that examined the effect of acute or chronic exercise training or physical activity levels on cerebrovascular function and cognition in female breast cancer survivors were searched for systematically. The 11 included studies were tabulated and described narratively. The included studies primarily focused on aerobic exercise training, while only four studies investigated the effect of resistance exercise training or concurrent training on cerebrovascular function and/or cognition in breast cancer survivors. Collectively, these studies provide preliminary evidence supporting the positive effect of exercise training on cerebrovascular function and cognition in breast cancer survivors, irrespective of their age, stage of breast cancer and treatment regimen. However, more research is required to comprehensively evaluate the effect of exercise training on cerebrovascular function and cognition in breast cancer survivors and the mechanisms leading to these potential improvements.
Collapse
Affiliation(s)
- Tahnee L. Downs
- School of Health and Medical Sciences, University of Southern Queensland, Toowoomba, QLD 4350, Australia
- Respiratory and Exercise Physiology Research Group, University of Southern Queensland, Ipswich, QLD 4305, Australia
| | - Eliza J. Whiteside
- School of Health and Medical Sciences, University of Southern Queensland, Toowoomba, QLD 4350, Australia
- Centre for Health Research, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Future Materials, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Joshua Denham
- School of Health and Medical Sciences, University of Southern Queensland, Toowoomba, QLD 4350, Australia
- Respiratory and Exercise Physiology Research Group, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Health Research, University of Southern Queensland, Ipswich, QLD 4305, Australia
| | - Dean E. Mills
- School of Health and Medical Sciences, University of Southern Queensland, Toowoomba, QLD 4350, Australia
- Respiratory and Exercise Physiology Research Group, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Health Research, University of Southern Queensland, Ipswich, QLD 4305, Australia
| | - Edward S. Bliss
- School of Health and Medical Sciences, University of Southern Queensland, Toowoomba, QLD 4350, Australia
- Respiratory and Exercise Physiology Research Group, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Health Research, University of Southern Queensland, Ipswich, QLD 4305, Australia
| |
Collapse
|
30
|
Sultan AA, Karthikeyan S, Grigorian A, Kennedy KG, Mio M, MacIntosh BJ, Goldstein BI. Cerebral blood flow in relation to peripheral endothelial function in youth bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111087. [PMID: 39004332 DOI: 10.1016/j.pnpbp.2024.111087] [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: 02/29/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Anomalous cerebral blood flow (CBF) is evident in bipolar disorder (BD), however the extent to which CBF reflects peripheral vascular function in BD is unknown. This study investigated endothelial function, an index of early atherosclerosis and cardiovascular disease risk, in relation to CBF among youth with BD. METHODS Participants included 113 youth, 13-20 years old (66 BD; 47 healthy controls [HC]). CBF was measured using arterial spin labeling with 3T MRI. Region of interest analyses (ROI; global grey matter, middle frontal gyrus, anterior cingulate cortex, temporal cortex, caudate) were undertaken alongside voxel-wise analyses. Reactive hyperemia index (RHI), a measure of endothelial function, was assessed non-invasively via pulse amplitude tonometry. General linear models were used to examine RHI and RHI-by-diagnosis associations with CBF, controlling for age, sex, and body mass index. Bonferroni correction for multiple comparisons was used for ROI analyses, such that the significance level was divided by the number of ROIs (α = 0.05/5 = 0.01). Cluster-extent thresholding was used to correct for multiple comparisons for voxel-wise analyses. RESULTS ROI findings were not significant after correction. Voxel-wise analyses found that higher RHI was associated with lower left thalamus CBF in the whole group (p < 0.001). Additionally, significant RHI-by-diagnosis associations with CBF were found in three clusters: left intracalcarine cortex (p < 0.001), left thalamus (p < 0.001), and right frontal pole (p = 0.006). Post-hoc analyses showed that in each cluster, higher RHI was associated with lower CBF in BD, but higher CBF in HC. CONCLUSION We found that RHI was differentially associated with CBF in youth with BD versus HC. The unanticipated association of higher RHI with lower CBF in BD could potentially reflect a compensatory mechanism. Future research, including prospective studies and experimental designs are warranted to build on the current findings.
Collapse
Affiliation(s)
- Alysha A Sultan
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sudhir Karthikeyan
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Anahit Grigorian
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Megan Mio
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Bradley J MacIntosh
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences, Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Harmon JN, Chandran P, Chandrasekaran A, Hyde JE, Hernandez GJ, Reed MJ, Bruce MF, Khaing ZZ. Contrast-Enhanced Ultrasound Imaging Detects Anatomical and Functional Changes in Rat Cervical Spine Microvasculature With Normal Aging. J Gerontol A Biol Sci Med Sci 2024; 80:glae215. [PMID: 39188137 PMCID: PMC11701746 DOI: 10.1093/gerona/glae215] [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: 03/01/2024] [Indexed: 08/28/2024] Open
Abstract
Normal aging is associated with significant deleterious cerebrovascular changes; these have been implicated in disease pathogenesis and increased susceptibility to ischemic injury. Although these changes are well documented in the brain, few studies have been conducted in the spinal cord. Here, we utilize specialized contrast-enhanced ultrasound (CEUS) imaging to investigate age-related changes in cervical spinal vascular anatomy and hemodynamics in male Fisher 344 rats, a common strain in aging research. Aged rats (24-26 months, N = 6) exhibited significant tortuosity in the anterior spinal artery and elevated vascular resistance compared to adults (4-6 months, N = 6; tortuosity index 2.20 ± 0.15 vs 4.74 ± 0.45, p < .05). Baseline blood volume was lower in both larger vessels and the microcirculation in the aged cohort, specifically in white matter (4.44e14 ± 1.37e13 vs 3.66e14 ± 2.64e13 CEUS bolus area under the curve, p < .05). To elucidate functional differences, animals were exposed to a hypoxia challenge, whereas adult rats exhibited significant functional hyperemia in both gray matter (GM) and white matter (WM) (GM: 1.13 ± 0.10-fold change from normoxia, p < .05; WM: 1.16 ± 0.13, p < .05), aged rats showed no response. Immunohistochemistry revealed reduced pericyte coverage and activated microglia behavior in aged rats, which may partially explain the lack of vascular response. This study provides the first in vivo description of age-related hemodynamic differences in the cervical spinal cord.
Collapse
Affiliation(s)
- Jennifer N Harmon
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - Preeja Chandran
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | | | - Jeffrey E Hyde
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - Gustavo J Hernandez
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - May J Reed
- Department of Gerontology and Geriatric Medicine, University of Washington, Seattle, Washington, USA
| | - Matthew F Bruce
- Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | - Zin Z Khaing
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
32
|
da C. Pinaffi-Langley AC, Szarvas Z, Peterfi A, Kaposzta Z, Mukli P, Shahriari A, Muranyi M, Pinto CB, Owens CD, Adams C, Karfonta B, Rohan M, Tarantini S, Yabluchanskiy A. Time-restricted eating for prevention of age-related vascular cognitive decline in older adults: A protocol for a single-arm open-label interventional trial. PLoS One 2024; 19:e0314871. [PMID: 39652561 PMCID: PMC11627372 DOI: 10.1371/journal.pone.0314871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024] Open
Abstract
Age-related cerebromicrovascular endothelial dysfunction underlies the initiation and progression of cognitive dysfunction and dementia, thus increasing the susceptibility of older adults to such conditions. Normal brain function requires dynamic adjustment of cerebral blood flow to meet the energetic demands of active neurons, which is achieved the homeostatic mechanism neurovascular coupling (NVC). In this context, therapeutical strategies aimed at rescuing or preserving NVC responses can delay the incidence or mitigate the severity of age-related cognitive dysfunction, and time-restricted eating (TRE) is a potential candidate for such a strategy. Studies have reported that TRE can improve cardiometabolic risk factors in older adults. However, the effect of TRE on cerebrovascular endothelial function remains unexplored. Thus, this protocol outlines the study procedures to test our hypothesis that a 6-month TRE regimen of 10-h eating window will improve NVC responses and endothelial function in community-dwelling older adults. This is a single-arm, open-label interventional trial. We aim to recruit 32 adults aged 55-80 years. Participants are instructed to maintain a TRE regimen of 10 h of free eating followed by 14 h of fasting for 6 months. Before and after fasting, participants are assessed for cognitive performance, peripheral micro- and macrovascular endothelial function, and NVC responses, as well as for several confounding factors, including body composition, dietary, and physical activity data. We expect that 6 months of TRE will improve NVC response and endothelial function in older adults compared with baseline, and that these improvements will be accompanied by improvements in cognitive performance. The study proposed herein will provide critical insight into a new potential therapeutical strategy for targeting age-related cognitive dysfunction. Ultimately, slowing down or alleviating cognitive decline will translate into improved quality of life and longer healthspan for aging adults. This study was prospectively registered at ClinicalTrials.gov (NCT06019195) on August 24, 2023.
Collapse
Affiliation(s)
- Ana Clara da C. Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Zalan Kaposzta
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Ali Shahriari
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Mihaly Muranyi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Camila B. Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Cheryl Adams
- Oklahoma Shared Clinical and Translational Resources, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Brittany Karfonta
- Oklahoma Shared Clinical and Translational Resources, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Michael Rohan
- Laureate Institute for Brain Research, Tulsa, OK, United States of America
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, United States of America
| |
Collapse
|
33
|
Russell SJ, Parker K, Lehoczki A, Lieberman D, Partha IS, Scott SJ, Phillips LR, Fain MJ, Nikolich JŽ. Post-acute sequelae of SARS-CoV-2 infection (Long COVID) in older adults. GeroScience 2024; 46:6563-6581. [PMID: 38874693 PMCID: PMC11493926 DOI: 10.1007/s11357-024-01227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
Long COVID, also known as PASC (post-acute sequelae of SARS-CoV-2), is a complex infection-associated chronic condition affecting tens of millions of people worldwide. Many aspects of this condition are incompletely understood. Among them is how this condition may manifest itself in older adults and how it might impact the older population. Here, we briefly review the current understanding of PASC in the adult population and examine what is known on its features with aging. Finally, we outline the major gaps and areas for research most germane to older adults.
Collapse
Affiliation(s)
- Samantha J Russell
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Karen Parker
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - David Lieberman
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Indu S Partha
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Serena J Scott
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Linda R Phillips
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- College of Nursing, University of Arizona, Tucson, AZ, USA
| | - Mindy J Fain
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Banner University Medicine-Tucson, Tucson, AZ, USA.
- College of Nursing, University of Arizona, Tucson, AZ, USA.
| | - Janko Ž Nikolich
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
34
|
Sanchez-Martinez Y, Lopez-Lopez JP, Gomez-Montoya I, Hernandez-Quiñones D, Ruiz-Uribe G, Rincón-Rueda Z, Garcia RG, Lopez-Jaramillo P. Muscular strength, endothelial function and cognitive disorders: state of the art. J Physiol 2024. [PMID: 39612371 DOI: 10.1113/jp285939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 12/01/2024] Open
Abstract
In recent years, the ageing population has increasingly grown. This process carries a range of pathophysiological changes involving alterations in the skeletal muscle, vascular endothelium and brain function, becoming an important risk factor for developing cognitive disorders and cardiovascular diseases. With ageing, there is a decrease in muscle mass and muscle strength, and a relationship between muscle strength decrease and cognitive decline has been shown. Lower handgrip strength has been linked to memory impairment, lower global cognitive function, decreased attention and reduced visuospatial abilities in the elderly, but understanding of the underlying mechanisms that explain the link between altered skeletal muscle function and structure, endothelial dysfunction, and the role of endothelial dysfunction in the onset of cognitive disorders has been scarcely explored. This review aims to detail the cellular and molecular mechanisms by which the progressive changes associated with ageing can alter healthy skeletal muscle and endothelial function, creating an environment of oxidative stress, inflammation and mitochondrial dysfunction. These changes can lead to reduced muscle strength, and the secretion of detrimental endothelial factors, resulting in endothelial dysfunction, blood-brain barrier disruption, and damage to neurons and microglia, ultimately accelerating the onset of cognitive disorders in the elderly. In addition, we aimed to describe the mechanisms that potentially explain how preserving muscular function with resistance training could prevent brain function deterioration, including the production of different factors that allow an improved endothelial function, haemodynamic parameters and brain plasticity, ultimately delaying the onset of cognitive impairment and chronic diseases.
Collapse
Affiliation(s)
| | - Jose P Lopez-Lopez
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | | | | | - Gabriela Ruiz-Uribe
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | - Zully Rincón-Rueda
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | - Ronald G Garcia
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Patricio Lopez-Jaramillo
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
- Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
35
|
Olejnik P, Golenia A. Vascular Cognitive Impairment-The Molecular Basis and Potential Influence of the Gut Microbiota on the Pathological Process. Cells 2024; 13:1962. [PMID: 39682711 PMCID: PMC11639845 DOI: 10.3390/cells13231962] [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: 11/03/2024] [Revised: 11/18/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Cognitive impairment is a major healthcare challenge worldwide, with vascular cognitive impairment (VCI) being its second leading cause after Alzheimer's disease. VCI is a heterogeneous group of cognitive disorders resulting from various vascular pathologies. Therefore, it is particularly difficult to determine its underlying cause and exact molecular basis. Nevertheless, the current understanding of the pathophysiological processes underlying VCI has changed and evolved in the last decades. The aim of this narrative review is to summarize the current state of knowledge on VCI pathogenesis and to analyze the potential role of the gut microbiota in this process, considering the most recent scientific reports and in accordance with the current understanding of these processes. Chronic cerebral hypoperfusion, which results in impaired blood supply, i.e., oxygen and nutrient deficiency, is the main underlying mechanism of VCI. Furthermore, chronic cerebral hypoperfusion triggers a cascade of molecular changes, starting with an energy imbalance, leading to glutamate excitotoxicity, acidotoxicity, and oxidative stress. Also, all of the above provoke the activation of microglia and the release of pro-inflammatory cytokines that recruit systemic immune cells and lead to their infiltration into the central nervous system, resulting in neuroinflammation. Blood-brain barrier dysfunction may occur at various stages of chronic cerebral hypoperfusion, ultimately increasing its permeability and allowing potentially toxic substances to enter the brain parenchyma. Gut microbiota and their metabolites, which have been identified in numerous inflammatory conditions, may also influence the pathophysiological processes of VCI.
Collapse
Affiliation(s)
- Piotr Olejnik
- Department of Neurology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | | |
Collapse
|
36
|
Lauriola M, Esposito L, D’Onofrio G, Ciccone F, la Torre A, Addante F, Cocomazzi A, Cascavilla L, Ariano O, Serviddio G, Greco A. Risk of Stroke or Heart Attack in Mild Cognitive Impairment and Subjective Cognitive Impairment. Neurol Int 2024; 16:1528-1539. [PMID: 39585072 PMCID: PMC11587450 DOI: 10.3390/neurolint16060113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The study aimed to identify Mild Cognitive Impairment (MCI) as an alert clinical manifestation of increased probability of major acute vascular events (MVEs), such as Ischemic Stroke and heart attack. METHODS In a longitudinal study, 181 (M = 81, F = 100; mean age of 75.8 ± 8.69 years) patients were enrolled and divided into three groups based on diagnosis: Subjective Cognitive Impairment (SCI), amnestic MCI Single Domain (aMCI-SD), and amnestic MCI More Domain (aMCI-MD). Clinical assessment and the presence of vascular risk factors were collected. RESULTS The distribution of MVEs showed a higher incidence in the first two years of follow-up of 7.4% in SCI, 12.17% in aMCI-SD, and 8.57% in aMCI-MD. Acute Myocardial Infarction showed a major incidence in one year of follow-up (41%) and in two years of follow-up (29%). Also, Ischemic Stroke showed a major incidence in one year of follow-up (30%) and in two years of follow-up (40%). A statistically significant difference in the progression to dementia was shown (SCI 3.75%; aMCI-SD 10.43%; aMCI-MD 37%; p-value < 0.001). CONCLUSIONS MCI is considered an expression of the systemic activation of mechanisms of endothelial damage, representing a diagnosis predictive of increased risk of MVEs.
Collapse
Affiliation(s)
- Michele Lauriola
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| | - Luigi Esposito
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| | - Grazia D’Onofrio
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Filomena Ciccone
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Annamaria la Torre
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Filomena Addante
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| | - Annagrazia Cocomazzi
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| | - Leandro Cascavilla
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| | - Olga Ariano
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (O.A.); (G.S.)
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (O.A.); (G.S.)
| | - Antonio Greco
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (M.L.); (L.E.); (F.A.); (A.C.); (L.C.); (A.G.)
| |
Collapse
|
37
|
Alateeq K, Walsh EI, Cherbuin N. High Blood Pressure and Impaired Brain Health: Investigating the Neuroprotective Potential of Magnesium. Int J Mol Sci 2024; 25:11859. [PMID: 39595928 PMCID: PMC11594239 DOI: 10.3390/ijms252211859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
High blood pressure (BP) is a significant contributor to the disease burden globally and is emerging as an important cause of morbidity and mortality in the young as well as the old. The well-established impact of high BP on neurodegeneration, cognitive impairment, and dementia is widely acknowledged. However, the influence of BP across its full range remains unclear. This review aims to explore in more detail the effects of BP levels on neurodegeneration, cognitive function, and dementia. Moreover, given the pressing need to identify strategies to reduce BP levels, particular attention is placed on reviewing the role of magnesium (Mg) in ageing and its capacity to lower BP levels, and therefore potentially promote brain health. Overall, the review aims to provide a comprehensive synthesis of the evidence linking BP, Mg and brain health. It is hoped that these insights will inform the development of cost-effective and scalable interventions to protect brain health in the ageing population.
Collapse
Affiliation(s)
- Khawlah Alateeq
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
- Radiological Science, College of Applied Medical Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Erin I. Walsh
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
| | - Nicolas Cherbuin
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
| |
Collapse
|
38
|
Callewaert B, Gsell W, Lox M, Himmelreich U, Jones EAV. A timeline study on vascular co-morbidity induced cerebral endothelial dysfunction assessed by perfusion MRI. Neurobiol Dis 2024; 202:106709. [PMID: 39433136 DOI: 10.1016/j.nbd.2024.106709] [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: 12/12/2023] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Endothelial dysfunction is considered a key element in the early pathogenesis of neurodegenerative disorders. Dysfunction of the cerebral endothelial cells can result in dysregulation of cerebral perfusion and disruption of the Blood Brain Barrier (BBB), leading to brain damage, neurodegeneration and cognitive decline. It has been shown that the presence of modifiable risk factors exacerbates endothelial dysfunction. This study primarily aimed to identify which among various perfusion MRI methodologies could be effectively utilized to non-invasively identify early pathological alterations as a result of endothelial dysfunction. We compared these perfusion MRI measurements to invasive immunohistochemistry to detect early pathological alterations in the cerebral vasculature of a rat model of multiple cardiovascular co-morbidities (the ZSF1 Obese rat) at several stages of the cerebrovascular pathology. We observed cerebral hyperperfusion, expressed by increased Cerebral Blood Flow (CBF) and increased BBB permeability in the ZSF1 Obese rats, at an early stage of disease development. The increase in CBF observed with Arterial Spin Labeling (ASL) was lost during later stages of disease progression. These findings are in line with recent clinical findings in early stages of Alzheimer's disease (AD), that also show early increases in CBF.
Collapse
Affiliation(s)
- Bram Callewaert
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Marleen Lox
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; School for Cardiovascular Diseases (CARIM), Department of Cardiology, Maastricht University, 6200 Maastricht, the Netherlands.
| |
Collapse
|
39
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
40
|
Lim XR, Abd-Alhaseeb MM, Ippolito M, Koide M, Senatore AJ, Plante C, Hariharan A, Weir N, Longden TA, Laprade KA, Stafford JM, Ziemens D, Schwaninger M, Wenzel J, Postnov DD, Harraz OF. Endothelial Piezo1 channel mediates mechano-feedback control of brain blood flow. Nat Commun 2024; 15:8686. [PMID: 39375369 PMCID: PMC11458797 DOI: 10.1038/s41467-024-52969-0] [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: 03/04/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Hyperemia in response to neural activity is essential for brain health. A hyperemic response delivers O2 and nutrients, clears metabolic waste, and concomitantly exposes cerebrovascular endothelial cells to hemodynamic forces. While neurovascular research has primarily centered on the front end of hyperemia-neuronal activity-to-vascular response-the mechanical consequences of hyperemia have gone largely unexplored. Piezo1 is an endothelial mechanosensor that senses hyperemia-associated forces. Using genetic mouse models and pharmacologic approaches to manipulate endothelial Piezo1 function, we evaluated its role in blood flow control and whether it impacts cognition. We provide evidence of a built-in brake system that sculpts hyperemia, and specifically show that Piezo1 activation triggers a mechano-feedback system that promotes blood flow recovery to baseline. Further, genetic Piezo1 modification led to deficits in complementary memory tasks. Collectively, our findings establish a role for endothelial Piezo1 in cerebral blood flow regulation and a role in its behavioral sequelae.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Michael Ippolito
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Amanda J Senatore
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Curtis Plante
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Kathryn A Laprade
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - James M Stafford
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Dorothea Ziemens
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Dmitry D Postnov
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, 8200, Denmark
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
41
|
Chen B, Yu X, Horvath-Diano C, Ortuño MJ, Tschöp MH, Jastreboff AM, Schneeberger M. GLP-1 programs the neurovascular landscape. Cell Metab 2024; 36:2173-2189. [PMID: 39357509 DOI: 10.1016/j.cmet.2024.09.003] [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: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Readily available nutrient-rich foods exploit our inherent drive to overconsume, creating an environment of overnutrition. This transformative setting has led to persistent health issues, such as obesity and metabolic syndrome. The development of glucagon-like peptide-1 receptor (GLP-1R) agonists reveals our ability to pharmacologically manage weight and address metabolic conditions. Obesity is directly linked to chronic low-grade inflammation, connecting our metabolic environment to neurodegenerative diseases. GLP-1R agonism in curbing obesity, achieved by impacting appetite and addressing associated metabolic defects, is revealing additional benefits extending beyond weight loss. Whether GLP-1R agonism directly impacts brain health or does so indirectly through improved metabolic health remains to be elucidated. In exploring the intricate connection between obesity and neurological conditions, recent literature suggests that GLP-1R agonism may have the capacity to shape the neurovascular landscape. Thus, GLP-1R agonism emerges as a promising strategy for addressing the complex interplay between metabolic health and cognitive well-being.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Claudia Horvath-Diano
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - María José Ortuño
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
42
|
Supthut W, Nuding S, Wienke A, Müller-Werdan U, Werdan K, Ebelt H. [Relationship between cardiac output, heart rate and microcirculation in patients with multiorgan dysfunction syndrome]. Med Klin Intensivmed Notfmed 2024; 119:538-545. [PMID: 38038767 DOI: 10.1007/s00063-023-01086-9] [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: 06/18/2023] [Revised: 10/15/2023] [Accepted: 10/29/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Multiple organ dysfunction syndrome (MODS) is one of the main causes of death in intensive care units. There is evidence that microcirculation in sepsis and coronary shock is regulated separately from hemodynamics. This study investigates the relationship between heart rate (HR), cardiac output (CO) and microcirculation in patients with MODS. METHODS This is a partial analysis of the "MODIFY study" (Reducing Elevated Heart Rate in Patients With Multiple Organ Dysfunction Syndrome [MODS] by Ivabradine). During the period 05/2010-09/2011, the microcirculation of 46 patients with septic and coronary MODS was measured using the sidestream dark field technique on the day of inclusion and 96 h later. Patients were randomized into a control and ivabradine treatment group. RESULTS Overall, there is a relevant improvement in microcirculation over time small perfused vessels, SPV [%] on day 0, d0:56.5 ± 34.2/d4:73.2 ± 22.1 (p = 0.03); perfused vessel density, PVDsmall [1/mm2] d0:7.5 ± 5.0/d4:9.8 ± 3.4 (p = 0.04); proportion of perfused vessels, PPVsmall [%] d0:51.6 ± 31.6/d4:66.7 ± 21.8 (p = 0.04); microcirculatory flow index, MFI d0:1.7 ± 1.0/d4:2.2 ± 0.7 (p = 0.05). Administration of ivabradine shows no effect. In patients with coronary MODS, there is a relevant correlation between microcirculatory parameters and cardiac output (SPV [%]: r = 0.98, p = 0.004). Patients with coronary MODS show better microcirculation values at high heart rates (> 100 bpm), while patients with septic MODS show an opposite relationship. CONCLUSION The results indicate that in critically ill patients, depending on the genesis of the MODS, there are different relationships between HF or CO values, on the one hand, and the parameters of the microcirculation, on the other.
Collapse
Affiliation(s)
- Wiebke Supthut
- Klinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Deutschland.
| | - Sebastian Nuding
- Medizinische Klinik II, Krankenhaus St. Elisabeth und St. Barbara, Halle (Saale), Deutschland
| | - Andreas Wienke
- Institut für Medizinische Epidemiologie, Biometrie und Informatik, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Deutschland
| | - Ursula Müller-Werdan
- Klinik für Geriatrie und Altersmedizin und EGZB, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Karl Werdan
- Klinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle (Saale), Deutschland
| | - Henning Ebelt
- Klinik für Innere Medizin II, Katholisches Krankenhaus St. Johann Nepomuk, Erfurt, Deutschland
| |
Collapse
|
43
|
Gulej R, Nyúl-Tóth Á, Csik B, Patai R, Petersen B, Negri S, Chandragiri SS, Shanmugarama S, Mukli P, Yabluchanskiy A, Conley S, Huffman D, Tarantini S, Csiszar A, Ungvari Z. Young blood-mediated cerebromicrovascular rejuvenation through heterochronic parabiosis: enhancing blood-brain barrier integrity and capillarization in the aged mouse brain. GeroScience 2024; 46:4415-4442. [PMID: 38727872 PMCID: PMC11336025 DOI: 10.1007/s11357-024-01154-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/05/2024] [Indexed: 06/15/2024] Open
Abstract
Age-related cerebromicrovascular changes, including blood-brain barrier (BBB) disruption and microvascular rarefaction, play a significant role in the development of vascular cognitive impairment (VCI) and neurodegenerative diseases. Utilizing the unique model of heterochronic parabiosis, which involves surgically joining young and old animals, we investigated the influence of systemic factors on these vascular changes. Our study employed heterochronic parabiosis to explore the effects of young and aged systemic environments on cerebromicrovascular aging in mice. We evaluated microvascular density and BBB integrity in parabiotic pairs equipped with chronic cranial windows, using intravital two-photon imaging techniques. Our results indicate that short-term exposure to young systemic factors leads to both functional and structural rejuvenation of cerebral microcirculation. Notably, we observed a marked decrease in capillary density and an increase in BBB permeability to fluorescent tracers in the cortices of aged mice undergoing isochronic parabiosis (20-month-old C57BL/6 mice [A-(A)]; 6 weeks of parabiosis), compared to young isochronic parabionts (6-month-old, [Y-(Y)]). However, aged heterochronic parabionts (A-(Y)) exposed to young blood exhibited a significant increase in cortical capillary density and restoration of BBB integrity. In contrast, young mice exposed to old blood from aged parabionts (Y-(A)) rapidly developed cerebromicrovascular aging traits, evidenced by reduced capillary density and increased BBB permeability. These findings underscore the profound impact of systemic factors in regulating cerebromicrovascular aging. The rejuvenation observed in the endothelium, following exposure to young blood, suggests the existence of anti-geronic elements that counteract microvascular aging. Conversely, pro-geronic factors in aged blood appear to accelerate cerebromicrovascular aging. Further research is needed to assess whether the rejuvenating effects of young blood factors could extend to other age-related cerebromicrovascular pathologies, such as microvascular amyloid deposition and increased microvascular fragility.
Collapse
Affiliation(s)
- Rafal Gulej
- 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
| | - 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, 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
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Benjamin Petersen
- 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
| | - Sharon Negri
- 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
| | - Siva Sai Chandragiri
- 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
| | - Santny Shanmugarama
- 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
| | - 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Shannon Conley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Derek Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, 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.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
44
|
Chen Z, Liu W, Balu N, Chen L, Ortega D, Huang X, Hatsukami TS, Yang J, Yuan C. Associations of Intracranial Artery Length and Branch Number on Time-of-Flight MRA With Cognitive Impairment in Hypertensive Older Males. J Magn Reson Imaging 2024; 60:1720-1728. [PMID: 38263621 DOI: 10.1002/jmri.29242] [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: 11/14/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Hypertension-induced impairment of the cerebral artery network contributes to cognitive impairment. Characterizing the structure and function of cerebral arteries may facilitate the understanding of hypertension-related pathological mechanisms and lead to the development of new indicators for cognitive impairment. PURPOSE To investigate the associations between morphological features of the intracranial arteries distal to the circle of Willis on time-of-flight MRA (TOF-MRA) and cognitive performance in a hypertensive cohort. STUDY TYPE Prospective observational study. POPULATION 189 hypertensive older males (mean age 64.9 ± 7.2 years). FIELD STRENGTH/SEQUENCE TOF-MRA sequence with a 3D spoiled gradient echo readout and arterial spin labeling perfusion imaging sequence with a 3D stack-of-spirals fast spin echo readout at 3T. ASSESSMENT The intracranial arteries were segmented from TOF-MRA and the total length of distal arteries (TLoDA) and number of arterial branches (NoB) were calculated. The mean gray matter cerebral blood flow (GM-CBF) was extracted from arterial spin labeling perfusion imaging. The cognitive level was assessed with short-term and long-term delay-recall auditory verbal learning test (AVLT) scores, and with montreal cognitive assessment. STATISTICAL TESTS Univariable and multivariable linear regression were used to analyze the associations between TLoDA, NoB, GM-CBF and the cognitive assessment scores, with P < 0.05 indicating significance. RESULTS TLoDA (r = 0.314) and NoB (r = 0.346) were significantly correlated with GM-CBF. Multivariable linear regression analyses showed that TLoDA and NoB, but not GM-CBF (P = 0.272 and 0.141), were significantly associated with short-term and long-term delay-recall AVLT scores. These associations remained significant after adjusting for GM-CBF. DATA CONCLUSION The TLoDA and NoB of distal intracranial arteries on TOF-MRA are significantly associated with cognitive impairment in hypertensive subjects. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Zhensen Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Wenjin Liu
- Department of Nephrology, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, China
- Yangzhou Institute of Precision Medicine for Kidney Diseases, Yangzhou, China
| | - Niranjan Balu
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Li Chen
- Department of Electrical and Computer Engineering, University of Washington, Seattle, Washington, USA
| | - Dakota Ortega
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Xiaoqin Huang
- Department of Nephrology, The First People's Hospital of Yancheng, Yancheng, China
| | - Thomas S Hatsukami
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Yuan
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
45
|
Ha JC, Yoon JH. Differential associations of physical job demands with cognitive impairment in Korean workers aged 45 and older: a 14-year longitudinal study using the Korean Longitudinal Study of Aging (KLoSA). BMJ Open 2024; 14:e088000. [PMID: 39353692 PMCID: PMC11448154 DOI: 10.1136/bmjopen-2024-088000] [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] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVES To investigate the association between subelements of physical job demands and cognitive impairment risk in middle-aged and older workers in Korea. DESIGN Longitudinal study using eight waves (2006-2020) of the Korean Longitudinal Study of Aging. SETTING Nationally representative sample of the Korean population aged 45 years and older. PARTICIPANTS 2170 workers aged 45 and older at baseline. PRIMARY OUTCOME MEASURES Cognitive function was evaluated using the Korean version of the Mini-Mental State Examination and cognitive impairment was defined as a score below 24. RESULTS High physical strength demands were inversely associated with cognitive impairment (OR 0.31, 95% CI 0.14 to 0.68 for 'always' vs 'never' category). Conversely, frequent heavy lifting (OR 2.67, 95% CI 1.36 to 5.26) and bending, kneeling or squatting (OR 1.69, 95% CI 0.82 to 3.47) tasks were associated with increased impairment risk. Dose-response relationships were observed between all physical job demands and cognitive impairment, persisting among those with lower education but not among those with higher education. CONCLUSIONS Different types of physical job demands have varying relationships with cognitive impairment in middle-aged and older workers. Tasks requiring high physical strength may protect against cognitive impairment while tasks involving heavy lifting and bending, kneeling or squatting may increase the risk. These findings highlight the need for tailored interventions that consider the type of physical job demands and workers' educational levels to mitigate cognitive impairment risks. Further research is needed to explore the underlying mechanisms and validate these findings.
Collapse
Affiliation(s)
- Jea Chul Ha
- Department of Public Health, Yonsei University Graduate School, Seoul, Republic of Korea
- Occupational and Environmental Medicine, Keimyung University College of Medicine, Daegu, Republic of Korea
| | - Jin-Ha Yoon
- Department of Public Health, Yonsei University Graduate School, Seoul, Republic of Korea
- Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Godos J, Micek A, Currenti W, Franchi C, Poli A, Battino M, Dolci A, Ricci C, Ungvari Z, Grosso G. Fish consumption, cognitive impairment and dementia: an updated dose-response meta-analysis of observational studies. Aging Clin Exp Res 2024; 36:171. [PMID: 39162889 PMCID: PMC11335789 DOI: 10.1007/s40520-024-02823-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/28/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Cognitive impairment is projected to affect a preponderant proportion of the aging population. Lifelong dietary habits have been hypothesized to play a role in preventing cognitive decline. Among the most studied dietary components, fish consumptionhas been extensively studied for its potential effects on the human brain. AIMS To perform a meta-analysis of observational studies exploring the association between fish intake and cognitive impairment/decline and all types of dementia. METHODS A systematic search of electronic databases was performed to identify observational studies providing quantitative data on fish consumption and outcomes of interest. Random effects models for meta-analyses using only extreme exposure categories, subgroup analyses, and dose-response analyses were performed to estimate cumulative risk ratios (RRs) and 95% confidence intervals (CIs). RESULTS The meta-analysis comprised 35 studies. Individuals reporting the highest vs. the lowest fish consumption were associated with a lower likelihood of cognitive impairment/decline (RR = 0.82, 95% CI: 0.75, 0.90, I2 = 61.1%), dementia (RR = 0.82, 95% CI: 0.73, 0.93, I2 = 38.7%), and Alzheimer's disease (RR = 0.80, 95% CI: 0.67, 0.96, I2 = 20.3%). The dose-response relation revealed a significantly decreased risk of cognitive impairment/decline and all cognitive outcomes across higher levels of fish intake up to 30% for 150 g/d (RR = 0.70, 95% CI: 0.52, 0.95). The results of this relation based on APOE ε4 allele status was mixed based on the outcome investigated. CONCLUSIONS Current findings suggest fish consumption is associated with a lower risk of cognitive impairment/decline in a dose-response manner, while for dementia and Alzheimer's disease there is a need for further studies to improve the strength of evidence.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| | - Agnieszka Micek
- Statistical Laboratory, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, 31-501, Poland
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Carlotta Franchi
- Laboratory of Pharmacoepidemiology and Human Nutrition, Department of Health Policy, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, 20156, Italy
- Italian Institute for Planetary Health (IIPH), Milan, 20124, Italy
| | - Andrea Poli
- Nutrition Foundation of Italy (NFI), Milan, 20124, Italy
| | - Maurizio Battino
- Department of Clinical Sciences, Università Politecnica Delle Marche, Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, Santander, 39011, Spain
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Alberto Dolci
- Sustainable Development Department, Bolton Food SpA, Milan, 20124, Italy
| | - Cristian Ricci
- Africa Unit for Transdisciplinary Health Research (AUTHeR), North-West University, Potchefstroom, 2531, South Africa
| | - 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
- International Training Program in Geroscience, Department of Public Health, Doctoral College, Semmelweis University, Budapest, Hungary
- 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
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy.
| |
Collapse
|
47
|
Che J, Sun Y, Deng Y, Zhang J. Blood-brain barrier disruption: a culprit of cognitive decline? Fluids Barriers CNS 2024; 21:63. [PMID: 39113115 PMCID: PMC11305076 DOI: 10.1186/s12987-024-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
48
|
Diorio TC, Abderezzai J, Nauman E, Kurt M, Tong Y, Rayz VL. MRI-based quantification of cardiac-driven brain biomechanics for early detection of neurological disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606246. [PMID: 39149257 PMCID: PMC11326150 DOI: 10.1101/2024.08.01.606246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
We present a pipeline to quantify biomechanical environment of the brain using solely MRI-derived data in order to elucidate the role of biomechanical factors in neurodegenerative disorders. Neurological disorders, like Alzheimer's and Parkinson's diseases, are associated with physical changes, including the accumulation of amyloid-β and tau proteins, damage to the cerebral vasculature, hypertension, atrophy of the cortical gray matter, and lesions of the periventricular white matter. Alterations in the external mechanical environment of cells can trigger pathological processes, and it is known that AD causes reduced stiffness in the brain tissue during degeneration. However, there appears to be a significant lag time between microscale changes and macroscale obstruction of neurological function in the brain. Here, we present a pipeline to quantify the whole brain biomechanical environment to bridge the gap in understanding how underlying brain changes affect macroscale brain biomechanics. This pipeline enables image-based quantification of subject-specific displacement field of the whole brain to subject-specific strain, strain rate, and stress across 133 labeled functional brain regions. We have focused our development efforts on utilizing solely MRI-derived data to facilitate clinical applicability of our approach and have emphasized automation in all aspects of our methods to reduce operator dependance. Our pipeline has the potential to improve early detection of neurological disorders and facilitate the identification of disease before widespread, irreversible damage has occurred.
Collapse
Affiliation(s)
- Tyler C. Diorio
- Purdue University, (Weldon School of Biomedical Engineering), West Lafayette, (IN), USA
| | - Javid Abderezzai
- University of Washington, (Department of Mechanical Engineering), Seattle, (WA), USA
| | - Eric Nauman
- University of Cincinnati, (Department of Biomedical Engineering), Cincinnati, (OH), USA
| | - Mehmet Kurt
- University of Washington, (Department of Mechanical Engineering), Seattle, (WA), USA
| | - Yunjie Tong
- Purdue University, (Weldon School of Biomedical Engineering), West Lafayette, (IN), USA
| | - Vitaliy L. Rayz
- Purdue University, (Weldon School of Biomedical Engineering), West Lafayette, (IN), USA
| |
Collapse
|
49
|
Chen JR, Lin CJ, Chang FC, Lee IH, Lu CF. Territory-Related Functional Connectivity Changes Associated with Verbal Memory Decline in Patients with Unilateral Asymptomatic Internal Carotid Stenosis. AJNR Am J Neuroradiol 2024; 45:934-942. [PMID: 38871370 DOI: 10.3174/ajnr.a8248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/12/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND AND PURPOSE Verbal memory decline is a common complaint of patients with severe asymptomatic stenosis of the internal carotid artery (aICS). Previous publications explored the associations between verbal memory decline and altered functional connectivity (FC) after aICS. Patients with severe aICS may show reduced perfusion in the ipsilateral territory and redistribution of cerebral blood flow to compensate for the deficient regions, including expansion of the posterior and contralateral ICA territories via the circle of Willis. However, aICS-related FC changes in anterior and posterior territories and the impact of the sides of stenosis were less explored. This study aims to investigate the altered FC in anterior and posterior circulation territories of patients with left or right unilateral aICS and its association with verbal memory decline. MATERIALS AND METHODS We enrolled 15 healthy controls (HCs), 22 patients with left aICS (aICSL), and 33 patients with right aICS (aICSR) to receive fMRI, Mini-Mental State Examination (MMSE), the Digit Span Test (DST), and the 12-item Chinese version of Verbal Learning Tests. We selected brain regions associated with verbal memory within anterior and posterior circulation territories. Territory-related FC alterations and verbal memory decline were identified by comparing the aICSL and aICSR groups with HC groups (P < .05, corrected for multiple comparisons), respectively. Furthermore, the association between altered FC and verbal memory decline was tested with the Pearson correlation analysis. RESULTS Compared with HCs, patients with aICSL or aICSR had significant impairment in delayed recall of verbal memory. Decline in delayed recall of verbal memory was significantly associated with altered FC between the right cerebellum and right middle temporal pole in the posterior circulation territory (r = 0.40, P = .03) in the aICSR group and was significantly associated with altered FC between the right superior medial frontal gyrus and left lingual gyrus in the anterior circulation territory (r = 0.56, P = .01) in the aICSL group. CONCLUSIONS Patients with aICSL and aICSR showed different patterns of FC alterations in both anterior and posterior circulation territories, which suggests that the side of aICS influences the compensatory mechanism for decline in delayed recall of verbal memory.
Collapse
Affiliation(s)
- Jyun-Ru Chen
- From the Department of Biomedical Imaging and Radiological Sciences (J.-R.C., C.-F.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Jen Lin
- School of Medicine (C.-J.L., F.-C.C., I.-H.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
- Neurological Institute (C.-J.L., I.-H.L.), Taipei Veterans General Hospital, Taipei, Taiwan
| | - Feng-Chi Chang
- School of Medicine (C.-J.L., F.-C.C., I.-H.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Radiology (F.-C.C.), Taipei Veterans General Hospital, Taipei, Taiwan
| | - I-Hui Lee
- School of Medicine (C.-J.L., F.-C.C., I.-H.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
- Neurological Institute (C.-J.L., I.-H.L.), Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Brain Science (I.-H.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Feng Lu
- From the Department of Biomedical Imaging and Radiological Sciences (J.-R.C., C.-F.L.), National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
50
|
Wei W, Ma D, Li L, Zhang L. Cognitive impairment in cerebral small vessel disease induced by hypertension. Neural Regen Res 2024; 19:1454-1462. [PMID: 38051887 PMCID: PMC10883517 DOI: 10.4103/1673-5374.385841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/22/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Hypertension is a primary risk factor for the progression of cognitive impairment caused by cerebral small vessel disease, the most common cerebrovascular disease. However, the causal relationship between hypertension and cerebral small vessel disease remains unclear. Hypertension has substantial negative impacts on brain health and is recognized as a risk factor for cerebrovascular disease. Chronic hypertension and lifestyle factors are associated with risks for stroke and dementia, and cerebral small vessel disease can cause dementia and stroke. Hypertension is the main driver of cerebral small vessel disease, which changes the structure and function of cerebral vessels via various mechanisms and leads to lacunar infarction, leukoaraiosis, white matter lesions, and intracerebral hemorrhage, ultimately resulting in cognitive decline and demonstrating that the brain is the target organ of hypertension. This review updates our understanding of the pathogenesis of hypertension-induced cerebral small vessel disease and the resulting changes in brain structure and function and declines in cognitive ability. We also discuss drugs to treat cerebral small vessel disease and cognitive impairment.
Collapse
Affiliation(s)
- Weipeng Wei
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center; Beijing Engineering Research Center for Nervous System Drugs; National Center for Neurological Disorders; National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Denglei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center; Beijing Engineering Research Center for Nervous System Drugs; National Center for Neurological Disorders; National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center; Beijing Engineering Research Center for Nervous System Drugs; National Center for Neurological Disorders; National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center; Beijing Engineering Research Center for Nervous System Drugs; National Center for Neurological Disorders; National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|