1
|
Pai V, Singh BN, Singh AK. Transformative Advances in Modeling Brain Aging and Longevity: Success, Challenges and Future Directions. Ageing Res Rev 2025:102753. [PMID: 40222396 DOI: 10.1016/j.arr.2025.102753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/30/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Research on brain aging is crucial for understanding age-related neurodegenerative disorders and developing several therapeutic interventions. Numerous models ranging from two-dimensional (2D) cell-based, invertebrate, vertebrate, and sophisticated three-dimensional (3D) models have been used to understand the process of brain aging. Invertebrate models are ideal for researching conserved aging processes because of their simplicity, short lifespans, and genetic tractability. Moreover, vertebrate models, including zebrafish and rodents, exhibit more complex nervous systems and behaviors, enabling the exploration of age-related neurodegeneration and cognitive decline. 2D cell culture models derived from primary cells or immortalized cell lines are widely used for mechanistic studies at the cellular level but lack the physiological complexity of brain tissue. Recent advancements have shifted focus to 3D models, which better recapitulate the brain's microenvironment. Organoids derived from induced pluripotent stem cells mimic human brain architecture and enable the study of cell-cell interactions and aging in a human-specific context. Brain-on-a-chip systems integrate microfluidics and 3D cultures to model blood-brain barrier dynamics and neuronal networks. Additionally, scaffold-based 3D cultures and spheroids provide intermediate complexity, allowing researchers to study extracellular matrix interactions and age-related changes in neuronal function. These 3D models bridge the gap between traditional 2D cultures and animal-based in vivo studies, offering unprecedented insights into brain aging mechanisms. By combining these diverse models, researchers can unravel the multifaceted processes of brain aging and accelerate the development of targeted therapies for age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Varsha Pai
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Bhisham Narayan Singh
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
2
|
Scieszka D, Hulse J, Gu H, Barkley-Levenson A, Barr E, Garcia M, Begay JG, Herbert G, McCormick M, Brigman J, Ottens A, Bleske B, Bhaskar K, Campen MJ. Neurometabolomic impacts of wood smoke and protective benefits of anti-aging therapeutics in aged female C57BL/6J mice. RESEARCH SQUARE 2025:rs.3.rs-5936676. [PMID: 40166007 PMCID: PMC11957201 DOI: 10.21203/rs.3.rs-5936676/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Wildland fires have become progressively more extensive over the past 30 years in the United States, routinely generating smoke that deteriorates air quality for most of the country. We explored the neurometabolomic impact of biomass-derived smoke on older (18 months) female C57BL/6J mice, both acutely and after 10 weeks of recovery from exposures. Methods Mice were exposed to wood smoke (WS) 4 hours/day, every other day, for 2 weeks (7 exposures total) to an average concentration of 448 µg particulate matter (PM)/m 3 per exposure. One group was euthanized 24 hours after the last exposure. Other groups were then placed on 1 of 4 treatment regimens for 10 weeks after wood smoke exposures: vehicle; resveratrol in chow plus nicotinamide mononucleotide in water (RNMN); senolytics via gavage (dasatanib + quercetin; DQ); or both RNMN with DQ (RNDQ). Results Among the findings, the aging from 18 months to 21 months was associated with the greatest metabolic shift, including changes in nicotinamide metabolism, with WS exposure effects that were relatively modest. WS caused a reduction in NAD + within the prefrontal cortex immediately after exposure and a long-term reduction in serotonin that persisted for 10 weeks. The serotonin reductions were corroborated by behavioral changes, including increased immobility in a forced swim test, and neuroinflammatory markers that persisted for 10 weeks. RNMN had the most beneficial effects after WS exposure, while RNDQ caused markers of brain aging to be upregulated within WS-exposed mice. Discussion Taken together, these findings highlight the persistent neurometabolomic and behavioral effects of woodsmoke exposure in an aged mouse model. Further examination is necessary to determine the age-specific and species-determinant response pathways and duration before complete resolution occurs.
Collapse
|
3
|
Yu J, Ji L, Liu Y, Wang X, Wang J, Liu C. Bone-brain interaction: mechanisms and potential intervention strategies of biomaterials. Bone Res 2025; 13:38. [PMID: 40097409 PMCID: PMC11914511 DOI: 10.1038/s41413-025-00404-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 03/19/2025] Open
Abstract
Following the discovery of bone as an endocrine organ with systemic influence, bone-brain interaction has emerged as a research hotspot, unveiling complex bidirectional communication between bone and brain. Studies indicate that bone and brain can influence each other's homeostasis via multiple pathways, yet there is a dearth of systematic reviews in this area. This review comprehensively examines interactions across three key areas: the influence of bone-derived factors on brain function, the effects of brain-related diseases or injuries (BRDI) on bone health, and the concept of skeletal interoception. Additionally, the review discusses innovative approaches in biomaterial design inspired by bone-brain interaction mechanisms, aiming to facilitate bone-brain interactions through materiobiological effects to aid in the treatment of neurodegenerative and bone-related diseases. Notably, the integration of artificial intelligence (AI) in biomaterial design is highlighted, showcasing AI's role in expediting the formulation of effective and targeted treatment strategies. In conclusion, this review offers vital insights into the mechanisms of bone-brain interaction and suggests advanced approaches to harness these interactions in clinical practice. These insights offer promising avenues for preventing and treating complex diseases impacting the skeleton and brain, underscoring the potential of interdisciplinary approaches in enhancing human health.
Collapse
Affiliation(s)
- Jiaze Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Luli Ji
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yongxian Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiaogang Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China.
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
4
|
Ohene Y, Morrey WJ, Powell E, Smethers KF, Luka N, South K, Berks M, Lawrence CB, Parker GJM, Parkes LM, Boutin H, Dickie BR. MRI detects blood-brain barrier alterations in a rat model of Alzheimer's disease and lung infection. NPJ IMAGING 2025; 3:8. [PMID: 40051735 PMCID: PMC11879872 DOI: 10.1038/s44303-025-00071-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 01/23/2025] [Indexed: 03/09/2025]
Abstract
Pneumonia is a common infection in people suffering with Alzheimer's disease, leading to delirium, critical illness or severe neurological decline, which may be due to an amplified response of the blood-brain barrier (BBB) to peripheral insult. We assess the response of the BBB to repeated Streptococcus pneumoniae lung infection in rat model of Alzheimer's disease (TgF344-AD), at 13- and 18-months old, using dynamic contrast-enhanced (DCE) MRI and filter exchange imaging. Higher BBB water exchange rate is initially detected in infected TgF344-AD rats. BBB water exchange rates correlated with hippocampus aquaporin-4 water channel expression in infected animals. We detected no differences in BBB permeability to gadolinium contrast agent measured by DCE-MRI, confirmed by staining for tight junction proteins, occludin and claudin-5. These findings provide insight into the mechanisms of how peripheral inflammation impacts the BBB.
Collapse
Affiliation(s)
- Yolanda Ohene
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - William J. Morrey
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elizabeth Powell
- Department of Medical Physics and Biomedical Engineering and Department of Neuroinflammation, Centre for Medical Image Computing, UCL, London, UK
| | - Katherine F. Smethers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nadim Luka
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kieron South
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael Berks
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catherine B. Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Geoff. J. M. Parker
- Department of Medical Physics and Biomedical Engineering and Department of Neuroinflammation, Centre for Medical Image Computing, UCL, London, UK
- Bioxydyn Limited, Manchester, UK
| | - Laura M. Parkes
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Hervé Boutin
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Imaging Brain & Neuropsychiatry iBraiN, Université de Tours, INSERM, Tours, France
| | - Ben R. Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
5
|
Wei P, Lin K, Chen X, Fang C, Qiu L, Hu J, Chang J. Sequential Proteomic Analysis Reveals the Key APOE4-Induced Pathological and Molecular Features at the Presymptomatic Stage in Alzheimer's Disease Mice. CNS Neurosci Ther 2025; 31:e70306. [PMID: 40075551 PMCID: PMC11903334 DOI: 10.1111/cns.70306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
AIMS Alzheimer's disease (AD) involves a prolonged presymptomatic or preclinical stage with subtle pathological changes. Apolipoprotein E4 (APOE4) is a significant genetic risk factor for AD, yet its specific role at the presymptomatic stage is not fully understood. This study aimed to elucidate the cellular and molecular effects of APOE4 compared to APOE3 on AD progression during the presymptomatic stage. METHODS We generated 5xFAD AD mice carrying human APOE3 or APOE4 and their non-AD controls. Behavioral tests, immunostaining, quantitative proteomics and phosphoproteomics, Golgi staining, and Western blotting were conducted at 3 or 10 months of age, respectively. Cell culture experiments were performed to assess APOE4's direct impact on neuronal mitochondrial function. RESULTS APOE4 significantly increased β-amyloid (Aβ) deposition and microglial activation compared to APOE3 in 5xFAD mice at the presymptomatic stage, without aggravating the blood-brain barrier disruption. Proteomic and biochemical analysis revealed strong molecular features of synaptic degeneration and mitochondrial dysfunction associated with APOE4. Notably, APOE4 promoted mitochondrial fusion and mitophagy while inhibiting fission, leading to impaired neuronal energy supply and increased reactive oxygen species. CONCLUSION Our findings indicate that APOE4 accelerates AD pathologies at the presymptomatic stage by exacerbating Aβ deposition, neuroinflammation, and synaptic degeneration. The study highlights mitochondrial dysfunction as a critical mediator of APOE4-induced AD progression, providing potential targets for early intervention.
Collapse
Affiliation(s)
- Pengju Wei
- State Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Kaihua Lin
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Cheng Fang
- State Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Linhui Qiu
- State Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Jun Hu
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Junlei Chang
- State Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Bennett T, Walmsley S, Bendayan R. Aging with HIV and HIV-associated neurocognitive impairment. AIDS 2025; 39:215-228. [PMID: 39878669 DOI: 10.1097/qad.0000000000004057] [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: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 01/31/2025]
Abstract
Antiretroviral therapy (ART) is the most effective therapeutic intervention for HIV infection. With improved survival, comorbidities, including neuropsychiatric and HIV-associated neurocognitive impairment (NCI) are of increasing concern to aging people with HIV (PWH). The clinical features and the inter-individual variability of the aging process confound the elucidation of the diagnosis and underlying mechanisms of cognitive dysfunction in aging PWH. Herein, we review the clinical aspects of HIV-associated NCI in the aging PWH contrasting to the normative neuro-aging seen in people without HIV (PWoH) and address the growing role of biomarkers to predict the onset of age-related diseases in PWH and their clinical significance. There is an urgent need for further research into the role of specific immune brain biomarkers in predicting the aging process and how these biomarkers may assist in understanding the mechanisms and possible prognosis of age-related neurocognitive comorbidities in aging PWH as an endpoint for interventional studies.
Collapse
Affiliation(s)
- Teresa Bennett
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| | - Sharon Walmsley
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| |
Collapse
|
7
|
Rajput M, Malik IA, Methi A, Cortés Silva JA, Fey D, Wirths O, Fischer A, Wilting J, von Arnim CAF. Cognitive decline and neuroinflammation in a mouse model of obesity: An accelerating role of ageing. Brain Behav Immun 2025; 125:226-239. [PMID: 39730092 DOI: 10.1016/j.bbi.2024.12.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/30/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024] Open
Abstract
Obesity, a pandemic, worldwide afflicts almost one billion people. Obesity and ageing share several pathological pathways leading to neurological disorders. However, due to a lack of suitable animal models, the long-term effects of obesity on age-related disorders- cognitive impairment and dementia have not yet been thoroughly investigated. Therefore, the current investigation focuses on developing a suitable model to explore the effects of obese-ageing. It also aims to determine whether obesity affects cognitive abilities in an age-dependent manner, and to identify a potential biomarker(s) for cognitive decline. Cognitive tests were carried out on 6-months and 1-year-old melanocortin-4 receptor (Mc4r)-deficient-obese and lean (wildtype) mice. Additionally, brains and sera were harvested for molecular, histological and serological analyses from 6, 12, and 24-months-old mice. Finally, RT-PCR was carried out after hippocampal mRNA sequencing. The cognitive tests revealed that 1-year-old obese mice have cognitive impairment along with underlying neurodegenerative changes, such as enlarged lateral ventricles. Serum neurofilament light chain (sNfL) levels were also elevated. Lipid accumulation and neuroinflammation were apparent besides, a compromised blood-brain barrier (BBB) indicated by altered junction protein gene expression. Differentially-expressed genes associated with cognitive decline were identified by mRNA sequencing of hippocampi. One such gene, Secreted Phosphoprotein 1 (Spp1) had markedly increased expression in cognitively-impaired obese mice. Our findings present an obese-aged mouse model of cognitive decline with neuroinflammation, reduced BBB-integrity and predisposing neurodegenerative changes. Obese-ageing accelerates the progression of cognitive impairment. Furthermore, Spp1 appears to be a potential biomarker for early diagnosis of neuropathological disorders.
Collapse
Affiliation(s)
- Mansi Rajput
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Ihtzaz Ahmed Malik
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Aditi Methi
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Jonathan Alexis Cortés Silva
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Dorothea Fey
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.
| | - Oliver Wirths
- Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany.
| | - André Fischer
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany; Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| | - Jörg Wilting
- Institute of Anatomy and Embryology, University Medical Center Goettingen, Kreuzbergring 36, D-37075 Goettingen, Germany.
| | - Christine A F von Arnim
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
8
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2025; 36:139-168. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
9
|
Rajab HA, Al-Kuraishy HM, Shokr MM, Al-Gareeb AI, Al-Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GES. Statins for vascular dementia: A hype or hope. Neuroscience 2025; 567:45-55. [PMID: 39746645 DOI: 10.1016/j.neuroscience.2024.12.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Vascular dementia (VaD) is a second most common type of dementia subsequent to Alzheimer disease (AD). VaD is characterized by cognitive impairment and memory loss that may progress due to the development of cerebral amyloid angiopathy (CAA) a hallmark of AD. CAA triggers the progression of ischemic and hemorrhagic strokes with the subsequent the development of VaD and mixed dementia. Early diagnosis of patients with appropriate use of anti-inflammatory can prevent CAA-related inflammation and VaD development. Currently, there are no effective drugs in the management of VaD. Of note, cholesterol-lowering agent statins which are commonly used in patients with vascular diseases and dyslipidemia may affect the progression of VaD. Many previous studies highlighted the potential therapeutic efficacy of statins in treating VaD. Though, the underlying mechanisms of statins in prevention and treatment of VaD are not fully clarified. Consequently, this review aims to discuss the mechanistic role of statins in the management of VaD, and how statins may adversely affect the cognitive function in VaD patients.
Collapse
Affiliation(s)
- Hussein A Rajab
- Endocrinology Consultant, Medical School, Najran University, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, Jabir Ibn Hayyan Medical University, Kufa, Iraq
| | - Nasser A Al-Harchan
- Department of Clinical Pharmacology, College of Dentistry, Al-Rasheed University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia; Department of Research & Development, Funogen, Athens, Greece
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
10
|
Bocharova M, Borza T, Watne LO, Engedal K, O'Brien JT, Selbæk G, Idland AV, Hodsoll J, Young AH, Aarsland D. The role of plasma inflammatory markers in late-life depression and conversion to dementia: a 3-year follow-up study. Mol Psychiatry 2025:10.1038/s41380-025-02908-2. [PMID: 39922907 DOI: 10.1038/s41380-025-02908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/27/2024] [Accepted: 01/21/2025] [Indexed: 02/10/2025]
Abstract
Late-life depression (LLD) has been linked to increased likelihood of dementia, although mechanisms responsible for this association remain largely unknown. One feature frequently observed in both LLD and dementia is elevated levels of plasma inflammatory markers. The present study aimed to compare the levels of 12 plasma inflammatory markers between older people with LLD and controls, and to explore whether these markers, along with clinical characteristics, can predict dementia in patients with LLD within 3 years of follow-up. Using multiple linear regression with stepwise adjustment, we compared levels of plasma inflammatory markers (IL-1β, IL-1ra, IL-6, IL-10, IL-17a, IL-18, IL-33, TNFα, CD40L, IFN-γ, CCL-2 and CCL-4) between 136 inpatients with LLD (PRODE cohort) and 103 cognitively healthy non-depressed controls (COGNORM cohort). In the PRODE cohort, follow-up data was available for 139 patients (of them 123 had data on baseline plasma inflammatory markers); 36 (25.9%) developed dementia by Year 3 (n = 31 for those with cytokine data). Using Cox proportional hazards regression, we explored whether inflammatory markers and clinical characteristics of LLD (age of onset, treatment response, number of episodes) predicted progression to dementia during follow-up. Levels of IL-1ra, CCL-2, CCL-4, IFN-γ and IL-17a were significantly higher in LLD patients compared to controls in the majority of models. However, none of the inflammatory markers predicted progression from LLD to dementia in the PRODE cohort. Among clinical features, only poor response to treatment significantly predicted higher risk of progression to dementia.
Collapse
Affiliation(s)
- M Bocharova
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - T Borza
- Research Centre for Age-Related Functional Decline and Disease, Innlandet Hospital Trust, Ottestad, Norway
| | - L O Watne
- Department of Geriatric Medicine, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - K Engedal
- Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - J T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - G Selbæk
- Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - A V Idland
- Department of Anesthesiology, Akershus University Hospital, Lørenskog, Norway
| | - J Hodsoll
- Department of Biostatistics & Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - A H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Division of Psychiatry, Imperial College London, London, UK
| | - D Aarsland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
11
|
Janus A, Dumas D, Le Douce J, Marie S, Pasculli G, Bambury P, Lemarchant S, Kremer P, Godfrin Y. Safety, Tolerability and Pharmacokinetic-Pharmacodynamic Relationship of NX210c Peptide in Healthy Elderly Volunteers: Randomized, Placebo-Controlled, Double-Blind, Multiple Ascending Dose Study. Neurol Ther 2025; 14:357-377. [PMID: 39708220 PMCID: PMC11762061 DOI: 10.1007/s40120-024-00691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Blood-brain barrier (BBB) integrity is fundamental to brain homeostasis, enabling control of substance exchange and safeguarding neurons against harmful toxins, pathogens, and immune cells that lead to dysregulation and inflammation involved in ageing and neurodegenerative diseases (NDD). The cyclized peptide NX210c is a thrombospondin type 1 repeat analogue derived from subcommissural organ-spondin. It exerts beneficial effects in animal models of NDD owing to its effects on neurons and endothelial cells. NX210c demonstrated a good safety profile in a single ascending dose phase 1a clinical study. The present multiple ascending dose phase 1b study was performed to evaluate the tolerability and pharmacological effects of repeated doses of NX210c in healthy elderly (age: > 55 years) volunteers. METHODS This was a randomized, placebo-controlled, double-blind study (EudraCT No. 2022-002868-76), investigating safety/tolerability, pharmacokinetics, and pharmacodynamics (including blood and cerebrospinal fluid biomarkers). Participants received 5 or 10 mg/kg NX210c or placebo (10-min infusion) thrice weekly for 4 weeks in an ascending dose fashion. Follow-up was conducted 2 weeks after last dosing. RESULTS The investigation included 29 participants. No serious adverse events were recorded and all adverse events were mild. Dedicated central nervous system testing did not reveal neurotoxicity. Biomarker evaluation showed a statistically significant reduction in blood claudin-5 and a trend toward reduction of blood homocysteine. In silico data modelling revealed salient pharmacokinetic-pharmacodynamic relationships, including reduction of claudin-5, neurofilament light chain, and SPARC-like protein 1 release, and degradation of homocysteine. CONCLUSION Multiple doses of NX210c exhibited a good safety profile, showed non-cumulative pharmacokinetics, and exerted pharmacodynamic effects on biomarkers linked to BBB integrity. The effects of NX210c on claudin-5 and biomarkers influencing BBB integrity-and the overarching brain protection it offers-provide a novel therapeutic strategy targeting an underlying driver of neurodegenerative conditions for which disease-modifying treatments are limited or not available.
Collapse
Affiliation(s)
- Annette Janus
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France.
| | - Daniël Dumas
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Juliette Le Douce
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Sébastien Marie
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | | | | | - Sighild Lemarchant
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Philip Kremer
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Yann Godfrin
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
- Godfrin Life-Sciences, Caluire-Et-Cuire, France
| |
Collapse
|
12
|
S. DSN, Sundararajan V. Gene expression analysis reveals mir-29 as a linker regulatory molecule among rheumatoid arthritis, inflammatory bowel disease, and dementia: Insights from systems biology approach. PLoS One 2025; 20:e0316584. [PMID: 39813219 PMCID: PMC11734936 DOI: 10.1371/journal.pone.0316584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a degenerative autoimmune disease, often managed through symptomatic treatment. The co-occurrence of the reported extra-articular comorbidities such as inflammatory bowel disease (IBD), and dementia may complicate the pathology of the disease as well as the treatment strategies. Therefore, in our study, we aim to elucidate the key genes, and regulatory elements implicated in the progression and association of these diseases, thereby highlighting the linked potential therapeutic targets. METHODOLOGY Ten microarray datasets each for RA, and IBD, and nine datasets for dementia were obtained from Gene Expression Omnibus. We identified common differentially expressed genes (DEGs) and constructed a gene-gene interaction network. Subsequently, topology analysis for hub gene identification, cluster and functional enrichment, and regulatory network analysis were performed. The hub genes were then validated using independent microarray datasets from Gene Expression Omnibus. RESULTS A total of 198 common DEGs were identified from which CD44, FN1, IGF1, COL1A2, and POSTN were identified as the hub genes in our study. These hub genes were mostly enriched in significant processes and pathways like tissue development, collagen binding, cell adhesion, regulation of ERK1/2 cascade, PI3K-AKT signaling, and cell surface receptor signaling. Key transcription factors TWIST2, CEBPA, EP300, HDAC1, HDAC2, NFKB1, RELA, TWIST1, and YY1 along with the miRNA hsa-miR-29 were found to regulate the expression of the hub genes significantly. Among these regulatory molecules, miR-29 emerged as a significant linker molecule, bridging the molecular mechanisms of RA, IBD, and dementia. Validation of our hub genes demonstrated a similar expression trend in the independent datasets used for our study. CONCLUSION Our study underscores the significant role of miR-29 in modulating the expression of hub genes and the associated transcription factors, which are crucial in the comorbidity status of RA, dementia, and IBD. This regulatory mechanism highlights miR-29 as a key player in the pathogenesis of these comorbid diseases.
Collapse
Affiliation(s)
- Devi Soorya Narayana S.
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Vino Sundararajan
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
13
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
14
|
Wang T, Huang X, Sun S, Wang Y, Han L, Zhang T, Zhang T, Chen X. Recent Advances in the Mechanisms of Postoperative Neurocognitive Dysfunction: A Narrative Review. Biomedicines 2025; 13:115. [PMID: 39857699 PMCID: PMC11762480 DOI: 10.3390/biomedicines13010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Postoperative neurocognitive dysfunction (PND) is a prevalent and debilitating complication in elderly surgical patients, characterized by persistent cognitive decline that negatively affects recovery and quality of life. As the aging population grows, the rising number of elderly surgical patients has made PND an urgent clinical challenge. Despite increasing research efforts, the pathophysiological mechanisms underlying PND remain inadequately characterized, underscoring the need for a more integrated framework to guide targeted interventions. To better understand the molecular mechanisms and therapeutic targets of PND, this narrative review synthesized evidence from peer-reviewed studies, identified through comprehensive searches of PubMed, Embase, Cochrane Library, and Web of Science. Key findings highlight neuroinflammation, oxidative stress, mitochondrial dysfunction, neurotransmitter imbalances, microvascular changes, and white matter lesions as central to PND pathophysiology, with particular parallels to encephalocele- and sepsis-associated cognitive impairments. Among these, neuroinflammation, mediated by pathways such as the NLRP3 inflammasome and blood-brain barrier disruption, emerges as a pivotal driver, triggering cascades that exacerbate neuronal injury. Oxidative stress and mitochondrial dysfunction synergistically amplify these effects, while neurotransmitter imbalances and microvascular alterations, including white matter lesions, contribute to synaptic dysfunction and cognitive decline. Anesthetic agents modulate these interconnected pathways, exhibiting both protective and detrimental effects. Propofol and dexmedetomidine demonstrate neuroprotective properties by suppressing neuroinflammation and microglial activation, whereas inhalational anesthetics like sevoflurane intensify oxidative stress and inflammatory responses. Ketamine, with its anti-inflammatory potential, offers promise but requires further evaluation to determine its long-term safety and efficacy. By bridging molecular insights with clinical practice, this review highlights the critical role of personalized anesthetic strategies in mitigating PND and improving cognitive recovery in elderly surgical patients. It aims to inform future research and clinical decision-making to address this multifaceted challenge.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xin Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| |
Collapse
|
15
|
Yang C, Zhao E, Zhang H, Duan L, Han X, Ding H, Cheng Y, Wang D, Lei X, Diwu Y. Xixin Decoction's novel mechanism for alleviating Alzheimer's disease cognitive dysfunction by modulating amyloid-β transport across the blood-brain barrier to reduce neuroinflammation. Front Pharmacol 2025; 15:1508726. [PMID: 39834810 PMCID: PMC11743276 DOI: 10.3389/fphar.2024.1508726] [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: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose Xixin Decoction (XXD) is a classical formula that has been used to effectively treat dementia for over 300 years. Modern clinical studies have demonstrated its significant therapeutic effects in treating Alzheimer's disease (AD) without notable adverse reactions. Nevertheless, the specific mechanisms underlying its efficacy remain to be elucidated. This investigation sought to elucidate XXD's impact on various aspects of AD pathology, including blood-brain barrier (BBB) impairment, neuroinflammatory processes, and amyloid-β (Aβ) deposition, as well as the molecular pathways involved in these effects. Methods In vitro experiments were conducted using hCMEC/D3 and HBVP cell coculture to establish an in vitro blood-brain barrier (BBB) model. BBB damage was induced in this model by 24-h exposure to 1 μg/mL lipopolysaccharide (LPS). After 24, 48, and 72 h of treatment with 10% XXD-medicated serum, the effects of XXD were assessed through Western blotting, RT-PCR, and immunofluorescence techniques. In vivo, SAMP8 mice were administered various doses of XXD via gavage for 8 weeks, including high-dose XXD group (H-XXD) at 5.07 g kg-1·d-1, medium-dose XXD group (M-XXD) at 2.535 g kg-1·d-1, and low-dose XXD group (L-XXD) at 1.2675 g kg-1·d-1. Cognitive function was subsequently evaluated using the Morris water maze test. BBB integrity was evaluated using Evans blue staining, and protein expression levels were analyzed via ELISA, Western blotting, and immunofluorescence. Results In vitro experiments revealed that XXD-containing serum, when cultured for 24, 48, and 72 h, could upregulate the expression of P-gp mRNA and protein, downregulate CB1 protein expression, and upregulate CB2 and Mfsd2a protein expression. In vivo studies demonstrated that XXD improved spatial learning and memory abilities in SAMP8 mice, reduced the amount of Evans blue extravasation in brain tissues, modulated the BBB-associated P-gp/ECS axis, RAGE/LRP1 receptor system, as well as MRP2 and Mfsd2a proteins, and decreased the accumulation of Aβ in the brains of SAMP8 mice. Additionally, XXD upregulated the expression of TREM2, downregulated IBA1, TLR1, TLR2, and CMPK2 expression, and reduced the levels of pro-inflammatory factors NLRP3, NF-κB p65, COX-2, TNF-α, and IL-1β in the hippocampal tissues. Conclusion XXD may exert its effects by regulating the P-gp/ECS axis, the RAGE/LRP1 receptor system, and the expression of MRP2 and Mfsd2a proteins, thereby modulating the transport function of the BBB to expedite the clearance of Aβ, reduce cerebral Aβ accumulation, and consequently inhibit the activation of microglia induced by Aβ aggregation. This process may suppress the activation of the CMPK2/NLRP3 and TLRs/NF-κB pathways, diminish the production of inflammatory cytokines and chemokines, alleviate neuroinflammation associated with microglia in the brain of AD, and ultimately improve AD pathology.
Collapse
Affiliation(s)
- Chaokai Yang
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Enlong Zhao
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hu Zhang
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Liqi Duan
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinyue Han
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hongli Ding
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Cheng
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Dengkun Wang
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang, China
- Key Research Laboratory for Prevention and Treatment of Cerebrospinal diseases, Shaanxi Provincial Administration of Traditional Chinese Medicine, Xianyang, China
- Discipline Innovation Team for Neurodegenerative Diseases of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaojing Lei
- Key Research Laboratory for Prevention and Treatment of Cerebrospinal diseases, Shaanxi Provincial Administration of Traditional Chinese Medicine, Xianyang, China
- Discipline Innovation Team for Neurodegenerative Diseases of Shaanxi University of Chinese Medicine, Xianyang, China
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yongchang Diwu
- Key Research Laboratory for Prevention and Treatment of Cerebrospinal diseases, Shaanxi Provincial Administration of Traditional Chinese Medicine, Xianyang, China
- Discipline Innovation Team for Neurodegenerative Diseases of Shaanxi University of Chinese Medicine, Xianyang, China
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
16
|
Hamblin PS, Russell AW, Talic S, Zoungas S. The growing range of complications of diabetes mellitus. Trends Endocrinol Metab 2025:S1043-2760(24)00328-X. [PMID: 39755491 DOI: 10.1016/j.tem.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025]
Abstract
With the rising prevalence of type 2 diabetes mellitus (T2DM) and obesity, several previously under-recognised complications associated with T2DM are becoming more evident. The most common of these emerging complications are metabolic dysfunction-associated steatotic liver disease (MASLD), cancer, dementia, sarcopenia, and frailty, as well as other conditions involving the lung, heart, and intestinal tract. Likely causative factors are chronic inflammation and insulin resistance, whereas blood glucose levels appear to play a lesser role. We discuss these complications and the new approaches being developed to prevent and manage them, especially incretin-based therapies. We argue that these new interventions may work in a complementary way to other proven cardiorenal protective therapies to reduce the burden of T2DM complications.
Collapse
Affiliation(s)
- Peter S Hamblin
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, Alfred Health, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, Western Health, St Albans, VIC, Australia.
| | - Anthony W Russell
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, Alfred Health, Melbourne, VIC, Australia
| | - Stella Talic
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Sophia Zoungas
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
17
|
He Z, Sun J. The role of the neurovascular unit in vascular cognitive impairment: Current evidence and future perspectives. Neurobiol Dis 2025; 204:106772. [PMID: 39710068 DOI: 10.1016/j.nbd.2024.106772] [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: 10/17/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
Vascular cognitive impairment (VCI) is a progressive cognitive impairment caused by cerebrovascular disease or vascular risk factors. It is the second most common type of cognitive impairment after Alzheimer's disease. The pathogenesis of VCI is complex, and neurovascular unit destruction is one of its important mechanisms. The neurovascular unit (NVU) is responsible for combining blood flow with brain activity and includes endothelial cells, pericytes, astrocytes and many regulatory nerve terminals. The concept of an NVU emphasizes that interactions between different types of cells are essential for maintaining brain homeostasis. A stable NVU is the basis of normal brain function. Therefore, understanding the structure and function of the neurovascular unit and its role in VCI development is crucial for gaining insights into its pathogenesis. This article reviews the structure and function of the neurovascular unit and its contribution to VCI, providing valuable information for early diagnosis and prevention.
Collapse
Affiliation(s)
- Zhidong He
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130031, Jilin, China
| | - Jing Sun
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130031, Jilin, China..
| |
Collapse
|
18
|
Sasannia S, Leigh R, Bastani PB, Shin HG, van Zijl P, Knutsson L, Nyquist P. Blood-brain barrier breakdown in brain ischemia: Insights from MRI perfusion imaging. Neurotherapeutics 2025; 22:e00516. [PMID: 39709246 PMCID: PMC11840350 DOI: 10.1016/j.neurot.2024.e00516] [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: 09/08/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
Brain ischemia is a major cause of neurological dysfunction and mortality worldwide. It occurs not only acutely, such as in acute ischemic stroke (AIS), but also in chronic conditions like cerebral small vessel disease (cSVD). Any other conditions resulting in brain hypoperfusion can also lead to ischemia. Ischemic events can cause blood-brain barrier (BBB) disruption and, ultimately, white matter alterations, contributing to neurological deficits and long-term functional impairments. Hence, understanding the mechanisms of BBB breakdown and white matter injury across various ischemic conditions is critical for developing effective interventions and improving patient outcomes. This review discusses the proposed mechanisms of ischemia-related BBB breakdown. Moreover, magnetic resonance imaging (MRI) based perfusion-weighted imaging (PWI) techniques sensitive to BBB permeability changes are described, including dynamic contrast-enhanced (DCE-MRI) and dynamic susceptibility contrast MRI (DSC-MRI), two perfusion-weighted imaging (PWI). These PWI techniques provide valuable insights that improve our understanding of the complex early pathophysiology of brain ischemia, which can lead to better assessment and management. Finally, in this review, we explore the implications of the mentioned neuroimaging findings, which emphasize the potential of neuroimaging biomarkers to guide personalized treatment and inform novel neuroprotective strategies. This review highlights the importance of investigating BBB changes in brain ischemia and the critical role of advanced neuroimaging in improving patient care and advancing stroke research.
Collapse
Affiliation(s)
- Sarvin Sasannia
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States.
| | - Richard Leigh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Pouya B Bastani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Hyeong-Geol Shin
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Peter van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Linda Knutsson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States; Department of Medical Radiation Physics, Lund University, Lund, Sweden.
| | - Paul Nyquist
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Neurocritical Care Division, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, MD, United States; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
19
|
AlRawili N, Al‐Kuraishy HM, Al‐Gareeb AI, Abdel‐Fattah MM, Al‐Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GE. Trajectory of Cardiogenic Dementia: A New Perspective. J Cell Mol Med 2025; 29:e70345. [PMID: 39828641 PMCID: PMC11742966 DOI: 10.1111/jcmm.70345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/27/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
The functions of the heart and brain are closely linked and essential to support human life by the heart-brain axis, which is a complex interconnection between the heart and brain. Also, cardiac function and cerebral blood flow regulate the brain's metabolism and function. Therefore, deterioration of cardiac function may affect cognitive function and may increase the risk of dementia. Cardiogenic dementia is defined as a cognitive deterioration due to heart diseases such as heart failure, myocardial infarction, and atrial fibrillation. The prevalence of cognitive impairment in patients with heart failure was 29%. In addition, coronary artery disease (CAD) is also associated with the development of cognitive impairment. CAD and reduction of myocardial contractility reduced cerebral blood flow and increased the risk of dementia in CAD patients. Furthermore, myocardial infarction and subsequent systemic haemodynamic instability promote the development and progression of cardiogenic dementia. These findings indicated that many cardiac diseases are implicated in the development and progression of cognitive impairment. Nevertheless, the underlying mechanism for the development of cardiogenic dementia was not fully elucidated. Consequently, this review aims to discuss the potential mechanisms involved in the pathogenesis of cardiogenic dementia.
Collapse
Affiliation(s)
- Nawaf AlRawili
- Department of Internal Medicine, College of MedicineNorthern Border UniversityArarSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical PharmacologyJabir ibn Hayyan Medical UniversityKufaIraq
| | - Maha M. Abdel‐Fattah
- Department of Pharmacology and Toxicology, Faculty of PharmacyBeni‐Suef UniversityBeni‐SuefEgypt
| | - Nasser A. Al‐Harchan
- Department of Clinical Pharmacology, College of DentistryAl‐Rasheed UniversityBaghdadIraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of MedicineJouf UniversitySaudi Arabia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐ HerdeckeWuppertalGermany
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliIndia
- Department of Science and EngineeringNovel Global Community Educational FoundationNew South WalesAustralia
- Department of Research & DevelopmentAthensGreece
| | - Gaber El‐Saber Batiha
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
20
|
Power L, Shuhmaher R, Houtz P, Chen J, Rudolph S, Yuen J, Machour M, Levy E, Wu L, Levenberg S, Whalen M, Chen Y, Kaplan DL. 3D Neurovascular Unit Tissue Model to Assess Responses to Traumatic Brain Injury. J Biomed Mater Res A 2025; 113:e37816. [PMID: 39440483 DOI: 10.1002/jbm.a.37816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
The neurovascular unit (NVU) is a critical interface in the central nervous system that links vascular interactions with glial and neural tissue. Disruption of the NVU has been linked to the onset and progression of neurodegenerative diseases. Despite its significance the NVU remains challenging to study in a physiologically relevant manner. Here, a 3D cell triculture model of the NVU is developed that incorporates human primary brain microvascular endothelial cells, astrocytes, and pericytes into a tissue system that can be sustained in vitro for several weeks. This tissue model helps recapitulate the complexity of the NVU and can be used to interrogate the mechanisms of disease and cell-cell interactions. The NVU tissue model displays elevated cell death and inflammatory responses following mechanical damage, to emulate traumatic brain injury (TBI) under controlled laboratory conditions, including lactate dehydrogenase (LDH) release, elevated inflammatory markers TNF-α and monocyte chemoattractant cytokines MCP-2 and MCP-3 and reduced expression of the tight junction marker ZO-1. This 3D tissue model serves as a tool for deciphering mechanisms of TBIs and immune responses associated with the NVU.
Collapse
Affiliation(s)
- Liam Power
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Rita Shuhmaher
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Philip Houtz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Jinpeng Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - John Yuen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Majd Machour
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Emily Levy
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Limin Wu
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
21
|
Gogishvili D, Honey MIJ, Verberk IMW, Vermunt L, Hol EM, Teunissen CE, Abeln S. The GFAP proteoform puzzle: How to advance GFAP as a fluid biomarker in neurological diseases. J Neurochem 2025; 169:e16226. [PMID: 39289040 DOI: 10.1111/jnc.16226] [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: 06/05/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Glial fibrillary acidic protein (GFAP) is a well-established biomarker of reactive astrogliosis in the central nervous system because of its elevated levels following brain injury and various neurological disorders. The advent of ultra-sensitive methods for measuring low-abundant proteins has significantly enhanced our understanding of GFAP levels in the serum or plasma of patients with diverse neurological diseases. Clinical studies have demonstrated that GFAP holds promise both as a diagnostic and prognostic biomarker, including but not limited to individuals with Alzheimer's disease. GFAP exhibits diverse forms and structures, herein referred to as its proteoform complexity, encompassing conformational dynamics, isoforms and post-translational modifications (PTMs). In this review, we explore how the proteoform complexity of GFAP influences its detection, which may affect the differential diagnostic performance of GFAP in different biological fluids and can provide valuable insights into underlying biological processes. Additionally, proteoforms are often disease-specific, and our review provides suggestions and highlights areas to focus on for the development of new assays for measuring GFAP, including isoforms, PTMs, discharge mechanisms, breakdown products, higher-order species and interacting partners. By addressing the knowledge gaps highlighted in this review, we aim to support the clinical translation and interpretation of GFAP in both CSF and blood and the development of reliable, reproducible and specific prognostic and diagnostic tests. To enhance disease pathology comprehension and optimise GFAP as a biomarker, a thorough understanding of detected proteoforms in biofluids is essential.
Collapse
Affiliation(s)
- Dea Gogishvili
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Madison I J Honey
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Sanne Abeln
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
22
|
Owens CD, Pinto CB, Szarvas Z, Muranyi M, da C. Pinaffi-Langley AC, Peterfi A, Mukli P, Detwiler S, Olay L, Kaposzta Z, Smith K, Kirkpatrick AC, Saleh Velez F, Tarantini S, Csiszar A, Ungvari ZI, Prodan CI, Yabluchanskiy A. COVID-19 Exacerbates Neurovascular Uncoupling and Contributes to Endothelial Dysfunction in Patients with Mild Cognitive Impairment. Biomolecules 2024; 14:1621. [PMID: 39766328 PMCID: PMC11726736 DOI: 10.3390/biom14121621] [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: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Mild cognitive impairment (MCI) affects nearly 20% of older adults worldwide, with no targetable interventions for prevention. COVID-19 adversely affects cognition, with >70% of older adults with Long COVID presenting with cognitive complaints. Neurovascular coupling (NVC), an essential mechanism of cognitive function, declines with aging and is further attenuated in neurocognitive disorders. The effect of COVID-19 on NVC responses has yet to be addressed in older adults who are vulnerable to dementia progression. Participants with MCI and a history of COVID-19 (COV+, N = 31) and MCI participants with no history of infection (COV- N = 11) participated in this cross-sectional study to determine if COVID-19 affects cerebrocortical NVC responses and vascular function. Functional near-infrared spectroscopy was used to measure cerebrocortical NVC responses, and endothelial function was assessed via insonation of the brachial artery during a flow-mediated dilation protocol. NVC responses were elicited by the working memory n-back paradigm. NVC in the left dorsolateral prefrontal cortex and endothelial function was decreased in the COV+ group compared to the COV- group. These data provide mechanistic insight into how COVID-19 may exacerbate long-term cognitive sequela seen in older adults, highlighting the urgent need for further research and clinical trials to explore novel therapeutic interventions aimed at preserving/restoring NVC.
Collapse
Affiliation(s)
- Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Camila B. Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Mihaly Muranyi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Ana Clara da C. Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1087 Budapest, Hungary
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Zalan Kaposzta
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1087 Budapest, Hungary
| | - Kenneth Smith
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA;
| | - Angelia C. Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; (A.C.K.); (C.I.P.)
- Cardiovascular Disease Section, Department of Medicine, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA
| | - Faddi Saleh Velez
- Department of Neurology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA;
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltan I. Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Calin I. Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; (A.C.K.); (C.I.P.)
- Department of Neurology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA;
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA; (C.D.O.); (C.B.P.); (Z.S.); (M.M.); (A.C.d.C.P.-L.); (A.P.); (P.M.); (S.D.); (L.O.); (Z.K.); (S.T.); (A.C.); (Z.I.U.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Neurosurgery, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, 1089 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
23
|
Scheinman SB, Dong H. The impact of sex on memory during aging and Alzheimer's disease progression: Epigenetic mechanisms. J Alzheimers Dis 2024; 102:562-576. [PMID: 39539121 PMCID: PMC11721493 DOI: 10.1177/13872877241288709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, disability, and death in the elderly. While the etiology of AD is unknown, there are several established risk factors for the disease including, aging, female sex, and genetics. However, specific genetic mutations only account for a small percentage (1-5%) of AD cases and the much more common sporadic form of the disease has no causative genetic basis, although certain risk factor genes have been identified. While the genetic code remains static throughout the lifetime, the activation and expression levels of genes change dynamically over time via epigenetics. Recent evidence has emerged linking changes in epigenetics to the pathogenesis of AD, and epigenetic alterations also modulate cognitive changes during physiological aging. Aging is the greatest risk factor for the development of AD and two-thirds of all AD patients are women, who experience an increased rate of symptom progression compared to men of the same age. In humans and other mammalian species, males and females experience aging differently, raising the important question of whether sex differences in epigenetic regulation during aging could provide an explanation for sex differences in neurodegenerative diseases such as AD. This review explores distinct epigenetic changes that impact memory function during aging and AD, with a specific focus on sexually divergent epigenetic alterations (in particular, histone modifications) as a potential mechanistic explanation for sex differences in AD.
Collapse
Affiliation(s)
- Sarah B Scheinman
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
24
|
Contreras JA, Fujisaki K, Ortega NE, Barisano G, Sagare A, Pappas I, Chui H, Ringman JM, Joe EB, Zlokovic BV, Toga AW, Pa J. Decreased functional connectivity is associated with increased levels of Cerebral Spinal Fluid soluble-PDGFRβ, a marker of blood brain barrier breakdown, in older adults. Brain Imaging Behav 2024; 18:1343-1354. [PMID: 39254921 PMCID: PMC11680618 DOI: 10.1007/s11682-024-00912-8] [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] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
Resting-state functional connectivity (FC) is suggested to be cross-sectionally associated with both vascular burden and Alzheimer's disease (AD) pathology. For instance, studies in pre-clinical AD subjects have shown increases of cerebral spinal fluid soluble platelet-derived growth factor receptor-β (CSF sPDGFRβ, a marker of BBB breakdown) but have not demonstrated if this vascular impairment affects neuronal dysfunction. It's possible that increased levels of sPDGFRβ in the CSF may correlate with impaired FC in metabolically demanding brain regions (i.e. Default Mode Network, DMN). Our study aimed to investigate the relationship between these two markers in older individuals that were cognitively normal and had cognitive impairment. Eighty-nine older adults without dementia from the University of Southern California were selected from a larger cohort. Region of interest (ROI) to ROI analyses were conducted using DMN seed regions. Linear regression models measured significant associations between BOLD FC strength among seed-target regions and sPDGFRβ values, while covarying for age and sex. Comparison of a composite ROI created by averaging FC values between seed and all target regions among cognitively normal and impaired individuals was also examined. Using CSF sPDGFRβ as a biomarker of BBB breakdown, we report that increased breakdown correlated with decreased functional connectivity in DMN areas, specifically the PCC, and while the hippocampus exhibited an interaction effect using CDR score, this was an exploratory analysis that we feel can lead to further research. Ultimately, we found that BBB breakdown, as measured by CSF sPDGFRβ, is associated with neural networks, and decreased functional connections.
Collapse
Affiliation(s)
- Joey A Contreras
- Department of Neurosciences, Alzheimer's Disease Cooperative Study (ADCS), University of California, San Diego, CA, USA
| | - Kimiko Fujisaki
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Nancy E Ortega
- Department of Neurosciences, Alzheimer's Disease Cooperative Study (ADCS), University of California, San Diego, CA, USA
| | - Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Ioannis Pappas
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Helena Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth B Joe
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Judy Pa
- Department of Neurosciences, Alzheimer's Disease Cooperative Study (ADCS), University of California, San Diego, CA, USA.
| |
Collapse
|
25
|
Kim S, Jung UJ, Kim SR. Role of Oxidative Stress in Blood-Brain Barrier Disruption and Neurodegenerative Diseases. Antioxidants (Basel) 2024; 13:1462. [PMID: 39765790 PMCID: PMC11673141 DOI: 10.3390/antiox13121462] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Upregulation of reactive oxygen species (ROS) levels is a principal feature observed in the brains of neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). In these diseases, oxidative stress can disrupt the blood-brain barrier (BBB). This disruption allows neurotoxic plasma components, blood cells, and pathogens to enter the brain, leading to increased ROS production, mitochondrial dysfunction, and inflammation. Collectively, these factors result in protein modification, lipid peroxidation, DNA damage, and, ultimately, neural cell damage. In this review article, we present the mechanisms by which oxidative damage leads to BBB breakdown in brain diseases. Additionally, we summarize potential therapeutic approaches aimed at reducing oxidative damage that contributes to BBB disruption in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
26
|
Salian VS, Tang X, Thompson KJ, Curan GL, Lowe VJ, Li L, Kalari KR, Kandimalla KK. Molecular Mechanisms Underlying Amyloid Beta Peptide Mediated Upregulation of Vascular Cell Adhesion Molecule-1 in Alzheimer Disease. J Pharmacol Exp Ther 2024; 391:430-440. [PMID: 39455283 PMCID: PMC11585316 DOI: 10.1124/jpet.124.002280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Amyloid β(Aβ) deposition and neurofibrillary tangles are widely considered the primary pathological hallmarks of familial and sporadic forms of Alzheimer disease (AD). However, cerebrovascular inflammation, which is prevalent in 70% of AD patients, is emerging as another core feature of AD pathology. In our current work, we investigated the hypothesis that Aβ42 exposure drives an increase in vascular cell adhesion molecule-1 (VCAM-1) expression, a cerebrovascular inflammatory marker expressed on the blood-brain barrier (BBB) endothelium in humans and murine models. We have demonstrated that the inflammation signaling pathway is upregulated in AD patient brains, and VCAM-1 expression is increased in AD patients compared with healthy controls. Furthermore, dynamic SPEC/CT imaging in APP,PS1 transgenic mice (a mouse model that overexpresses Aβ42) demonstrated VCAM-1 upregulation at the BBB. Although there is a strong association between Aβ42 exposure and an increase in VCAM-1 expression, the underlying mechanisms remain partially understood. Molecular mechanisms driving VCAM-1 expression at the BBB were investigated in polarized human cerebral microvascular endothelial cell monolayers. Moreover, by employing reverse-phase protein array assays and immunocytochemistry we demonstrated that Aβ42 increases VCAM-1 expression via the Src/p38/MEK signaling pathway. Therefore, targeting the Src/p38/MEK pathway may help modulate VCAM-1 expression at the BBB and help mitigate cerebrovascular inflammation in Alzheimer disease. SIGNIFICANCE STATEMENT: Although considered a core pathological feature of Alzheimer disease, molecular pathways leading to cerebrovascular inflammation remain only partially understood. Moreover, clinical diagnostic methods for detecting cerebrovascular inflammation are underdeveloped. This study demonstrated the detection of VCAM-1 using radio-iodinated VCAM-1 antibody and single-photon emission computed tomography/computed tomography imaging. Additionally, exposure to Aβ42 increases VCAM-1 expression on the blood-brain barrier endothelium via the Src/p38/MEK pathway. These findings are expected to aid in the development of diagnostic and therapeutic approaches for addressing cerebrovascular inflammation in AD.
Collapse
Affiliation(s)
- Vrishali S Salian
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Xiaojia Tang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Kevin J Thompson
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Geoffry L Curan
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Krishna R Kalari
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| |
Collapse
|
27
|
Yoon G, Kam MK, Koh YH, Jo C. Palmitoyl-L-carnitine induces tau phosphorylation and mitochondrial dysfunction in neuronal cells. PLoS One 2024; 19:e0313507. [PMID: 39536002 PMCID: PMC11560007 DOI: 10.1371/journal.pone.0313507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and memory loss, involving mechanisms such as tau hyperphosphorylation and mitochondrial dysfunction. Increasing evidence suggests that age-related alterations in metabolite levels are crucial for the pathogenesis of AD. Here, we analyzed serum metabolites from mice of various ages (2, 4, 14, and 21 months old) using mass spectrometry. We identified palmitoyl-L-carnitine as a key metabolite with significantly increased levels in aged mice. In vitro experiments with SH-SY5Y neuronal cells demonstrated that palmitoyl-L-carnitine treatment enhanced tau phosphorylation, increased mitochondrial fission, and elevated intracellular calcium levels. Furthermore, the increased levels of tau phosphorylation were significantly reduced by the inhibition of GSK-3β, CDK5, and calpain, indicating that tau kinases activated by calcium overload are directly involved in the increase of tau phosphorylation. Considering that mitochondrial fission is related to mitochondrial dysfunction, we propose that the elevated level of serum palmitoyl-L-carnitine during aging contributes to AD pathology through these pathways. These findings highlight the significant role of lipid metabolism in neurodegeneration and offer potential therapeutic targets for age-related diseases, including AD.
Collapse
Affiliation(s)
- Gwangho Yoon
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Min Kyoung Kam
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Young Ho Koh
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
28
|
Bai R, Ge X. Blood-brain barrier disruption following brain injury: Implications for clinical practice. Histol Histopathol 2024; 39:1435-1441. [PMID: 38618940 DOI: 10.14670/hh-18-740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The blood-brain barrier (BBB) plays a critical role in regulating the exchange of substances between peripheral blood and the central nervous system and in maintaining the stability of the neurovascular unit in neurological diseases. To guide clinical treatment and basic research on BBB protection following brain injury, this manuscript reviews how BBB disruption develops and influences neural recovery after stroke and traumatic brain injury (TBI). By summarizing the pathological mechanisms of BBB damage, we underscore the critical role of promoting BBB repair in managing brain injury. We also emphasize the potential for personalized and precise therapeutic strategies and the need for continued research and innovation. From this, broadening insights into the mechanisms of BBB disruption and repair could pave the way for breakthroughs in the treatment of brain injury-related diseases.
Collapse
Affiliation(s)
- Ruojing Bai
- Department of Geriatrics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Xintong Ge
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Geriatrics Institute, Tianjin, PR China.
| |
Collapse
|
29
|
Tseng PH, Huang LC, Huang XL, Huang BR, Lin SZ, Tsai ST, Huang HY. Blood-brain barrier-associated biomarker correlated with cerebral small vessel disease and shunt outcome in normal pressure hydrocephalus: a prospective cohort study. Int J Surg 2024; 110:6962-6971. [PMID: 39166950 PMCID: PMC11573114 DOI: 10.1097/js9.0000000000002038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Blood-brain barrier (BBB) breakdown is associated with neurodegeneration and cognitive impairment. Cerebral small vessel disease (CSVD) is also common in idiopathic normal pressure hydrocephalus (iNPH). Biomarkers in the cerebrospinal fluid (CSF) may reflect the severity of neuropathological damage and indicate a relationship between BBB integrity and iNPH and its surgical outcome. The authors investigated the association of CSVD and comorbidity-related CSF biomarkers with shunt outcomes in iNPH. MATERIALS AND METHODS This prospective cohort study recruited 53 patients with iNPH, who were subgrouped by CSVD severity. CSF proteins were analyzed, including soluble platelet-derived growth factor receptor-β (sPDGFR-β), Alzheimer's disease biomarkers, neurofilament light chain (NfL), and triggering receptor expressed on myeloid cells 2 (Trem2). We assessed symptom improvement, investigated its association with biomarkers levels, calculated protein cutoffs for surgical outcomes using receiver operating characteristic (ROC) curves, and compared model predictions using different proteins through hierarchical regression analysis. RESULTS Among patients with iNPH, 74% had comorbid CSVD. Patients with severe CSVD exhibited significantly higher sPDGFR-β levels ( P =0.019) and better postoperative performance (β=0.332, t=2.174, P =0.039; r =0.573, P =0.001). Analysis of the predictive potential of the biomarkers showed that sPDGFR-β was predictive of surgical outcomes (area under curve=0.82, sensitivity=66.8%, specificity=94.7%). A Comparison of the models revealed a greater effect of sPDGFR-β (Adjusted R 2 =0.247, ∆R 2 =0.160, ∆F(1, 37)=8.238, P =0.007) on cognitive improvement. CONCLUSION This study highlighted the relevance of CSF biomarkers in assessing CSVD severity and predicting iNPH surgical outcomes. CSF shunt surgery may provide an alternative treatment for neurodegenerative diseases with BBB breakdown and dysfunctional CSF clearance.
Collapse
Affiliation(s)
- Pao-Hui Tseng
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Department of Nursing, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
| | - Li-Chuan Huang
- Department of Medical Imaging, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
| | - Xiang-Ling Huang
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Department of Nursing, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung
- School of Medicine, Tzu Chi University
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Buddhist Tzu Chi Bioinnovation Center, Buddhist Tzu Chi Medical Foundation
| | - Sheng-Tzung Tsai
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
- School of Medicine, Tzu Chi University
| | - Hsin-Yi Huang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
| |
Collapse
|
30
|
Khodadadi M, Pirzad Jahromi G, Meftahi GH, Khodadadi H, Hadipour M, Ezami M. Crocin nano-chitosan-coated compound mitigates hippocampal blood-brain barrier disruption, anxiety, and cognitive deficits in chronic immobilization stress-induced rats. Heliyon 2024; 10:e39203. [PMID: 39640648 PMCID: PMC11620202 DOI: 10.1016/j.heliyon.2024.e39203] [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: 12/12/2023] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Stressful conditions can disrupt the central nervous system's normal homeostasis and physiological functions, resulting in blood-brain barrier malfunction, memory and learning impairment, anxiety, etc. Crocin is a long-investigated natural compound that has been documented to have anti-inflammation and neuroprotective effects, albeit it comes with some limitations such as low stability and bioavailability. Therefore, we aimed to overcome crocin's limitations by coating crocin with a nano-carrier (chitosan) in the chronic immobilization stress-induced rat model. Crocin was encapsulated into chitosan nanoparticles by a modified method. A total of 35 male Wistar rats were selected as our study subjects (220-250 g) which were randomly divided into 5 groups (control, stress, nanoparticle, crocin, and chitosan). Chronic immobilization stress was induced by placing rats for 2 h into a plastic bottle with specific measurements (for 14 consecutive days) to prevent animals from moving. To evaluate the memory and learning changes, we used the Barnes maze test and the Passive avoidance test followed by the evaluation of the N-methyl-D-aspartate |(NMDA) receptor subunits genes (GRIN1 and GRIN2A) expression. Anxiety levels were evaluated by elevated plus maze test. Furthermore, the changes in the expression of genes responsible for encoding the tight junction proteins of BBB including ZO1, CLDN5, and OCLN were assessed by RT-PCR. Compared to intact crocin, the administration of crocin nano-chitosan-coated compound resulted in significant improvement of specific memory and learning indicators as well as a significant reduction of anxiety levels in chronic immobilization stress-induced rats. Finally, we observed that treatment with the crocin nano-chitosan-coated compound can elevate the expression levels of the genes responsible for encoding NMDA receptor subunits, and the genes responsible for encoding the tight junction proteins of blood-brain barriers in the hippocampus.
Collapse
Affiliation(s)
- Mohsen Khodadadi
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gila Pirzad Jahromi
- Neuroscience Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khodadadi
- The Polish Academy of Sciences, Institute of Genetics and Animal Biotechnology, Warsaw, Poland
| | | | - Masoud Ezami
- Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Almanza DLV, Koletar MM, Lai AY, Lam WW, Joo L, Hill ME, Stanisz GJ, McLaurin J, Stefanovic B. High caloric intake improves neuronal metabolism and functional hyperemia in a rat model of early AD pathology. Theranostics 2024; 14:7405-7423. [PMID: 39659583 PMCID: PMC11626934 DOI: 10.7150/thno.98793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction: While obesity has been linked to both increased and decreased rate of cognitive decline in Alzheimer's Disease (AD) patients, there is no consensus on the interaction between obesity and AD. Methods: The TgF344-AD rat model was used to investigate the effects of high carbohydrate, high fat (HCHF) diet on brain glucose metabolism and hemodynamics in the presence or absence of AD transgenes, in presymptomatic (6-month-old) vs. symptomatic (12-month-old) stages of AD progression using non-invasive neuroimaging. Results: In presymptomatic AD, HCHF exerted detrimental effects, attenuating both hippocampal glucose uptake and resting perfusion in both non-transgenic and TgAD cohorts, when compared to CHOW-fed cohorts. In contrast, HCHF consumption was beneficial in established AD, resolving the AD-progression associated attenuation in hippocampal glucose uptake and functional hyperemia. Discussion: Whereas HCHF was harmful to the presymptomatic AD brain, it ameliorated deficits in hippocampal metabolism and neurovascular coupling in symptomatic TgAD rats.
Collapse
Affiliation(s)
- Dustin Loren V. Almanza
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Aaron Y. Lai
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Wilfred W. Lam
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Lewis Joo
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Mary E. Hill
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Greg J. Stanisz
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - JoAnne McLaurin
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Bojana Stefanovic
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
32
|
Naranjo O, Osborne OM, Torices S, Schmidlin S, Tiburcio D, Park M, Toborek M. Microvessel isolation protocol for RNA visualization and profiling. Sci Rep 2024; 14:25558. [PMID: 39462110 PMCID: PMC11513094 DOI: 10.1038/s41598-024-77501-8] [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: 03/27/2024] [Accepted: 10/23/2024] [Indexed: 10/28/2024] Open
Abstract
Disruptions in pericyte and endothelial cell communication can compromise the integrity of the blood-brain barrier (BBB), leading to neurovascular dysfunction and the development of neurological disorders. However, the evaluation of microvessel RNAs has been limited to tissue homogenates, with spatial visualization only available for protein targets. The aim of the present study is the development of an innovative microvessel isolation technique that is RNA-friendly for the purpose of coupling with in situ hybridization RNAscope analysis. RNA-friendly microvessel isolation combined with RNAscope analysis enables the visualization of cell-specific RNA within the spatial and histological context of the BBB. Using this approach, we have gained valuable insights into the structural and functional differences associated with the microvessels of 5xFAD mice, a mouse model of Alzheimer's disease (AD). RNAscope analysis revealed a decrease in pericytes from microvessels isolated from 5xFAD mice in comparison to wild-type mice. Additionally, the microvessels of 5xFAD mice exhibited an increase in TYRO protein tyrosine kinase binding protein (TYROBP) mRNA expression. These findings significantly advance our understanding of neurovascular interactions and hold great promise for guiding the development of targeted therapeutic interventions. This innovative approach enables visualization of cell RNA while preserving the spatial and histological context of the BBB, shedding light on the mechanisms underlying neurovascular unit communication.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sarah Schmidlin
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Destiny Tiburcio
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Minseon Park
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Institute of Physiotherapy and Health Sciences, the Blood-Brain Barrier Research Center, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
33
|
Zemke NR, Lee S, Mamde S, Yang B, Berchtold N, Maximiliano Garduño B, Indralingam HS, Bartosik WM, Lau PK, Dong K, Yang A, Tani Y, Chen C, Zeng Q, Ajith V, Tong L, Seng C, Li D, Wang T, Xu X, Ren B. Epigenetic and 3D genome reprogramming during the aging of human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618338. [PMID: 39463924 PMCID: PMC11507755 DOI: 10.1101/2024.10.14.618338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Age-related cognitive decline is associated with altered physiology of the hippocampus. While changes in gene expression have been observed in aging brain, the regulatory mechanisms underlying these changes remain underexplored. We generated single-nucleus gene expression, chromatin accessibility, DNA methylation, and 3D genome data from 40 human hippocampal tissues spanning adult lifespan. We observed a striking loss of astrocytes, OPC, and endothelial cells during aging, including astrocytes that play a role in regulating synapses. Microglia undergo a dramatic switch from a homeostatic state to a primed inflammatory state through DNA methylome and 3D genome reprogramming. Aged cells experience erosion of their 3D genome architecture. Our study identifies age-associated changes in cell types/states and gene regulatory features that provide insight into cognitive decline during human aging.
Collapse
Affiliation(s)
- Nathan R. Zemke
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Seoyeon Lee
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Sainath Mamde
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Bing Yang
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Nicole Berchtold
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
- Immunis Inc, 18301 Von Karman Ave; Irvine, CA, USA
| | - B. Maximiliano Garduño
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Hannah S. Indralingam
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Weronika M. Bartosik
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Pik Ki Lau
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Keyi Dong
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Amanda Yang
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Yasmine Tani
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Chumo Chen
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Qiurui Zeng
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
| | - Varun Ajith
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Liqi Tong
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
| | - Chanrung Seng
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine; St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine; St. Louis, MO, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine; Irvine, CA, USA
- The Center for Neural Circuit Mapping, University of California; Irvine, CA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine; La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego School of Medicine; La Jolla, CA, USA
| |
Collapse
|
34
|
Wang J, Lv C, Wei X, Li F. Molecular mechanisms and therapeutic strategies for ferroptosis and cuproptosis in ischemic stroke. Brain Behav Immun Health 2024; 40:100837. [PMID: 39228970 PMCID: PMC11369453 DOI: 10.1016/j.bbih.2024.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Ischemic stroke, as one of the most severe and prevalent neurological disorders, poses a significant threat to the health and quality of life of affected individuals. Stemming from the obstruction of blood flow, ischemic stroke, leads to cerebral tissue hypoxia and ischemia, instigating a cascade of pathophysiological changes that markedly exacerbate neuronal damage and may even culminate in cell death. In recent years, emerging research has increasingly focused on novel cell death mechanisms such as ferroptosis and cuproptosis. Mounting evidence underscores the independent roles of ferroptosis and cuproptosis in ischemic stroke. This review aims to elucidate potential cross-regulatory mechanisms between ferroptosis and cuproptosis, exploring their regulatory roles in ischemic stroke. The objective is to provide targeted therapeutic intervention strategies.
Collapse
Affiliation(s)
- Jing Wang
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
- Bengbu Medical College, Anhui, China
| | - Cunming Lv
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Wei
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Feng Li
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
| |
Collapse
|
35
|
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
|
36
|
Shaw BC, Anders VR, Tinkey RA, Habean ML, Brock OD, Frostino BJ, Williams JL. Immunity impacts cognitive deficits across neurological disorders. J Neurochem 2024; 168:3512-3535. [PMID: 37899543 PMCID: PMC11056485 DOI: 10.1111/jnc.15999] [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: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
Cognitive deficits are a common comorbidity with neurological disorders and normal aging. Inflammation is associated with multiple diseases including classical neurodegenerative dementias such as Alzheimer's disease (AD) and autoimmune disorders such as multiple sclerosis (MS), in which over half of all patients experience some form of cognitive deficits. Other degenerative diseases of the central nervous system (CNS) including frontotemporal lobe dementia (FTLD), and Parkinson's disease (PD) as well as traumatic brain injury (TBI) and psychological disorders like major depressive disorder (MDD), and even normal aging all have cytokine-associated reductions in cognitive function. Thus, there is likely commonality between these secondary cognitive deficits and inflammation. Neurological disorders are increasingly associated with substantial neuroinflammation, in which CNS-resident cells secrete cytokines and chemokines such as tumor necrosis factor (TNF)α and interleukins (ILs) including IL-1β and IL-6. CNS-resident cells also respond to a wide variety of cytokines and chemokines, which can have both direct effects on neurons by changing the expression of ion channels and perturbing electrical properties, as well as indirect effects through glia-glia and immune-glia cross-talk. There is significant overlap in these cytokine and chemokine expression profiles across diseases, with TNFα and IL-6 strongly associated with cognitive deficits in multiple disorders. Here, we review the involvement of various cytokines and chemokines in AD, MS, FTLD, PD, TBI, MDD, and normal aging in the absence of dementia. We propose that the neuropsychiatric phenotypes observed in these disorders may be at least partially attributable to a dysregulation of immunity resulting in pathological cytokine and chemokine expression from both CNS-resident and non-resident cells.
Collapse
Affiliation(s)
- Benjamin C. Shaw
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Victoria R. Anders
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Maria L. Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - Orion D. Brock
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin J. Frostino
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- College of Science, University of Notre Dame, South Bend, IN, USA
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
37
|
Greene C, Rebergue N, Fewell G, Janigro D, Godfrin Y, Campbell M, Lemarchant S. NX210c drug candidate peptide strengthens mouse and human blood-brain barriers. Fluids Barriers CNS 2024; 21:76. [PMID: 39334382 PMCID: PMC11438064 DOI: 10.1186/s12987-024-00577-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Alterations of blood-brain barrier (BBB) and blood-spinal cord barrier have been documented in various animal models of neurodegenerative diseases and in patients. Correlations of these alterations with functional deficits suggest that repairing barriers integrity may represent a disease-modifying approach to prevent neuroinflammation and neurodegeneration induced by the extravasation of blood components into the parenchyma. Here, we screened the effect of a subcommissural organ-spondin-derived peptide (NX210c), known to promote functional recovery in several models of neurological disorders, on BBB integrity in vitro and in vivo. METHODS In vitro, bEnd.3 endothelial cell (EC) monolayers and two different primary human BBB models containing EC, astrocytes and pericytes, in static and microfluidic conditions, were treated with NX210c (1-100 µM), or its vehicle, for 4 h and up to 5 days. Tight junction (TJ) protein levels, permeability to dextrans and transendothelial electrical resistance (TEER) were evaluated. In vivo, young and old mice (3- and 21-month-old, respectively) were treated daily intraperitoneally with NX210c at 10 mg/kg or its vehicle for 5 days and their brains collected at day 6 to measure TJ protein levels by immunohistochemistry. RESULTS NX210c induced an increase in claudin-5 protein expression after 24-h and 72-h treatments in mouse EC. Occludin level was also increased after a 24-h treatment. Accordingly, NX210c decreased by half the permeability of EC to a 40-kDa FITC-dextran and increased TEER. In the human static BBB model, NX210c increased by ∼ 25% the TEER from 3 to 5 days. NX210c also increased TEER in the human 3D dynamic BBB model after 4 h, which was associated with a reduced permeability to a 4-kDa FITC-dextran. In line with in vitro results, after only 5 days of daily treatments in mice, NX210c restored aging-induced reduction of claudin-5 and occludin levels in the hippocampus, and also in the cortex for occludin. CONCLUSIONS In summary, we have gathered preclinical data showing the capacity of NX210c to strengthen BBB integrity. Through this property, NX210c holds great promises of being a disease-modifying treatment for several neurological disorders with high unmet medical needs.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | | | | | | - Yann Godfrin
- Axoltis Pharma, 60 avenue Rockefeller, Lyon, 69008, France
- Godfrin Life-Sciences, Caluire-et-Cuire, 69300, France
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | |
Collapse
|
38
|
Taheri S, Prestopnik J, Rosenberg GA. Barriers of the CNS transfer rate dynamics in patients with vascular cognitive impairment and dementia. Front Aging Neurosci 2024; 16:1462302. [PMID: 39385834 PMCID: PMC11461252 DOI: 10.3389/fnagi.2024.1462302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Background Advances in in vivo MRI techniques enable cerebral barrier transfer rates (K trans ) measurement in patients with vascular cognitive impairment and dementia (VCID). However, a consensus has not been reached on the dynamic contribution and importance of cerebral barrier abnormalities to the differential diagnosis of dementia subtypes. Our goal was to investigate the dynamics of blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) K trans in patients with VCID longitudinally and determine the effect of aging. Methods We studied subjects at two time points over two years; they were 65.5 years of age (SD = 15.94, M/F = 24/14) at the first visit. We studied 38 patients, 18 of whom had two visits. We calculated the BBB and BCSFB K trans with dynamic contrast-enhanced T1 MR, and we used 1H-MR spectroscopy to measure N-acetylaspartate (NAA) levels in the white matter as a marker of injury. In addition, we measured CSF levels of active-matrix metalloproteinase-3 (MMP3) as an inflammatory biomarker to aid in patient clustering. Results Longitudinal BBB measurements revealed variable dynamic behavior: after two years, the BBB K trans increased in 55% of patients and decreased in the remaining 45% unpredictably. We did not find a significant linear model of BBB K trans versus age for VCID. For healthy controls, the model was K trans = 0.0014 + 0.0002 × age, which was significant (p = 0.046). VCID patients showed a reduction in BCSFB K trans compared to healthy controls (p = 0.01). Combining NAA, CSF MMP3, and K trans in a clustering analysis separated patients into groups. Conclusion These results suggest that BBB K trans in VCID is dynamic and BCSFB K trans reduced by age. By combining inflammatory biomarkers with BBB K trans data, it is possible to separate VCID patients into distinct groups with different underlying pathologies.
Collapse
Affiliation(s)
- Saeid Taheri
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, United States
- Center for Functional and Molecular Imaging, University of South Florida (USF) Heart Institute, Tampa, FL, United States
| | | | - Gary A. Rosenberg
- Center for Memory and Aging, Albuquerque, NM, United States
- Department of Neurology, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
39
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
40
|
Kim SY, Cheon J. Senescence-associated microvascular endothelial dysfunction: A focus on the blood-brain and blood-retinal barriers. Ageing Res Rev 2024; 100:102446. [PMID: 39111407 DOI: 10.1016/j.arr.2024.102446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The blood-brain barrier (BBB) and blood-retinal barrier (BRB) constitute critical physiochemical interfaces, precisely orchestrating the bidirectional communication between the brain/retina and blood. Increased permeability or leakage of these barriers has been demonstrably linked to age-related vascular and parenchymal damage. While it has been suggested that the gradual aging process may coincide with disruptions in these barriers, this phenomenon is significantly exacerbated in individuals with age-related neurodegenerative disorders (ARND). This review focuses on the microvascular endothelium, a key constituent of BBB and BRB, highlighting the impact of endothelial senescence on barrier dysfunction and exploring recent discoveries regarding core pathways implicated in its breakdown. Subsequently, we address the "vascular senescence hypothesis" for ARND, with a particular emphasis on Alzheimer's disease and age-related macular degeneration, centered on endothelial senescence. Finally, we discuss potential senotherapeutic strategies targeting barrier dysfunction.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea; Research Institute of Medical Science, Konkuk University, Republic of Korea; IBST, Konkuk University, Republic of Korea.
| | - Jaejoung Cheon
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea
| |
Collapse
|
41
|
Arshavsky YI. Autoimmune hypothesis of Alzheimer's disease: unanswered question. J Neurophysiol 2024; 132:929-942. [PMID: 39163023 DOI: 10.1152/jn.00259.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since β-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
42
|
Ma YZ, Cao JX, Zhang YS, Su XM, Jing YH, Gao LP. T Cells Trafficking into the Brain in Aging and Alzheimer's Disease. J Neuroimmune Pharmacol 2024; 19:47. [PMID: 39180590 DOI: 10.1007/s11481-024-10147-5] [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: 10/20/2022] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
The meninges, choroid plexus (CP) and blood-brain barrier (BBB) are recognized as important gateways for peripheral immune cell trafficking into the central nervous system (CNS). Accumulation of peripheral immune cells in brain parenchyma can be observed during aging and Alzheimer's disease (AD). However, the mechanisms by which peripheral immune cells enter the CNS through these three pathways and how they interact with resident cells within the CNS to cause brain injury are not fully understood. In this paper, we review recent research on T cells recruitment in the brain during aging and AD. This review focuses on the possible pathways through which T cells infiltrate the brain, the evidence that T cells are recruited to the brain, and how infiltrating T cells interact with the resident cells in the CNS during aging and AD. Unraveling these issues will contribute to a better understanding of the mechanisms of aging and AD from the perspective of immunity, and hopefully develop new therapeutic strategies for brain aging and AD.
Collapse
Affiliation(s)
- Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
43
|
Yang Y, Rao T, Wei S, Cheng J, Zhan Y, Lin T, Chen J, Zhong X, Jiang Y, Yang S. Role of inflammatory cytokines and the gut microbiome in vascular dementia: insights from Mendelian randomization analysis. Front Microbiol 2024; 15:1398618. [PMID: 39247699 PMCID: PMC11380139 DOI: 10.3389/fmicb.2024.1398618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/10/2024] [Indexed: 09/10/2024] Open
Abstract
Background Both inflammatory cytokines and the gut microbiome are susceptibility factors for vascular dementia (VaD). The trends in the overall changes in the dynamics of inflammatory cytokines and in the composition of the gut microbiome are influenced by a variety of factors, making it difficult to fully explain the different effects of both on the different subtypes of VaD. Therefore, this Mendelian randomization (MR) study identified the inflammatory cytokines and gut microbiome members that influence the risk of developing VaD and their causal effects, and investigated whether inflammatory cytokines are gut microbiome mediators affecting VaD. Methods We obtained pooled genome-wide association study (GWAS) data for 196 gut microbiota and 41 inflammatory cytokines and used GWAS data for six VaD subtypes, namely, VaD (mixed), VaD (multiple infarctions), VaD (other), VaD (subcortical), VaD (sudden onset), and VaD (undefined). We used the inverse-variance weighted (IVW) method as the primary MR analysis method. We conducted sensitivity analyses and reverse MR analyses to examine reverse causal associations, enhancing the reliability and stability of the conclusions. Finally, we used multivariable MR (MVMR) analysis to assess the direct causal effects of inflammatory cytokines and the gut microbiome on the risk of VaD, and performed mediation MR analysis to explore whether inflammatory factors were potential mediators. Results Our two-sample MR study revealed relationships between the risk of six VaD subtypes and inflammatory cytokines and the gut microbiota: 7 inflammatory cytokines and 14 gut microbiota constituents were positively correlated with increased VaD subtype risk, while 2 inflammatory cytokines and 11 gut microbiota constituents were negatively correlated with decreased VaD subtype risk. After Bonferroni correction, interleukin-18 was correlated with an increased risk of VaD (multiple infarctions); macrophage migration inhibitory factor was correlated with an increased risk of VaD (sudden onset); interleukin-4 was correlated with a decreased risk of VaD (other); Ruminiclostridium 6 and Bacillales were positively and negatively correlated with the risk of VaD (undefined), respectively; Negativicutes and Selenomonadales were correlated with a decreased risk of VaD (mixed); and Melainabacteria was correlated with an increased risk of VaD (multiple infarctions). Sensitivity analyses revealed no multilevel effects or heterogeneity and no inverse causality between VaD and inflammatory cytokines or the gut microbiota. The MVMR results further confirmed that the causal effects of Negativicutes, Selenomonadales, and Melainabacteria on VaD remain significant. Mediation MR analysis showed that inflammatory cytokines were not potential mediators. Conclusion This study helps us to better understand the pathological mechanisms of VaD and suggests the potential value of targeting increases or decreases in inflammatory cytokines and gut microbiome members for VaD prevention and intervention.
Collapse
Affiliation(s)
- Yihan Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ting Rao
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| | - Sheng Wei
- Department of General Practice, The Second Affiliated Hospital of Wannan Medical College, Anhui, China
| | - Jing Cheng
- Fujian Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| | - Ying Zhan
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Teng Lin
- The First Clinical Medical College, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jincheng Chen
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| | - Xiaoling Zhong
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yijing Jiang
- Fujian Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| | - Shanli Yang
- Fujian Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| |
Collapse
|
44
|
Yong HYF, Batty NJ, Tottenham I, Koch M, Camara-Lemarroy CR. Soluble adhesion molecules: Cognitive worsening biomarkers in primary progressive multiple sclerosis? J Neuroimmunol 2024; 393:578384. [PMID: 38850673 DOI: 10.1016/j.jneuroim.2024.578384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/26/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Affiliation(s)
- Heather Y F Yong
- Department of Clinical Neurosciences, University of Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicholas J Batty
- Department of Clinical Neurosciences, University of Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Isabelle Tottenham
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Koch
- Department of Clinical Neurosciences, University of Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carlos R Camara-Lemarroy
- Department of Clinical Neurosciences, University of Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
45
|
Liu R, Collier JM, Abdul-Rahman NH, Capuk O, Zhang Z, Begum G. Dysregulation of Ion Channels and Transporters and Blood-Brain Barrier Dysfunction in Alzheimer's Disease and Vascular Dementia. Aging Dis 2024; 15:1748-1770. [PMID: 38300642 PMCID: PMC11272208 DOI: 10.14336/ad.2023.1201] [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: 09/13/2023] [Accepted: 12/01/2023] [Indexed: 02/02/2024] Open
Abstract
The blood-brain barrier (BBB) plays a critical role in maintaining ion and fluid homeostasis, essential for brain metabolism and neuronal function. Regulation of nutrient, water, and ion transport across the BBB is tightly controlled by specialized ion transporters and channels located within its unique cellular components. These dynamic transport processes not only influence the BBB's structure but also impact vital signaling mechanisms, essential for its optimal function. Disruption in ion, pH, and fluid balance at the BBB is associated with brain pathology and has been implicated in various neurological conditions, including stroke, epilepsy, trauma, and neurodegenerative diseases such as Alzheimer's disease (AD). However, knowledge gaps exist regarding the impact of ion transport dysregulation on BBB function in neurodegenerative dementias. Several factors contribute to this gap: the complex nature of these conditions, historical research focus on neuronal mechanisms and technical challenges in studying the ion transport mechanisms in in vivo models and the lack of efficient in vitro BBB dementia models. This review provides an overview of current research on the roles of ion transporters and channels at the BBB and poses specific research questions: 1) How are the expression and activity of key ion transporters altered in AD and vascular dementia (VaD); 2) Do these changes contribute to BBB dysfunction and disease progression; and 3) Can restoring ion transport function mitigate BBB dysfunction and improve clinical outcomes. Addressing these gaps will provide a greater insight into the vascular pathology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ruijia Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jenelle M Collier
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | - Okan Capuk
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Zhongling Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Gulnaz Begum
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
46
|
Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024; 13:e2303180. [PMID: 38430211 PMCID: PMC11338747 DOI: 10.1002/adhm.202303180] [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: 09/20/2023] [Revised: 02/22/2024] [Indexed: 03/03/2024]
Abstract
The blood-brain barrier (BBB) is a highly controlled microenvironment that regulates the interactions between cerebral blood and brain tissue. Due to its selectivity, many therapeutics targeting various neurological disorders are not able to penetrate into brain tissue. Pre-clinical studies using animals and other in vitro platforms have not shown the ability to fully replicate the human BBB leading to the failure of a majority of therapeutics in clinical trials. However, recent innovations in vitro and ex vivo modeling called organs-on-chips have shown the potential to create more accurate disease models for improved drug development. These microfluidic platforms induce physiological stressors on cultured cells and are able to generate more physiologically accurate BBBs compared to previous in vitro models. In this review, different approaches to create BBBs-on-chips are explored alongside their application in modeling various neurological disorders and potential therapeutic efficacy. Additionally, organs-on-chips use in BBB drug delivery studies is discussed, and advances in linking brain organs-on-chips onto multiorgan platforms to mimic organ crosstalk are reviewed.
Collapse
Affiliation(s)
- Atharva R. Mulay
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Jihyun Hwang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218
| |
Collapse
|
47
|
Bertolini M, Clark D. Periodontal disease as a model to study chronic inflammation in aging. GeroScience 2024; 46:3695-3709. [PMID: 37285008 PMCID: PMC11226587 DOI: 10.1007/s11357-023-00835-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/20/2023] [Indexed: 06/08/2023] Open
Abstract
Periodontal disease is a chronic inflammatory condition that results in the destruction of the teeth supporting tissues, eventually leading to the loss of teeth and reduced quality of life. In severe cases, periodontal disease can limit proper nutritional intake, cause acute pain and infection, and cause a withdrawal from social situations due to esthetic and phonetic concerns. Similar to other chronic inflammatory conditions, periodontal disease increases in prevalence with age. Research into what drives periodontal disease pathogenesis in older adults is contributing to our general understanding of age-related chronic inflammation. This review will present periodontal disease as an age-related chronic inflammatory disease and as an effective geroscience model to study mechanisms of age-related inflammatory dysregulation. The current understanding of the cellular and molecular mechanisms that drive inflammatory dysregulation as a function of age will be discussed with a focus on the major pathogenic immune cells in periodontal disease, which include neutrophils, macrophages, and T cells. Research in the aging biology field has shown that the age-related changes in these immune cells result in the cells becoming less effective in the clearance of microbial pathogens, expansion of pathogenic subpopulations, or an increase in pro-inflammatory cytokine secretions. Such changes can be pathogenic and contribute to inflammatory dysregulation that is associated with a myriad of age-related disease including periodontal disease. An improved understanding is needed to develop better interventions that target the molecules or pathways that are perturbed with age in order to improve treatment of chronic inflammatory conditions, including periodontal disease, in older adult populations.
Collapse
Affiliation(s)
- Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Daniel Clark
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
48
|
Cunha S, Bicker J, Sereno J, Falcão A, Fortuna A. Blood brain barrier dysfunction in healthy aging and dementia: Why, how, what for? Ageing Res Rev 2024; 99:102395. [PMID: 38950867 DOI: 10.1016/j.arr.2024.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/03/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
The blood brain barrier (BBB) is an indispensable structure that maintains the central nervous system (CNS) microenvironment for a correct neuronal function. It is composed by highly specialized microvessels, surrounded by astrocytes, pericytes, neurons and microglia cells, which tightly control the influx and efflux of substances to the brain parenchyma. During aging, the BBB becomes impaired, and it may contribute to the development of neurodegenerative and neurological disorders including Alzheimer's disease and other dementias. Restoring the BBB can be a strategy to prevent disease onset and development, reducing the symptoms of these conditions. This work critically reviews the major mechanisms underlying BBB breakdown in healthy and unhealthy aging, as well as biomarkers and methodologies that accurately assess its impairment. Complementarily, potential therapeutic targets are discussed as new strategies to restore the normal function of the BBB in aging.
Collapse
Affiliation(s)
- Susana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - José Sereno
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
49
|
Xie DY, Lin M, Luo YM, Dong L, Wei Y, Gao JM, Zhu YZ, Gong QH. Trilobatin suppresses aging-induced cognitive impairment by targeting SIRT2: Involvement of remodeling gut microbiota to mediate the brain-gut axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155744. [PMID: 38763011 DOI: 10.1016/j.phymed.2024.155744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Aging is associated with learning and memory disorder, affecting multiple brain areas, especially the hippocampus. Previous studies have demonstrated trilobatin (TLB), as a natural food additive, can extend the life of Caenorhabditis elegans and exhibit neuroprotection in Alzheimer's disease mice. However, the possible significance of TLB in anti-aging remains elusive. PURPOSE This study aimed to delve into the physiological mechanism by which TLB ameliorated aging-induced cognitive impairment in senescence-accelerated mouse prone 8 (SAMP8) mice. METHODS 6-month-old SAMP8 mice were administrated with TLB (5, 10, 20 mg/kg/day, i.g.) for 3 months. The therapeutic effect of TLB on aging-induced cognitive impairment was assessed in mice using behavioral tests and aging score. The gut microbiota composition in fecal samples was analyzed by metagenomic analysis. The protective effects of TLB on blood-brain barrier (BBB) and intestinal barrier were detected by transmission electron microscope, H&E staining and western blot (WB) assay. The inhibitive effects of TLB on inflammation in brain and intestine were assessed using immunofluorescence, WB and ELISA assay. Molecular docking and surface plasma resonance (SPR) assay were utilized to investigate interaction between TLB and sirtuin 2 (SIRT2). RESULTS Herein, the findings exhibited TLB mitigated aging-induced cognitive impairment, neuron injury and neuroinflammation in hippocampus of aged SAMP8 mice. Moreover, TLB treatment repaired imbalance of gut microbiota in aged SAMP8 mice. Furthermore, TLB alleviated the damage to BBB and intestinal barrier, concomitant with reducing the expression of SIRT2, phosphorylated levels of c-Jun NH2 terminal kinases (JNK) and c-Jun, and expression of MMP9 protein in aged SAMP8 mice. Molecular docking and SPR unveiled TLB combined with SIRT2 and down-regulated SIRT2 protein expression. Mechanistically, the potential mechanism of SIRT2 in TLB that exerted anti-aging effect was validated in vitro. As expected, SIRT2 deficiency attenuated phosphorylated level of JNK in HT22 cells treated with d-galactose. CONCLUSION These findings reveal, for the first time, SIRT2-mediated brain-gut barriers contribute to aging and aging-related diseases, and TLB can rescue aging-induced cognitive impairment by targeting SIRT2 and restoring gut microbiota disturbance to mediate the brain-gut axis. Overall, this work extends the potential application of TLB as a natural food additive in aging-related diseases.
Collapse
Affiliation(s)
- Dian-You Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China
| | - Mu Lin
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China; Guizhou Aerospace Hospital, Zunyi 563000, China
| | - Yun-Mei Luo
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China
| | - Lan Dong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China
| | - Yu Wei
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Jian-Mei Gao
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China
| | - Yi Zhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China.
| | - Qi-Hai Gong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Macau SAR, Taipa, 999078, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, 6 Xuefu West Road, Zunyi, Guizhou 563006, China.
| |
Collapse
|
50
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [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: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|