1
|
Bassalo D, Matthews SG, Bloise E. The canine blood-brain barrier in health and disease: focus on brain protection. Vet Q 2025; 45:12-32. [PMID: 39791202 PMCID: PMC11727060 DOI: 10.1080/01652176.2025.2450041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/18/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025] Open
Abstract
This review examines the role of the canine blood-brain barrier (BBB) in health and disease, focusing on the impact of the multidrug resistance (MDR) transporter P-glycoprotein (P-gp) encoded by the ABCB1/MDR1 gene. The BBB is critical in maintaining central nervous system homeostasis and brain protection against xenobiotics and environmental drugs that may be circulating in the blood stream. We revise key anatomical, histological and functional aspects of the canine BBB and examine the role of the ABCB1/MDR1 gene mutation in specific dog breeds that exhibit reduced P-gp activity and disrupted drug brain pharmacokinetics. The review also covers factors that may disrupt the canine BBB, including the actions of aging, canine cognitive dysfunction, epilepsy, inflammation, infection, traumatic brain injury, among others. We highlight the critical importance of this barrier in maintaining central nervous system homeostasis and protecting against xenobiotics and conclude that a number of neurological-related diseases may increase vulnerability of the BBB in the canine species and discuss its profound impacts on canine health.
Collapse
Affiliation(s)
- Dimitri Bassalo
- Especialização em Farmacologia, Departamento de Farmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G. Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Department of Obstetrics & Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
2
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Yan R, Zhang L, Chen Y, Zheng Y, Xu P, Xu Z. Therapeutic potential of gut microbiota modulation in epilepsy: A focus on short-chain fatty acids. Neurobiol Dis 2025; 209:106880. [PMID: 40118219 DOI: 10.1016/j.nbd.2025.106880] [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: 11/17/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025] Open
Abstract
According to the criteria established by the International League Against Epilepsy (ILAE), epilepsy is defined as a disorder characterized by at least two unprovoked seizures occurring more than 24 h apart. Its pathogenesis is closely related to various physiological and pathological factors. Advances in high-throughput metagenomic sequencing have increasingly highlighted the role of gut microbiota dysbiosis in epilepsy. Short-chain fatty acids (SCFAs), the major metabolites of the gut microbiota and key regulators of the gut-brain axis, support physiological homeostasis through multiple mechanisms. Recent studies have indicated that SCFAs not only regulate seizures by maintaining intestinal barrier integrity and modulating intestinal immune responses, but also affect the structure and function of the blood-brain barrier (BBB) and regulate neuroinflammation. This review, based on current literatures, explores the relationship between SCFAs and epilepsy, emphasizing how SCFAs affect epilepsy by modulating the intestinal barrier and BBB. In-depth studies on SCFAs may reveal their therapeutic potential and inform the development of gut microbiota-targeted epilepsy treatments.
Collapse
Affiliation(s)
- Rong Yan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Linhai Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ya Chen
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongsu Zheng
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Function and Brain Disease Prevention and Treatment of Guizhou Province, Zunyi, China.
| |
Collapse
|
4
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
5
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
6
|
Mohammadi S, Dolatshahi M, Rahmani F, Raji CA. Altered Clearance in Alzheimer's Disease and Cerebral Amyloid Angiopathy. Neuroimaging Clin N Am 2025; 35:277-286. [PMID: 40210383 DOI: 10.1016/j.nic.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
With our focus on Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA), the authors herein provided a comprehensive overview of impaired amyloid beta and tau clearance pathways observed through advanced neuroimaging techniques such as dynamic contrast-enhanced MR imaging, arterial spin labeling, phase contrast MR imaging, PET, and functional MR imaging. The findings suggest the role of impaired degradation clearance, blood-brain barrier clearance, perivascular clearance, glymphatic system clearance, and cerebrospinal fluid dynamics in AD and CAA pathogenesis.
Collapse
Affiliation(s)
- Soheil Mohammadi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
7
|
Rodriguez-Lopez A, Esteban D, Domínguez-Romero AN, Gevorkian G. Tg-SwDI transgenic mice: A suitable model for Alzheimer's disease and cerebral amyloid angiopathy basic research and preclinical studies. Exp Neurol 2025; 387:115189. [PMID: 39978567 DOI: 10.1016/j.expneurol.2025.115189] [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: 11/22/2024] [Revised: 01/17/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and the most frequent cause of dementia. Characteristic features observed in the brain of AD patients are the accumulation of amyloid beta peptide (Aβ) aggregates, neurofibrillary tangles (NFT) composed of hyperphosphorylated Tau protein, neuronal and synaptic loss, and elevated levels of oxidative stress and inflammatory markers. Cerebral amyloid angiopathy (CAA) is another common cause of cognitive decline characterized by the accumulation of Aβ in the cerebral vasculature. The precise overlapping pathogenic mechanisms underlying the co-occurrence of AD and CAA are not very well understood. However, vascular dysfunction observed at early stages is considered a key phenomenon. Tg-SwDI transgenic mice expressing human Aβ precursor protein (AβPP) harboring the Swedish K670N/M671L and vasculotropic Dutch/Iowa E693Q/D694N mutations in the brain have been extensively used to study many pathological features observed in AD/CAA patients and to design biomarkers and therapeutic strategies. The present review summarizes studies addressing different features mimicking human disease in Tg-SwDI mice: parenchymal and cerebral vascular amyloid accumulation, neuroinflammation, complement overactivation, cerebrovascular, mitochondrial and GABAergic system dysfunction, altered NO synthesis, circadian rhythm disruptions, lead exposure effect, among others. Also, reports that evaluated anti-Aβ and anti-inflammatory strategies and compounds capable of delaying or reversing vascular dysfunction and the impairment of GABAergic transmission in Tg-SwDI mice are analyzed. This review may help researchers determine this model's appropriateness for future studies of a particular mechanism or a novel treatment protocol.
Collapse
Affiliation(s)
- Adrian Rodriguez-Lopez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Daniel Esteban
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Allan Noé Domínguez-Romero
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Apartado Postal 70228, Cuidad Universitaria, CDMX, CP 04510, Mexico.
| |
Collapse
|
8
|
Song C, Yang J, Gu Z. Latest developments of microphysiological systems (MPS) in aging-related and geriatric diseases research: A review. Ageing Res Rev 2025; 107:102728. [PMID: 40058462 DOI: 10.1016/j.arr.2025.102728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Aging is a gradual and irreversible process accompanied by the decline in tissue function and a significantly increased risk of various aging-related and geriatric diseases. Especially in the paradoxical context of accelerated global aging and the widespread emergence of pandemics, aging-related and geriatric diseases have become leading causes of individual mortality and disability, drawing increasing attention from researchers and investors alike. Despite the utility of current in vitro systems and in vivo animal models for studying aging, these approaches are limited by insurmountable inherent constraints. In response, microphysiological systems (MPS), leveraging advances in tissue engineering and microfluidics, have emerged as highly promising platforms. MPS are capable of replicating key features of the tissue microenvironment within microfabricated devices, offering biomimetic tissue culture conditions that enhance the in vitro simulation of intact or precise human body structure and function. This capability improves the predictability of clinical trial outcomes while reducing time and cost. In this review, we focus on recent advancements in MPS used to study age-related and geriatric diseases, with particular emphasis on the application of organoids and organ-on-a-chip technologies in understanding cardiovascular diseases, cerebrovascular diseases, neurodegenerative diseases, fibrotic diseases, locomotor and sensory degenerative disorders, and rare diseases. And we aim to provide readers with critical guidelines and an overview of examples for modeling age-related and geriatric diseases using MPS, exploring mechanisms, treatments, drug screening, and other subsequent applications, from a physiopathological perspective, emphasizing the characteristic of age-related and geriatric diseases and their established correlations with the aging process. We also discuss the limitations of current models and propose future directions for MPS in aging research, highlighting the potential of interdisciplinary approaches to address unresolved challenges in the field.
Collapse
Affiliation(s)
- Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jiachen Yang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
9
|
McIntyre CW, Jain A. Dialysis and cognitive impairment. Nat Rev Nephrol 2025:10.1038/s41581-025-00960-3. [PMID: 40275017 DOI: 10.1038/s41581-025-00960-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/26/2025]
Abstract
People with chronic kidney disease who require maintenance dialysis characteristically experience accelerated and aggravated cognitive decline compared with those with advanced kidney disease who are not receiving this form of kidney replacement therapy. This effect is inadequately appreciated, but of crucial importance to patients, their carers and the health-care systems that support them. Although many of the comorbid conditions prevalent in this patient population have the potential to affect brain structure and function, an evolving body of evidence indicates that the dialysis therapy itself has a central role in the pathophysiology of progressive cognitive impairment. Both haemodialysis and peritoneal dialysis are associated with structural and functional changes in the brain that can lead to characteristic short-term symptoms, such as headache, confusion, delirium and brain fog, as well as long-term reductions in cognitive functional ability. Here, we explore the mechanisms, both established and putative, underlying these effects and consider approaches to addressing this issue with both single and complex therapeutic interventions.
Collapse
Affiliation(s)
- Chris W McIntyre
- Lilibeth Caberto Kidney Clinical Research Unit, Lawson Health Research Institute, London, Ontario, Canada.
- Departments of Medicine, Medical Biophysics and Paediatrics, Western University, London, Ontario, Canada.
| | - Arsh Jain
- Lilibeth Caberto Kidney Clinical Research Unit, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
10
|
Garbarino VR, Palavicini JP, Melendez J, Barthelemy NR, He Y, Kautz TF, Lopez-Cruzan M, Mathews JJ, Xu P, Zhang B, Saliba A, Ragi N, Sharma K, Mason D, Johnson S, Hendrix S, Craft S, Petersen RC, Espindola-Netto JM, Xue A, Tchkonia T, Kirkland JL, Salardini A, Musi N, Bateman RJ, Gonzales MM, Orr ME. Evaluation of exploratory fluid biomarkers from a phase 1 senolytic trial in mild Alzheimer's disease. Neurotherapeutics 2025:e00591. [PMID: 40274471 DOI: 10.1016/j.neurot.2025.e00591] [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: 10/15/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Senescent cell accumulation contributes to the progression of age-related disorders including Alzheimer's disease (AD). Clinical trials focused on cellular senescence are in early stages and have yet to establish reliable outcome measures reflecting senescent cell burden or response to senolytics, therapeutics that clear senescent cells. Results from the first open-label trial of senolytics, dasatinib plus quercetin (D + Q), in older adults (N = 5) with early AD demonstrated central nervous system penetration of dasatinib and favorable safety and tolerability. Herein, we present exploratory analyses of senescence and AD-associated analytes in blood, cerebrospinal fluid (CSF) and urine from this study in effort to guide biomarker development for future senolytic trials. Immunoassays, mass spectrometry and transcriptomics were performed and changes in analyte levels were assessed from baseline to post-treatment using paired t-tests. Targeted cytokine and chemokine analyses revealed increases in plasma fractalkine and MMP-7 and CSF IL-6 from baseline to post-treatment. Mass spectrometry indicated stable levels of amyloid β and tau proteins in CSF, unchanged urinary metabolites, and modest treatment-associated lipid profile changes. Targeted transcriptomic analysis of peripheral blood mononuclear cells indicated downregulation of inflammatory genes including FOS, FOSB, IL1β, IL8, JUN, JUNB, PTGS2. The levels and treatment responses of the analytes identified here may help inform trial design and outcomes for senolytic studies. Independent validation will be necessary to develop standardized biomarker panels across senolytic trials for AD. ClinicalTrials.gov: NCT04063124.
Collapse
Affiliation(s)
- Valentina R Garbarino
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Juan Pablo Palavicini
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Justin Melendez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Nicolas R Barthelemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marisa Lopez-Cruzan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Julia J Mathews
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Peng Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Afaf Saliba
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | - Suzanne Craft
- Department of Internal Medicine Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | - Ailing Xue
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Arash Salardini
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nicolas Musi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Mitzi M Gonzales
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Miranda E Orr
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA; St Louis VA Medical Center, St Louis, MO, USA.
| |
Collapse
|
11
|
Cheemala A, Kimble AL, Burrage EN, Helming SB, Tyburski JD, Leclair NK, Omar OM, Zuberi AR, Murphy M, Jellison ER, Reese B, Hu X, Lutz CM, Yan R, Murphy PA. Amyotrophic lateral sclerosis and frontotemporal dementia mutation reduces endothelial TDP-43 and causes blood-brain barrier defects. SCIENCE ADVANCES 2025; 11:eads0505. [PMID: 40238886 PMCID: PMC12002129 DOI: 10.1126/sciadv.ads0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Mutations in the TARDBP gene encoding TDP-43 protein are linked to loss of function in neurons and familial frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). We recently identified reduced nuclear TDP-43 in capillary endothelial cells (ECs) of donors with ALS-FTD. Because blood-brain barrier (BBB) permeability increases in ALS-FTD, we postulated that reduced nuclear TDP-43 in ECs might contribute. Here, we show that nuclear TDP-43 is reduced in ECs of mice with an ALS-FTD-associated mutation in TDP-43 (TardbpG348C) and that this leads to cell-autonomous loss of junctional complexes and BBB integrity. Targeted excision of TDP-43 in brain ECs recapitulates BBB defects and loss of junctional complexes and ultimately leads to fibrin deposition, gliosis, phospho-Tau accumulation, and impaired memory and social interaction. Transcriptional changes in TDP-43-deficient ECs resemble diseased brain ECs. These data show that nuclear loss of TDP-43 in brain ECs disrupts the BBB and causes hallmarks of FTD.
Collapse
Affiliation(s)
- Ashok Cheemala
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Amy L. Kimble
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Emily N. Burrage
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stephen B. Helming
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Jordan D. Tyburski
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nathan K. Leclair
- MD/PhD Program, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Omar M. Omar
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Aamir R. Zuberi
- Rare Disease Translational Center and Technology Evaluation and Development Laboratory, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Melissa Murphy
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Evan R. Jellison
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Bo Reese
- Center for Genome Innovation, University of Connecticut, Storrs, CT, USA
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Cathleen M. Lutz
- Rare Disease Translational Center and Technology Evaluation and Development Laboratory, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Patrick A. Murphy
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
12
|
Altahrawi AY, James AW, Shah ZA. The Role of Oxidative Stress and Inflammation in the Pathogenesis and Treatment of Vascular Dementia. Cells 2025; 14:609. [PMID: 40277934 DOI: 10.3390/cells14080609] [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: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Vascular dementia (VaD) is a heterogeneous group of brain disorders caused by cerebrovascular pathologies and the second most common cause of dementia, accounting for over 20% of cases and posing an important global health concern. VaD can be caused by cerebral infarction or injury in critical brain regions, including the speech area of the dominant hemisphere or arcuate fasciculus of the dominant hemisphere, leading to notable cognitive impairment. Although the exact causes of dementia remain multifactorial and complex, oxidative stress (reactive oxygen species), neuroinflammation (TNFα, IL-6, and IL-1β), and inflammasomes are considered central mechanisms in its pathology. These conditions contribute to neuronal damage, synaptic dysfunction, and cognitive decline. Thus, antioxidants and anti-inflammatory agents have emerged as potential therapeutic targets in dementia. Recent studies emphasize that cerebrovascular disease plays a dual role: first, as a primary cause of cognitive impairment and then as a contributor to the manifestation of dementia driven by other factors, such as Alzheimer's disease and other neurodegenerative conditions. This comprehensive review of VaD focuses on molecular mechanisms and their consequences. We provided up-to-date knowledge about epidemiology, pathophysiological mechanisms, and current therapeutic approaches for VaD.
Collapse
Affiliation(s)
- Aseel Y Altahrawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
13
|
Vrillon A, Ashton NJ, Bouaziz-Amar E, Mouton-Liger F, Cognat E, Dumurgier J, Lilamand M, Karikari TK, Prevot V, Zetterberg H, Blennow K, Paquet C. Dissection of blood-brain barrier dysfunction through CSF PDGFRβ and amyloid, tau, neuroinflammation, and synaptic CSF biomarkers in neurodegenerative disorders. EBioMedicine 2025; 115:105694. [PMID: 40239464 PMCID: PMC12020895 DOI: 10.1016/j.ebiom.2025.105694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) dysfunction is an early event in neurodegenerative disorders. Pericytes are key cells for BBB maintenance. Upon pericyte injury, the platelet-derived growth factor receptor-β (PDGFRβ) is released in the cerebrospinal fluid (CSF). The relation of CSF PDGFRβ with markers of amyloid pathology, neuroinflammation, and axonal and synaptic damage across dementia remains unclear. METHODS Retrospectively, we quantified CSF PDGFRβ and CSF core Alzheimer's disease (AD), astrocytic (GFAP), microglial (sTREM 2, YKL-40), axonal (NfL), and synaptic (GAP-43, neurogranin) biomarkers in 210 patients from the Cognitive Neurology Centre, Paris, France, including n = 23 neurological controls (NC), n = 84 patients with mild cognitive impairment (MCI) [AD, n = 41; non-AD, n = 43], and n = 103 patients with dementia (AD, n = 73; non-AD, n = 30). FINDINGS Comparing clinical stages, CSF PDGFRβ levels were increased at the MCI stage (Cohen's d = 0.55 [CI95% 0.066, 1.0], P = 0.025) compared with NC. Non-AD MCI displayed higher levels than controls (Cohen's d = 0.74 [CI95% 0.22, 1.3], P = 0.042). No association was observed with CSF Aβ42/Aβ40 ratio but with p-tau 181 (β = 0.102 [CI95% 0.027, 0.176], P = 0.0080) and t-tau levels (β = 0.133 [0.054, 0.213], P = 0.0010). CSF PDGFRβ levels were positively associated with CSF neuroinflammation and synaptic markers levels. Higher CSF PDGFRβ levels were associated with lower MMSE scores at MCI (β = -1.23 [CI95% -2.33, -0.260], P = 0.015) and dementia stages (β = -2.24 [CI95% -3.62, -0.85], P = 0.0020). CSF neuroinflammation biomarkers mediated the association of CSF PDGFRβ with neurodegeneration and synaptic integrity markers. INTERPRETATION CSF PDGFRβ, a candidate biomarker of BBB dysfunction, is increased in the early stages of neurodegenerative disorders, in association with neuroinflammation and axonal and synaptic damage. FUNDING Association des Anciens Internes des Hôpitaux de Paris, Edmond de Rothschild Program, Fondation Vaincre Alzheimer, Demensförbundet, Gamla Tjänarinnor, Anna-Lisa och Bror Björnssons Stiftelse.
Collapse
Affiliation(s)
- Agathe Vrillon
- Cognitive Neurology Centre, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France; INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France; University of California San Francisco, San Francisco, CA, USA.
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Banner Alzheimer's Institute and University of Arizona, Phoenix, AZ, USA; Banner Sun Health Research Institute, Sun City, AZ 85351, USA; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Elodie Bouaziz-Amar
- INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France; Biochemistry Department, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | | | - Emmanuel Cognat
- Cognitive Neurology Centre, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France; INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Julien Dumurgier
- Cognitive Neurology Centre, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Matthieu Lilamand
- Cognitive Neurology Centre, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France; INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, DISTALZ, Lille, France
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Hong Kong Centre for Neurodegenerative Diseases, Clear Water Bay, Hong Kong Special Administrative Region of China; Wisconsin Alzheimer's Disease Research Centre, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Centre, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Claire Paquet
- Cognitive Neurology Centre, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France; INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| |
Collapse
|
14
|
Hou JH, Jiang DM, Chu M, Wu LY. Blood-brain barrier biomarkers modulate the associations of peripheral immunity with Alzheimer's disease. Transl Psychiatry 2025; 15:138. [PMID: 40210862 PMCID: PMC11986039 DOI: 10.1038/s41398-025-03347-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025] Open
Abstract
The association between peripheral immunity and Alzheimer's disease (AD) has been increasingly recognized, but the underlying mechanisms are still unclear. We used multiple linear regression models to explore the association between peripheral immune biomarkers / blood-brain barrier (BBB)-related biomarkers and AD biomarkers. And we used causal mediation analysis with 10,000 bootstrapped iterations to investigate the functions of BBB-related biomarkers in mediating the associations between peripheral immune biomarkers and AD pathology, cerebral atrophy degree, as well as cognitive function. A total of 543 participants (38.7% female, mean age of 74.8 years) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were involved. Neutrophils percent (NEU%), lymphocytes percent (LYM%), neutrophils / lymphocytes (NLR), and chemotactic factor-3 (CCL26) were significantly associated with cerebrospinal fluid (CSF) β-amyloid-42 (Aβ-42), phosphorylated-tau (P-tau), total tau (T-tau)/Aβ-42 and P-tau/Aβ-42, the associations of NEU% with AD pathology were mediated by CCL26 (proportion: 18-24%; p < 0.05). NEU%, LYM%, NLR, CCL26, CD40 and matrix metalloproteinase-10 (MMP10) were significantly associated with whole brain, hippocampal volume, middle temporal lobe (MTL) volume, and entorhinal cortex (EC) thickness, the associations of peripheral immune biomarkers with cerebral atrophy degree were mediated by BBB-related biomarkers (proportion: 7-17%; p < 0.05). NEU%, LYM%, NLR, CCL26, CD40 and MMP10 were significantly associated with global cognition, executive function, memory function, immediate recall, and delayed recall, the associations of peripheral immune biomarkers with cognitive function were mediated by BBB-related biomarkers (proportion: 9-24%; p < 0.05). This study suggests that peripheral immunity may influence AD through influencing BBB function, providing a more robust and comprehensive evidence chain for the potential role of inflammation in AD.
Collapse
Affiliation(s)
- Jia-Hui Hou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - De-Ming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Chu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Li-Yong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Moussiopoulou J, Yakimov V, Roell L, Rauchmann BS, Toth H, Melcher J, Jäger I, Lutz I, Kallweit MS, Papazov B, Boudriot E, Seelos K, Dehsarvi A, Campana M, Raabe F, Maurus I, Löhrs L, Brendel M, Stöcklein S, Falkai P, Hasan A, Group CW, Franzmeier N, Keeser D, Wagner E. Higher blood-brain barrier leakage in schizophrenia-spectrum disorders: A comparative dynamic contrast-enhanced magnetic resonance imaging study with healthy controls. Brain Behav Immun 2025; 128:256-265. [PMID: 40194748 DOI: 10.1016/j.bbi.2025.04.003] [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: 09/11/2024] [Revised: 02/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruptions are presumed to be implicated in schizophrenia-spectrum disorders (SSDs). Previous studies focused on cerebrospinal fluid (CSF) markers, which are imprecise for detecting subtle BBB disruption. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) enables sensitive investigation of subtle BBB leakage in vivo, yet remains unexplored in SSD research. We hypothesized higher leakage in SSDs compared to healthy controls (HCs), indicating a clinical sub-phenotype. METHODS Forty-one people with SSDs and forty age- and sex-matched HCs were included in this cross-sectional study employing DCE-MRI, clinical characterization, cognitive assessment, blood and CSF analyses. The volume transfer constant Ktrans, calculated using the Patlak method to estimate the contrast agent transfer between blood and extravascular space, was compared between groups to detect differences in BBB leakage. RESULTS Individuals with SSDs showed higher BBB leakage compared to HCs in a widespread pattern, in brain regions typically affected in SSDs. No significant association was detected between leakage and measures of cognition, symptom severity, peripheral inflammation markers and albumin CSF/serum ratio. CONCLUSIONS This is the first study to date reporting BBB leakage in SSDs compared to HCs in multiple brain regions implicated in the disorder. These findings provide insights into disease mechanisms, highlighting the need for further investigation into the role of the BBB in SSDs.
Collapse
Affiliation(s)
- Joanna Moussiopoulou
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; DZPG (German Center for Mental Health), partner site München, Augsburg, Germany; Neuroimaging Core Unit Munich, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany.
| | - Vladislav Yakimov
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany; DZPG (German Center for Mental Health), partner site München, Augsburg, Germany
| | - Lukas Roell
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; DZPG (German Center for Mental Health), partner site München, Augsburg, Germany; Neuroimaging Core Unit Munich, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Institute of Neuroradiology, LMU University Hospital, LMU Munich, Marchioninistr. 15 81377 Munich, Germany
| | - Hannah Toth
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Julian Melcher
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Iris Jäger
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Isabel Lutz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Marcel S Kallweit
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Boris Papazov
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Department of Radiology, LMU University Hospital, LMU Munich, Germany
| | - Emanuel Boudriot
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Klaus Seelos
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Marchioninistr. 15 81377 Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich Germany
| | - Mattia Campana
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Florian Raabe
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Isabel Maurus
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Lisa Löhrs
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Germany; DZNE (German Center for Neurodegenerative Diseases) Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, LMU University Hospital, LMU Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; DZPG (German Center for Mental Health), partner site München, Augsburg, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, University of Augsburg 86156 Augsburg, Germany; DZPG (German Center for Mental Health), partner site München, Augsburg, Germany
| | - Cdp Working Group
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, University of Augsburg 86156 Augsburg, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Evidence-based psychiatry and psychotherapy, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Neuroimaging Core Unit Munich, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nußbaumstraße 7, 80336 Munich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, University of Augsburg 86156 Augsburg, Germany; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Germany; Evidence-based psychiatry and psychotherapy, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| |
Collapse
|
16
|
Feng S, Lin Y, Wang R, Fang X, Shi J, Gu X, Yu Y. Elevated lipoprotein-associated phospholipase A2 as a biomarker for cognitive impairment in chronic kidney disease. Int Urol Nephrol 2025:10.1007/s11255-025-04469-3. [PMID: 40167984 DOI: 10.1007/s11255-025-04469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aimed to examine the relationship between serum lipoprotein-associated phospholipase A2 (Lp-PLA2) levels and cognitive impairment (CI) in patients with chronic kidney disease (CKD). METHODS A total of 125 patients with CKD treated in the Department of Nephrology, The Second Affiliated Hospital of Nantong University from July 2022 to May 2023 were selected and divided into observation group (44 patients with CI) and control group (81 patients with normal cognitive function). Multivariate logistic regression analysis was performed to analyze the risk factors of CI, and Spearman rank correlation analysis was used to analyze the correlation between serum Lp-PLA2 and Montreal Cognitive Assessment Scale (MoCA) score. The truncation value of Lp-PLA2 in CKD patients with CI was analyzed by receiver operating characteristic curve (ROC). RESULTS Serum Lp-PLA2 levels were significantly elevated in the observation group compared to the control group (P < 0.05). The area under the ROC curve for Lp-PLA2 was 0.849, with a cutoff value of 232 ng/mL for identifying CI in patients with CKD. Lp-PLA2 levels were independently associated with CI in patients with CKD (odds ratio [OR] = 0.988, 95% confidence interval [CI]: 0.982-0.993, P < 0.001). CONCLUSION Elevated serum Lp-PLA2 levels serve as an independent risk factor for CI in patients with CKD.
Collapse
Affiliation(s)
- Sujuan Feng
- Department of Nephrology, Affiliated Hospital 2 of Nantong University, Nantong, 226000, China
| | - Yanfeng Lin
- Hemodialysis center, Affiliated Haimen Hospital of Xinglin College, Nantong University, Nantong, 226000, China
| | - Rong Wang
- Department of Nephrology, Affiliated Hospital 2 of Nantong University, Nantong, 226000, China
| | - Xingxing Fang
- Department of Nephrology, Affiliated Hospital 2 of Nantong University, Nantong, 226000, China
| | - Jiaqi Shi
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Xiameng Gu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Yang Yu
- Department of Nephrology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), No. 881 of Yonghe Road, Nantong, 226000, China.
| |
Collapse
|
17
|
Wagner CA, Massy ZA, Capasso G, Mattace-Raso F, Pepin M, Bobot M, Zoccali C, Ferreira AC, Hoorn EJ, Imenez Silva PH, Unwin RJ, Pesic V. Translational research on cognitive impairment in chronic kidney disease. Nephrol Dial Transplant 2025; 40:621-631. [PMID: 39400744 DOI: 10.1093/ndt/gfae229] [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: 07/27/2024] [Indexed: 10/15/2024] Open
Abstract
Cognitive decline is common in patients with acute or chronic kidney disease. Several areas of brain function can be affected, including short- and long-term memory, attention and inhibitory control, sleep, mood, eating control and motor function. Cognitive decline in kidney disease shares risk factors with cognitive dysfunction in people without kidney disease, such as diabetes, high blood pressure, sedentary lifestyle and unhealthy diet. However, additional kidney-specific risk factors may contribute, such as uremic toxins, electrolyte imbalances, chronic inflammation, acid-base disorders or endocrine dysregulation. Traditional and kidney-specific risk factors may interact to cause damage to the blood-brain barrier, induce vascular damage in the brain and cause neurotoxicity or neuroinflammation. Here, we discuss recent insights into the pathomechanisms of cognitive decline from animal models and novel avenues for prevention and therapy. We focus on a several areas that influence cognition: blood-brain barrier disruption, the role of skeletal muscle, physical activity and the endocrine factor irisin, and the emerging therapeutic role of sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists. Taken together, these studies demonstrate the importance of animal models in providing a mechanistic understanding of this complex condition and their potential to explain the mechanisms of novel therapies.
Collapse
Affiliation(s)
- Carsten A Wagner
- Department of Physiology and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Ziad A Massy
- Clinical Epidemiology, Inserm Unit 1018, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA), Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt/Paris, France
| | - Giovambattista Capasso
- Biogem, Research Institute for Biology and Molecular Genetics, Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania, Luigi Vanvitelli, Napoli, Italy
| | - Francesco Mattace-Raso
- Department of Internal Medicine, Division of Geriatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marion Pepin
- Clinical Epidemiology, Inserm Unit 1018, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) Villejuif, France
- Geriatric Department, Ambroise Paré University Hospital, APHP, Versailles Saint-Quentin-en-Yvelines University (UVSQ), Boulogne-Billancourt, France
| | - Mickaël Bobot
- Aix-Marseille Univ, C2VN, INSERM 1263, INRAE 1260, CERIMED, Marseille, France
| | - Carmine Zoccali
- Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale, Grande Ospedale Metropolitano, c/o Nefrologia, Reggio Calabria, Italy
| | - Ana C Ferreira
- Unidade Local de Saúde de São José - Hospital Curry Cabral, Nephrology Department, Lisbon, Portugal
- Nova Medical School - Nephrology, Lisbon, Portugal
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Hypertension, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pedro H Imenez Silva
- Department of Internal Medicine, Division of Nephrology and Hypertension, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert J Unwin
- Department of Renal Medicine, Royal Free Hospital Trust, University College London (UCL), London, UK
| | - Vesna Pesic
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
18
|
Xia Y, Chen J, Dong P, Zhang L, Ding Y, Ding W, Han X, Wang X, Li D. Embryonic 6:2 Fluorotelomer Alcohol Exposure Disrupts the Blood‒Brain Barrier by Causing Endothelial‒to‒Mesenchymal Transition in the Male Mice. Mol Neurobiol 2025; 62:4203-4220. [PMID: 39417922 DOI: 10.1007/s12035-024-04540-7] [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: 07/03/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
6:2 Fluorotelomer alcohol (6:2 FTOH) is a raw material used in the manufacture of short-chain poly- and perfluoroalkyl substances. Our previous study revealed that gestational exposure to 6:2 FTOH can impair blood‒brain barrier (BBB) function in offspring, accompanied by anxiety-like behavior and learning memory deficits. The aim of this study was to further investigate the specific mechanism by which maternal exposure to 6:2 FTOH resulted in impaired BBB function in offspring mice. Pregnant mice were orally administered different doses of 6:2 FTOH (0, 5, 25, and 125 mg/kg/day) from gestation day 8.5 until delivery. These results confirmed that maternal 6:2 FTOH exposure impaired BBB function and disrupted the brain immune microenvironment. Subsequent investigations revealed that endothelial-to-mesenchymal transition (EndMT) in the cerebral microvascular endothelium of offspring may be the mechanism mediating functional disruption of the BBB. Mechanistic studies revealed that exposure to 6:2 FTOH upregulated ETS proto-oncogene 1 (ETS1) expression via the tumor necrosis factor-α/extracellular signal-regulated kinase 1/2 signaling pathway, which mediated disturbances in energy metabolism, leading to impaired actin dynamics and subsequently triggering the EndMT phenotype. This is the first finding indicating that gestational 6:2 FTOH exposure caused functional impairment of the BBB through ETS1-mediated EndMT in cerebral microvascular endothelial cells, potentially providing novel insight into the environmental origins of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yunhui Xia
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Junhan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Ping Dong
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Luqing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Yibing Ding
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
- Translational Medicine Core Facilities, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Weidong Ding
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, Jiangsu, China.
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
19
|
Kumar S, Earnest T, Yang B, Kothapalli D, Aschenbrenner AJ, Hassenstab J, Xiong C, Ances B, Morris J, Benzinger TLS, Gordon BA, Payne P, Sotiras A. Analyzing heterogeneity in Alzheimer disease using multimodal normative modeling on imaging-based ATN biomarkers. Alzheimers Dement 2025; 21:e70143. [PMID: 40235115 PMCID: PMC12000228 DOI: 10.1002/alz.70143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Previous studies have applied normative modeling on a single neuroimaging modality to investigate Alzheimer disease (AD) heterogeneity. We employed a deep learning-based multimodal normative framework to analyze individual-level variation across ATN (amyloid-tau-neurodegeneration) imaging biomarkers. METHODS We selected cross-sectional discovery (n = 665) and replication cohorts (n = 430) with available T1-weighted magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). Normative modeling estimated individual-level abnormal deviations in amyloid-positive individuals compared to amyloid-negative controls. Regional abnormality patterns were mapped at different clinical group levels to assess intra-group heterogeneity. An individual-level disease severity index (DSI) was calculated using both the spatial extent and magnitude of abnormal deviations across ATN. RESULTS Greater intra-group heterogeneity in ATN abnormality patterns was observed in more severe clinical stages of AD. Higher DSI was associated with worse cognitive function and increased risk of disease progression. DISCUSSION Subject-specific abnormality maps across ATN reveal the heterogeneous impact of AD on the brain. HIGHLIGHTS Normative modeling examined AD heterogeneity across multimodal imaging biomarkers. Heterogeneity in spatial patterns of gray matter atrophy, amyloid, and tau burden. Higher within-group heterogeneity for AD patients at advanced dementia stages. Patient-specific metric summarized extent of neurodegeneration and neuropathology. Metric is a marker of poor brain health and can monitor risk of disease progression.
Collapse
Affiliation(s)
- Sayantan Kumar
- Department of Computer Science and EngineeringWashington University in St LouisSaint LouisMissouriUSA
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Tom Earnest
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Braden Yang
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Deydeep Kothapalli
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | | - Jason Hassenstab
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - Chengie Xiong
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Beau Ances
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - John Morris
- Department of NeurologyWashington University School of MedicineSt louisMissouriUSA
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Philip Payne
- Department of Computer Science and EngineeringWashington University in St LouisSaint LouisMissouriUSA
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Aristeidis Sotiras
- Institute for InformaticsData Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | |
Collapse
|
20
|
Zhang X, Zhang H, Liu Z, Huang T, Yi R, Ma Z, Gao Y. Salidroside improves blood-brain barrier integrity and cognitive function in hypobaric hypoxia mice by inhibiting microglia activation through GSK3β. Phytother Res 2025; 39:1808-1825. [PMID: 39364585 DOI: 10.1002/ptr.8264] [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: 06/29/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 10/05/2024]
Abstract
Salidroside, an active component found in Rhodiola rosea L., has emerged as a potential therapeutic agent for the prevention and treatment of hypoxic brain injury, while the precise target and mechanism of salidroside were remain unclear. The study utilized techniques such as network pharmacology, transcriptome sequencing to investigate the mechanism and target of salidroside in regulating blood-brain barrier (BBB) function to protect hypoxic brain injury in vivo. Utilized macromolecular docking and molecular biology techniques to explore the molecular mechanism of salidroside in alleviating brain injury induced by hypoxia in BV2 cell model. The results show that salidroside alleviated the learning and memory dysfunction and pathological injury in mice exposed to hypobaric hypoxia, reduced brain water content and attenuate the inflammatory response and oxidative stress, effectively reversed S100β in serum and promoted the repair of BBB. GSK3β is an important therapeutic target of salidroside in the treatment of hypoxic cognitive impairment, and salidroside can specifically bind GSK3β in the ATP binding pocket, inducing the phosphorylation of GSK3β, targeting downstream Nrf-2 to regulate microglia activity, promoting the accumulation of β-catenin, thereby inhibiting microglial activation, improving the BBB integrity injury and achieving a neuroprotective effect. This study demonstrates that salidroside can inhibit the activation of microglia by inducing GSK3β phosphorylation, achieve neuroprotective effects and alleviate learning and memory dysfunction in hypobaric hypoxia mice. This study provides a theoretical basis for the development of salidroside and the clinical application of Rhodiola rosea L.
Collapse
Affiliation(s)
- Xianxie Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Huiting Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Zuoxu Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Tianke Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Ru Yi
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Zengchun Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| |
Collapse
|
21
|
Hardowar LD, Sheavyn M, McTernan PG, Bates DO, Hulse RP. Angiotensin II type 1 receptor activation induces dorsal horn capillary constriction and pain hypersensitivity. THE JOURNAL OF PAIN 2025; 30:105384. [PMID: 40174732 DOI: 10.1016/j.jpain.2025.105384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Vascular disturbance is a key factor in the development of neurological disease, with reduced integrity of the capillary network in the dorsal horn implicated in activation of nociceptive neural circuits and induction of pain states. Pericytes regulate capillary health and tone, with pericyte dysfunction in cerebral tissue associated with neurodegenerative disorders. Our work demonstrates that spinal cord nociceptive processing is influenced by angiotensin II type 1 (AT1) receptor mediated capillary constriction. Intravital imaging of the mouse spinal cord demonstrated angiotensin II induced cessation of spinal cord capillary perfusion. Intrathecal administration of angiotensin II induced narrowing of capillary diameter, which was accompanied by mechanical allodynia and heat hyperalgesia in adult male and female mice. Angiotensin II mediated reduction of spinal cord blood flow and pericyte activation, was prevented by AT1 receptor inhibition via losartan treatment. Losartan prevented angiotensin II induced pain. Integrity of dorsal horn capillary endothelium was protected by co-treatment with losartan preventing angiotensin II induced loss of CD31 immunoreactivity. This investigation demonstrates that AT1 regulates the dorsal horn capillary network and is fundamental in modulating nociceptive processing and perception of pain. Here we identify a novel cellular and mechanistic target for the induction of pain hypersensitivity. PERSPECTIVE: Intrathecally delivered Angiotensin II induced mechanical and heat hypersensitivity in male and female mice. Capillary constriction in the dorsal horn was induced by Angiotensin II treatment and led to degeneration of the endothelium. Angiotensin II induced pericyte activation was Angiotensin II type 1 receptor dependent.
Collapse
Affiliation(s)
- Lydia D Hardowar
- School of Science and Technology, Nottingham Trent University, UK
| | - Matthew Sheavyn
- School of Science and Technology, Nottingham Trent University, UK
| | | | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, Centre for Cancer Science, Biodiscovery Institute, University of Nottingham, Nottingham, UK; Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Richard P Hulse
- School of Science and Technology, Nottingham Trent University, UK.
| |
Collapse
|
22
|
Chung KJ, Abdelhafez YG, Spencer BA, Jones T, Tran Q, Nardo L, Chen MS, Sarkar S, Medici V, Lyo V, Badawi RD, Cherry SR, Wang G. Quantitative PET imaging and modeling of molecular blood-brain barrier permeability. Nat Commun 2025; 16:3076. [PMID: 40159510 PMCID: PMC11955546 DOI: 10.1038/s41467-025-58356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Neuroimaging of blood-brain barrier permeability has been instrumental in identifying its broad involvement in neurological and systemic diseases. However, current methods evaluate the blood-brain barrier mainly as a structural barrier. Here we developed a non-invasive positron emission tomography method in humans to measure the blood-brain barrier permeability of molecular radiotracers that cross the blood-brain barrier through its molecule-specific transport mechanism. Our method uses high-temporal resolution dynamic imaging and kinetic modeling for multiparametric imaging and quantification of the blood-brain barrier permeability-surface area product of molecular radiotracers. We show, in humans, our method can resolve blood-brain barrier permeability across three radiotracers and demonstrate its utility in studying brain aging and brain-body interactions in metabolic dysfunction-associated steatotic liver inflammation. Our method opens new directions to effectively study the molecular permeability of the human blood-brain barrier in vivo using the large catalogue of available molecular positron emission tomography tracers.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Quyen Tran
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Souvik Sarkar
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Valentina Medici
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Victoria Lyo
- Department of Surgery, University of California Davis Health, Sacramento, CA, USA
- Center for Alimentary and Metabolic Sciences, University of California Davis Health, Sacramento, CA, USA
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Simon R Cherry
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA.
| |
Collapse
|
23
|
Wang X, Zhong H, Kong X, Wei H, Li B. Artesunate alleviated hippocampal neuron pyroptosis by down-regulating NLRP3 in rats with cerebral small vessel disease. Metab Brain Dis 2025; 40:160. [PMID: 40138000 PMCID: PMC11947036 DOI: 10.1007/s11011-025-01590-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Our study aims to investigate the potential of artesunate (ART) in improving learning and memory function by down-regulating NLRP3 and consequently affecting pyroptosis levels in the brains of rats with cerebral small vessel disease (CSVD). Initially, Sprague-Dawley (SD) rats were randomly assigned to five groups: the solvent sham operation group, solvent model group, low-dose ART (ARTL) group, medium-dose ART (ARTM) group, and high-dose ART group (ARTH). CSVD rat models were established through bilateral common carotid artery occlusion (BCCAO). Subsequently, the rats were further divided into four groups: the empty plasmid control group (shNC) and three groups receiving NLRP3-shRNA interference plasmids (shNLRP3-1, shNLRP3-2, shNLRP3-3). We recorded animal behaviors and stained nerve cell changes. Hippocampal expression levels of Caspase-1, cleaved caspase-1, IL-18, IL-1β, GSDMD-N, β-actin, and NLRP3 were evaluated in each group. Our findings revealed that ART ameliorated cognitive dysfunction and brain tissue injury in CSVD rats. Moreover, expression levels of cleaved caspase-1, IL-18, IL-1β, GSDMD-N, and NLRP3 in the hippocampus were significantly reduced in the shNLRP3 group, resulting in improved cognitive function in these rats. These results suggest that NLRP3 could be a potential therapeutic target in CSVD development in rats, and modulating its expression might mitigate pathological alterations associated with CSVD. Subsequently, lipopolysaccharide (LPS) was injected into the tail vein, and inflammatory factors in peripheral blood of rats were found to be increased, suggesting that the level of intracranial NLRP3 was increased. In addition, MWM experiment showed that after the increase of NLRP3 expression, the repair effect of ART on learning and memory dysfunction was weakened. ART may enhance cognitive impairment in CSVD rats by downregulating NLRP3 expression in the brain, thereby inhibiting neuronal cell pyroptosis in the hippocampus.
Collapse
Affiliation(s)
- Xiaokun Wang
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Hequan Zhong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xiangyu Kong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Hongqiao Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530022, China.
| | - Bing Li
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China.
| |
Collapse
|
24
|
Yilmaz SN, Steiner K, Marksteiner J, Faserl K, Sarg B, Humpel C. Novel Plasma Biomarkers for Alzheimer's Disease: Insights from Organotypic Brain Slice and Microcontact Printing Techniques. FRONT BIOSCI-LANDMRK 2025; 30:36257. [PMID: 40152394 DOI: 10.31083/fbl36257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a severe neurodegenerative disorder characterized by beta-amyloid plaques and tau neurofibrillary tangles. The diagnosis of AD is complex, with the analysis of beta-amyloid and tau in cerebrospinal fluid being a well-established diagnostic approach. However, currently no blood biomarkers have been identified or validated for clinical use. In the present study, we will identify novel plasma biomarkers for AD using our well-established organotypic mouse brain slice model connected to microcontact prints. We hypothesize that AD plasma contains factors that affect endothelial cell migration and new vessel formation. METHODS In the present study, plasma from human patients is microcontact printed and connected to mouse brain slices. After 4 weeks in culture, laminin+ and lectin+ endothelial cells (ECs) and vessels are analyzed by immunostaining techniques. The most promising samples were processed by differential mass spectrometry. RESULTS Our data show that AD plasma significantly increased the migration length of laminin+ and lectin+ ECs along the microcontact prints. Using differential mass spectrometry, we could identify three potential biomarkers: C-reactive protein, basigin, and trem-like transcript 1 protein. CONCLUSION Here we show that brain slices connected to human plasma prints allow the identification of novel human AD biomarkers with subsequent mass spectrometry. This technique represents a novel and innovative approach to translate research findings from mouse models to human applications.
Collapse
Affiliation(s)
- Sakir Necat Yilmaz
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, 33343 Mersin, Turkey
| | - Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
25
|
Li Y, Izhar T, Kanekiyo T. HDAC3 as an Emerging Therapeutic Target for Alzheimer's Disease and other Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04866-w. [PMID: 40126601 DOI: 10.1007/s12035-025-04866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the aged population. Histone acetylation is a major epigenetic mechanism linked to memory formation and cognitive function. Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone proteins. Although pan-HDAC inhibitors are effective in ameliorating AD phenotypes in preclinical models, they are associated with potential unfavorable adverse effects and barely translated into clinical trials. Therefore, the development of novel HDAC inhibitors with a well isoform-selectivity has been desired in AD drug discovery. Among various HDAC isoforms, HDAC3 is highly expressed in neurons and exhibits detrimental effects on synaptic plasticity and cognitive function. Moreover, HDAC3 provokes neuroinflammation and neurotoxicity and contributes to AD pathogenesis. In this review, we highlight HDAC3 as an attractive therapeutic target for disease-modifying therapy in AD. In addition, we discuss the therapeutic potential of HDAC3 inhibitors in other neurological disorders.
Collapse
Affiliation(s)
- Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Taha Izhar
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
26
|
Yang B, Teymur A, Tang C, Wu T. V-set and immunoglobulin domain containing 4 as a potential predictor of Alzheimer's disease and advanced aging. J Alzheimers Dis 2025:13872877251329463. [PMID: 40116682 DOI: 10.1177/13872877251329463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
BackgroundV-set and immunoglobulin domain containing 4 (VSIG4) emerges as a significant player in the immune system pathways. It has been previously identified as a potential hub gene for Alzheimer's disease (AD) and aging, underscoring its importance in understanding these conditions.ObjectiveThis study aimed to evaluate the diagnostic potential of serum VSIG4 and identify trends in serum VSIG4 in relationship with other biomarkers and neurological tests.MethodsELISA was used to measure the serum concentration of VSIG4 in AD, compared to healthy subjects. The relationship between VSIG4 levels and the age of the subjects, as well as other AD-related serum proteins and various measures of cognition was examined.ResultsVSIG4 was significantly elevated in the serum of AD patients compared to healthy controls (p = 0.0074). Significant correlations were identified between serum VSIG4 and other notable proteins related to AD and inflammation, such as total tau, neurofilament light (NfL), YKL-40, CD14, FABP3, and TNF-α. Significant correlations were also identified between VSIG4 concentration and the results of neurological tests.ConclusionsSerum VSIG4 may reflect neuroinflammation and altered lipid processing, affecting the cognitive performance of AD and aging.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Aygun Teymur
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
27
|
Ungvari A, Nyúl-Tóth Á, Patai R, Csik B, Gulej R, Nagy D, Shanmugarama S, Benyó Z, Kiss T, Ungvari Z, Csiszar A. Cerebromicrovascular senescence in vascular cognitive impairment: does accelerated microvascular aging accompany atherosclerosis? GeroScience 2025:10.1007/s11357-025-01621-w. [PMID: 40113668 DOI: 10.1007/s11357-025-01621-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Vascular cognitive impairment (VCI) is a leading cause of age-related cognitive decline, driven by cerebrovascular dysfunction and cerebral small vessel disease (CSVD). Emerging evidence suggests that cerebromicrovascular endothelial senescence plays an important role in the pathogenesis of VCI by promoting cerebral blood flow dysregulation, neurovascular uncoupling, blood-brain barrier (BBB) disruption, and the development of cerebral microhemorrhages (CMHs). This review explores the concept of cerebromicrovascular senescence as a continuum of vascular aging, linking macrovascular atherosclerosis with microvascular dysfunction. It examines the mechanisms by which endothelial senescence drives neurovascular pathology and highlights the impact of cardiovascular risk factors in accelerating these processes. We examine preclinical and clinical studies that provide compelling evidence that atherosclerosis-induced microvascular senescence exacerbates cognitive impairment. In particular, findings suggest that targeting senescent endothelial cells through senolytic therapy can restore cerebrovascular function and improve cognitive outcomes in experimental models of atherosclerosis. Given the growing recognition of microvascular senescence as a therapeutic target, further research is warranted to explore novel interventions such as senolytics, anti-inflammatory agents, and metabolic modulators. The development of circulating biomarkers of vascular senescence (e.g., senescence-associated secretory phenotype [SASP] components and endothelial-derived extracellular vesicles) could enable early detection and risk stratification in individuals at high risk for VCI. Additionally, lifestyle modifications, including the Mediterranean diet, hold promise for delaying endothelial senescence and mitigating cognitive decline. In conclusion, cerebromicrovascular senescence is a key mechanistic link between atherosclerosis and cognitive impairment. Addressing microvascular aging as a modifiable risk factor through targeted interventions offers a promising strategy for reducing the burden of VCI and preserving cognitive function in aging populations.
Collapse
Affiliation(s)
- Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Dorina Nagy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
| | - Santny Shanmugarama
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
| | - Tamas Kiss
- Cerebrovascular and Neurocognitive Diseases Research Group, HUN-REN, Semmelweis University, Budapest, Hungary
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Gao L, Wang B, Cui X, Xia L, Li X, Figueredo YN, Li D, Liu K, Wang H, Jin M. Neochlorogenic acid ameliorates Alzheimer's disease-like pathology via scavenging oxidative stress and restoring blood-brain barrier function in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111334. [PMID: 40122506 DOI: 10.1016/j.pnpbp.2025.111334] [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: 05/06/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease is the most widespread neurodegenerative disease characterized by insidious onset and slow progression. At present, most available medications serve to attenuate the progression of Alzheimer's disease with side effects and drug resistance. Neochlorogenic acid is a natural polyphenolic compound with excellent antioxidant properties. Based on zebrafish Alzheimer's disease model induced by AlCl3, we found that neochlorogenic acid significantly improved motor dysfunction, reduced brain cell apoptosis, and Aβ plaque. Because of antioxidant stress and improvement of blood-brain barrier dysfunction are important in treating Alzheimer's disease, we explored the interaction between these two mechanisms in alleviating the pathological course of Alzheimer's disease. Neochlorogenic acid inhibited the overproduction of reactive oxygen species, suppressed the gene expression encoding antioxidant-related proteins, and protected brain cell integrity while enhancing Nrf2, improving blood-brain barrier nerve resilience. Meanwhile, neochlorogenic acid attenuated blood-brain barrier dysfunction in Alzheimer's disease zebrafish by reducing blood hemoglobin leakage and upregulating the gene expression encoding blood-brain barrier endothelial cell-related proteins, resulting in reactive oxygen species in a controllable state. In conclusion, our research suggests that neochlorogenic acid ameliorates Alzheimer's disease-like pathology by inhibiting oxidative stress and restoring blood-brain barrier function, indicating that neochlorogenic acid may be a potential drug for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Li Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Baokun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Pharmacy, Qingdao University, Qing'dao 266003, Shandong Province, People's Republic of China
| | - Xiaotong Cui
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Lijie Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Xinjia Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Yanier Nuñez Figueredo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, La Habana CP10600, Cuba
| | - Dong Li
- R&D Department, Jinan Perfect Biological Technology Co., Ltd., Jinan 250101, Shandong Province, People's Republic of China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Haitao Wang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China.
| |
Collapse
|
29
|
Rust R, Sagare AP, Zhang M, Zlokovic BV, Kisler K. The blood-brain barrier as a treatment target for neurodegenerative disorders. Expert Opin Drug Deliv 2025:1-20. [PMID: 40096820 DOI: 10.1080/17425247.2025.2480654] [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: 10/11/2024] [Revised: 02/14/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a vascular endothelial membrane which restricts entry of toxins, cells, and microorganisms into the brain. At the same time, the BBB supplies the brain with nutrients, key substrates for DNA and RNA synthesis, and regulatory molecules, and removes metabolic waste products from brain to blood. BBB breakdown and/or dysfunction have been shown in neurogenerative disorders including Alzheimer's disease (AD). Current data suggests that these BBB changes may initiate and/or contribute to neuronal, synaptic, and cognitive dysfunction, and possibly other aspects of neurodegenerative processes. AREAS COVERED We first briefly review recent studies uncovering molecular composition of brain microvasculature and examine the BBB as a possible therapeutic target in neurodegenerative disorders with a focus on AD. Current strategies aimed at protecting and/or restoring altered BBB functions are considered. The relevance of BBB-directed approaches to improve neuronal and synaptic function, and to slow progression of neurodegenerative processes are also discussed. Lastly, we review recent advancements in drug delivery across the BBB. EXPERT OPINION BBB breakdown and/or dysfunction can significantly affect neuronal and synaptic function and neurodegenerative processes. More attention should focus on therapeutics to preserve or restore BBB functions when considering treatments of neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mingzi Zhang
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
30
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [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: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
31
|
Sorets AG, Schwensen KR, Francini N, Kjar A, Lyons S, Park JC, Palmer D, Abdulrahman AM, Cowell RP, Katdare KA, Hoogenboezem EN, Wang A, Dani N, Duvall CL, Lippmann ES. Intravenous lipid-siRNA conjugate mediates gene silencing at the blood-brain barrier and blood-CSF barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.642142. [PMID: 40166171 PMCID: PMC11957068 DOI: 10.1101/2025.03.14.642142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Barriers of the central nervous system (CNS), such as the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB), regulate the two-way exchange of material between the blood and CNS. These barriers pose a considerable challenge for efficacious delivery of intravenously administered therapies into the CNS, motivating exploration of their function and ways to modulate their properties. While the BBB and BCSFB can become dysfunctional in patients with chronic CNS diseases, few studies have focused on strategies for targeting these interfaces. Here, we showed that an intravenously administered albumin-binding lipid-siRNA conjugate was delivered to and silences genes within brain endothelial cells and choroid plexus epithelial cells, which comprise the BBB and BCSFB, respectively. A single intravenous dose of lipid-siRNA conjugate was delivered to ~100% of brain endothelial cells and major choroid plexus cell types, without any substantial delivery into brain parenchymal tissue. Sustained gene silencing was achieved in both brain endothelial cells (over two weeks) and bulk choroid plexus tissues (up to one month). Moreover, single cell RNA sequencing demonstrated gene knockdown in capillaries, venous endothelial cells, and choroid plexus epithelial cells without silencing genes in parenchymal cell populations. Collectively, this work establishes an effective nonviral framework to mediate gene inhibition in the brain barriers.
Collapse
Affiliation(s)
- Alexander G. Sorets
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Katrina R. Schwensen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sarah Lyons
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Joshua C. Park
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Dillon Palmer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Adam M. Abdulrahman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rebecca P. Cowell
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A. Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | | | - Angela Wang
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Neil Dani
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S. Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
32
|
Omar OMF, Kimble AL, Cheemala A, Tyburski JD, Pandey S, Wu Q, Reese B, Jellison ER, Hao B, Li Y, Yan R, Murphy PA. Endothelial TDP-43 depletion disrupts core blood-brain barrier pathways in neurodegeneration. Nat Neurosci 2025:10.1038/s41593-025-01914-5. [PMID: 40087396 DOI: 10.1038/s41593-025-01914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/05/2025] [Indexed: 03/17/2025]
Abstract
Endothelial cells (ECs) help maintain the blood-brain barrier but deteriorate in many neurodegenerative disorders. Here we show, using a specialized method to isolate EC and microglial nuclei from postmortem human cortex (92 donors, 50 male and 42 female, aged 20-98 years), that intranuclear cellular indexing of transcriptomes and epitopes enables simultaneous profiling of nuclear proteins and RNA transcripts at a single-nucleus resolution. We identify a disease-associated subset of capillary ECs in Alzheimer's disease, amyotrophic lateral sclerosis and frontotemporal degeneration. These capillaries exhibit reduced nuclear β-catenin and β-catenin-downstream genes, along with elevated TNF/NF-κB markers. Notably, these transcriptional changes correlate with the loss of nuclear TDP-43, an RNA-binding protein also depleted in neuronal nuclei. TDP-43 disruption in human and mouse ECs replicates these alterations, suggesting that TDP-43 deficiency in ECs is an important factor contributing to blood-brain barrier breakdown in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omar M F Omar
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA
| | - Amy L Kimble
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA
| | - Ashok Cheemala
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA
| | - Jordan D Tyburski
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA
| | - Swati Pandey
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA
| | - Qian Wu
- Department of Pathology, University of Connecticut Medical School, Farmington, CT, USA
| | - Bo Reese
- Center for Genome Innovation, University of Connecticut, Storrs, CT, USA
| | - Evan R Jellison
- Department of Immunology, University of Connecticut Medical School, Farmington, CT, USA
| | - Bing Hao
- Department of Molecular Biology and Biophysics, University of Connecticut Medical School, Farmington, CT, USA
| | - Yunfeng Li
- Department of Molecular Biology and Biophysics, University of Connecticut Medical School, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA
| | - Patrick A Murphy
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT, USA.
- Department of Immunology, University of Connecticut Medical School, Farmington, CT, USA.
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA.
| |
Collapse
|
33
|
Wagner CA, Frey-Wagner I, Ortiz A, Unwin R, Liabeuf S, Suzumoto Y, Iervolino A, Stasi A, Di Marzo V, Gesualdo L, Massy ZA. The role of the intestinal microbiome in cognitive decline in patients with kidney disease. Nephrol Dial Transplant 2025; 40:ii4-ii17. [PMID: 40080091 PMCID: PMC11905753 DOI: 10.1093/ndt/gfae253] [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: 08/15/2024] [Indexed: 03/15/2025] Open
Abstract
Cognitive decline is frequently seen in patients with chronic kidney disease (CKD). The causes of cognitive decline in these patients are likely to be multifactorial, including vascular disease, uraemic toxins, blood-brain barrier leakage, and metabolic and endocrine changes. Gut dysbiosis is common in patients with CKD and contributes to the increase in uraemic toxins. However, the gut microbiome modulates local and systemic levels of several metabolites such as short-chain fatty acids or derivatives of tryptophan metabolism, neurotransmitters, endocannabinoid-like mediators, bile acids, hormones such as glucagon-like peptide 1 (GLP1) or cholecystokinin (CCK). These factors can affect gut function, immunity, autonomic nervous system activity and various aspects of brain function. Key areas include blood-brain barrier integrity, nerve myelination and survival/proliferation, appetite, metabolism and thermoregulation, mood, anxiety and depression, stress and local inflammation. Alterations in the composition of the gut microbiota and the production of biologically active metabolites in patients with CKD are well documented and are favoured by low-fiber diets, elevated urea levels, sedentary lifestyles, slow stool transit times and polypharmacy. In turn, dysbiosis can modulate brain function and cognitive processes, as discussed in this review. Thus, the gut microbiome may contribute to alterations in cognition in patients with CKD and may be a target for therapeutic interventions using diet, prebiotics and probiotics.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center, University of Zurich, Switzerland
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, RICORS2040, Madrid, Spain
| | - Robert Unwin
- Department of Renal Medicine, University College London, London, UK
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Yoko Suzumoto
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Anna Iervolino
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- University of Campania “L. Vanvitelli”, Naples, Italy
| | - Alessandra Stasi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and INAF, Centre NUTRISS, Faculties of Medicine and Agriculture and Food Sciences, Université Laval, Québec City, Canada
- Joint International Research Unit for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (JIRU-MicroMeNu) between Université Laval Québec, Canada and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA) Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt, Paris, France
| |
Collapse
|
34
|
Hassanzadeh-Khanmiri M, Moshari A, Kheradmand R, Haghgouei T, Homaei M, Charsouei S, Mobed A. Nanomedicine: a cost-effective and powerful platform for managing neurodegenerative diseases. Metab Brain Dis 2025; 40:142. [PMID: 40067468 DOI: 10.1007/s11011-025-01564-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/28/2025] [Indexed: 03/26/2025]
Abstract
Neurodegenerative diseases (NDDs) are characterized by the chronic and progressive deterioration of the structure and function of the nervous system, imposing a significant burden on patients, their families, and society. These diseases have a gradual onset and continually worsen, making early diagnosis challenging. Current drugs on the market struggle to effectively cross the blood-brain barrier (BBB), leading to poor outcomes and limited therapeutic success. Consequently, there is an urgent need for new diagnostic tools and treatment strategies. To address these challenges, nanotechnology-based drug delivery systems-such as liposomes, micelles, dendrimers, and solid lipid nanoparticles (SLNs)-have emerged as promising solutions. This study provides a comprehensive review of recent advances in nanomedicine and nanotechnology-based platforms, alongside an exploration of ND mechanisms. The authors conducted a systematic literature search across relevant databases such as PubMed, Scopus, and Web of Science, focusing on peer-reviewed articles, reviews, and clinical studies published within the last 5 to 10 years. Additionally, this paper addresses the challenges faced by nanomedicines and delivery systems, offering insights into future directions in the field and the need for further research to establish their clinical viability as alternatives to current therapies.
Collapse
Affiliation(s)
| | - Amirreza Moshari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Tannaz Haghgouei
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Maryam Homaei
- Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Division of Pharmacology and Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
- Tabriz Neuroscience Research Center (NRSC), Neurology Department, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Tabriz, Iran
| | - Saeid Charsouei
- Tabriz Neuroscience Research Center (NRSC), Neurology Department, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Tabriz, Iran.
| |
Collapse
|
35
|
Antal BB, van Nieuwenhuizen H, Chesebro AG, Strey HH, Jones DT, Clarke K, Weistuch C, Ratai EM, Dill KA, Mujica-Parodi LR. Brain aging shows nonlinear transitions, suggesting a midlife "critical window" for metabolic intervention. Proc Natl Acad Sci U S A 2025; 122:e2416433122. [PMID: 40030017 PMCID: PMC11912423 DOI: 10.1073/pnas.2416433122] [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: 08/16/2024] [Accepted: 01/13/2025] [Indexed: 03/19/2025] Open
Abstract
Understanding the key drivers of brain aging is essential for effective prevention and treatment of neurodegenerative diseases. Here, we integrate human brain and physiological data to investigate underlying mechanisms. Functional MRI analyses across four large datasets (totaling 19,300 participants) show that brain networks not only destabilize throughout the lifetime but do so along a nonlinear trajectory, with consistent temporal "landmarks" of brain aging starting in midlife (40s). Comparison of metabolic, vascular, and inflammatory biomarkers implicate dysregulated glucose homeostasis as the driver mechanism for these transitions. Correlation between the brain's regionally heterogeneous patterns of aging and gene expression further supports these findings, selectively implicating GLUT4 (insulin-dependent glucose transporter) and APOE (lipid transport protein). Notably, MCT2 (a neuronal, but not glial, ketone transporter) emerges as a potential counteracting factor by facilitating neurons' energy uptake independently of insulin. Consistent with these results, an interventional study of 101 participants shows that ketones exhibit robust effects in restabilizing brain networks, maximized from ages 40 to 60, suggesting a midlife "critical window" for early metabolic intervention.
Collapse
Affiliation(s)
- Botond B. Antal
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Helena van Nieuwenhuizen
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Physics, State University of New York at Stony Brook, Stony Brook, NY
| | - Anthony G. Chesebro
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Helmut H. Strey
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Kieran Clarke
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eva-Maria Ratai
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ken A. Dill
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
| | - Lilianne R. Mujica-Parodi
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY
- Laufer Center for Physical and Quantitative Biology, State University of New York at Stony Brook, Stony Brook, NY
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Physics, State University of New York at Stony Brook, Stony Brook, NY
- Santa Fe Institute, Santa Fe, NM
| |
Collapse
|
36
|
Zhang L, Ge Q, Sun Z, Zhang R, Li X, Luo X, Tian R, Cao Y, Pu C, Li L, Wu D, Jiang P, Yu C, Nosarti C, Xiao C, Liu Z. Association and shared biological bases between birth weight and cortical structure. Transl Psychiatry 2025; 15:74. [PMID: 40044659 PMCID: PMC11882966 DOI: 10.1038/s41398-025-03294-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/14/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Associations between birth weight and cortical structural phenotypes have been detected; however, the understanding is incomprehensive, and the potential biological bases are not well defined. Leveraging data from genome-wide association studies, we investigated the associations and the shared transcriptomic, proteomic and cellular bases of birth weight and 13 cortical structural phenotypes. Mendelian randomization analyses were performed to examine associations between birth weight and cortical structure. Downstream transcriptome-wide association study (TWAS), proteome-wide association study (PWAS) and summary-based Mendelian randomization (SMR) analyses were utilized to identify the shared cis-regulated gene expressions and proteins. Finally, cell-type expression-specific integration for complex traits (CELLECT) analyses were conducted to explore the enriched cell types. The Mendelian randomization analyses found positive associations between birth weight and global cortical folding index, intrinsic curvature index, local gyrification index, surface area and volume. Downstream transcriptomic-level TWAS and SMR identified three gene expressions both linked to birth weight and at least one cortical structural phenotype (CNNM2, RABGAP1 and CENPW). Parallel PWAS and SMR analyses at the proteomic level identified four proteins linked to both phenotypes (CNNM2, RAB7L1, RAB5B and PPA2), of which CNNM2 was replicated. CELLECT analyses revealed brain cell types enriched in birth weight, including pericytes, inhibitory GABAergic neurons and cerebrovascular cells. These findings support the importance of early life growth to cortical structure, and suggest underlying transcriptomic, proteomic and cellular bases. These results provide intriguing targets for further research into the mechanisms of cortical development.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qiaoyue Ge
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zeyuan Sun
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Rui Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xinxi Li
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Luo
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Run Tian
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuheng Cao
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunyan Pu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Li
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Dongsheng Wu
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ping Jiang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- West China Medical Publishers, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Chuan Yu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chiara Nosarti
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Chenghan Xiao
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Zhenmi Liu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
37
|
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
|
38
|
Bateman GA, Bateman AR. Brain Ischemia in Alzheimer's Disease May Partly Counteract the Disruption of the Blood-Brain Barrier. Brain Sci 2025; 15:269. [PMID: 40149790 PMCID: PMC11940560 DOI: 10.3390/brainsci15030269] [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: 02/11/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND In normal pressure hydrocephalus (NPH) there is blood-brain barrier (BBB) disruption, which should increase the CSF formation rate (CSFfr) and, therefore, also increase the intracranial pressure (ICP). However, the ICP is normal in NPH. A lumped parameter study was performed to look at the interrelation between the ICP, cerebral blood flow (CBF), and the degree of BBB disruption in NPH. The model suggested that the CSFfr could be reduced in this condition if the BBB disruption was moderated by a reduction in the capillary transmural pressure (TMP) secondary to arteriolar constriction and a reduced CBF. In early Alzheimer's disease (AD), there is BBB disruption, reduced ICP, and global ischemia. This raises the possibility that the same physiology may occur in AD as occurs in NPH. METHODS A lumped parameter model previously used to describe the hydrodynamics of NPH was modified to investigate the effects of changes in CSF pressure and blood flow in patients with mild cognitive impairment (MCI) and AD. RESULTS The model indicates that the average capillary TMP is normal in MCI, but decreases as AD progresses. Removing CSF in AD patients during a tap test initially increases the capillary TMP. The brain in AD responds to a tap test by increasing its level of ischemia, and this reduces the capillary TMP. CONCLUSIONS A hypothesis is put forward that the BBB disruption in AD is partially mitigated by the brain making itself ischemic. Modelling gives support to this hypothesis. The model can suggest a cause for the development of ischemic neuronal loss and amyloid accumulation secondary to glymphatic flow disruption as AD progresses.
Collapse
Affiliation(s)
- Grant A. Bateman
- Department of Medical Imaging, John Hunter Hospital, Newcastle, NSW 2310, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, Newcastle University, Callaghan Campus, Newcastle, NSW 2308, Australia
| | - Alexander R. Bateman
- School of Engineering, College of Engineering, Science and Environment, Newcastle University, Callaghan Campus, Newcastle, NSW 2308, Australia;
| |
Collapse
|
39
|
Pavlou G, Spitz S, Pramotton FM, Tsai A, Li BM, Wang X, Barr OM, Ko EC, Zhang S, Ashley SJ, Maaser-Hecker A, Choi SH, Jorfi M, Tanzi RE, Kamm RD. Engineered 3D human neurovascular model of Alzheimer's disease to study vascular dysfunction. Biomaterials 2025; 314:122864. [PMID: 39357152 PMCID: PMC12012383 DOI: 10.1016/j.biomaterials.2024.122864] [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: 03/22/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
The blood-brain barrier (BBB) serves as a selective filter that prevents harmful substances from entering the healthy brain. Dysfunction of this barrier is implicated in several neurological diseases. In the context of Alzheimer's disease (AD), BBB breakdown plays a significant role in both the initiation and progression of the disease. This study introduces a three-dimensional (3D) self-assembled in vitro model of the human neurovascular unit to recapitulate some of the complex interactions between the BBB and AD pathologies. It incorporates primary human brain endothelial cells, pericytes and astrocytes, and stem cell-derived neurons and astrocytes harboring Familial AD (FAD) mutations. Over an extended co-culture period, the model demonstrates increased BBB permeability, dysregulation of key endothelial and pericyte markers, and morphological alterations mirroring AD pathologies. The model enables visualization of amyloid-beta (Aβ) accumulation in both neuronal and vascular compartments. This model may serve as a versatile tool for neuroscience research and drug development to provide insights into the dynamic relationship between vascular dysfunction and AD pathogenesis.
Collapse
Affiliation(s)
- Georgios Pavlou
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Alice Tsai
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brent M Li
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Xun Wang
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Olivia M Barr
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Eunkyung Clare Ko
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Shun Zhang
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Savannah J Ashley
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Anna Maaser-Hecker
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Roger D Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA.
| |
Collapse
|
40
|
Gutiérrez‐Jiménez E, Rasmussen PM, Mikkelsen IK, Kura S, Fruekilde SK, Hansen B, Bordoni L, Carlsen J, Palmfeldt J, Boas DA, Sakadžić S, Vinogradov S, Khatib ME, Ramos‐Cejudo J, Wied B, Leduc‐Galindo D, Canepa E, Mar AC, Gamallo‐Lana B, Fossati S, Østergaard L. Carbonic anhydrase inhibitors prevent presymptomatic capillary flow disturbances in a model of cerebral amyloidosis. Alzheimers Dement 2025; 21:e70023. [PMID: 40133235 PMCID: PMC11936728 DOI: 10.1002/alz.70023] [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: 08/28/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION Disturbances in microvascular flow dynamics are hypothesized to precede the symptomatic phase of Alzheimer's disease (AD). However, evidence in presymptomatic AD remains elusive, underscoring the need for therapies targeting these early vascular changes. METHODS We employed a multimodal approach, combining in vivo optical imaging, molecular techniques, and ex vivo magnetic resonance imaging, to investigate early capillary dysfunction in C57BL/6-Tg(Thy1-APPSwDutIowa)BWevn/Mmjax (Tg-SwDI) mice without memory impairment. We also assessed the efficacy of carbonic anhydrase inhibitors (CAIs) in preventing capillary flow disturbances. RESULTS Our study revealed capillary flow disturbances associated with alterations in capillary morphology, adhesion molecule expression, and amyloid beta (Aβ) load in 9- to 10-month-old Tg-SwDI mice without memory impairment. CAI treatment ameliorated these capillary flow disturbances, enhanced oxygen availability, and reduced Aβ load. DISCUSSION These findings underscore the importance of capillary flow disturbances as early biomarkers in presymptomatic AD and highlight the potential of CAIs for preserving vascular integrity in the early stages of AD. HIGHLIGHTS Uncovered early capillary dysfunction in a presymptomatic Alzheimer's disease (AD) mouse model. Evidence linking capillary stalls and capillary dysfunction with oxygen delivery issues in AD. Novel use of carbonic anhydrase inhibitors to prevent early capillary flow disturbances in AD.
Collapse
Affiliation(s)
- Eugenio Gutiérrez‐Jiménez
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Peter Mondrup Rasmussen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Irene Klærke Mikkelsen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Sreekanth Kura
- Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
| | - Signe K. Fruekilde
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Luca Bordoni
- GliaLab and Letten Centre, Division of AnatomyDepartment of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jasper Carlsen
- Research Unit for Molecular Medicine (MMF), Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine (MMF), Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - David A. Boas
- Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Sergei Vinogradov
- Department of Biochemistry and BiophysicsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of ChemistrySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Mirna El Khatib
- Department of Biochemistry and BiophysicsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of ChemistrySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jaime Ramos‐Cejudo
- Department of Psychiatry and NeurologyNew York University (NYU) Grossman School of MedicineNew York CityNew YorkUSA
| | - Boris Wied
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Desiree Leduc‐Galindo
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Elisa Canepa
- Alzheimer's Center at TempleDepartment of Neural SciencesLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Adam C. Mar
- Department of Neuroscience and PhysiologyNeuroscience InstituteNew York University (NYU) Grossman School of MedicineNew YorkNew YorkUSA
| | - Begona Gamallo‐Lana
- Department of Neuroscience and PhysiologyNeuroscience InstituteNew York University (NYU) Grossman School of MedicineNew YorkNew YorkUSA
| | - Silvia Fossati
- Alzheimer's Center at TempleDepartment of Neural SciencesLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Section of NeuroradiologyDepartment of RadiologyAarhus University HospitalAarhusDenmark
| |
Collapse
|
41
|
Chen G, Li H, Shao X, Wang DJJ, Hu W, Han Y, Yang Q. Decreased water exchange rate across the blood-brain barrier throughout the Alzheimer's disease continuum: Evidence from Chinese data. Alzheimers Dement 2025; 21:e70089. [PMID: 40145328 PMCID: PMC11947737 DOI: 10.1002/alz.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Water exchange rate (Kw) across the blood-brain barrier (BBB) is used in magnetic resonance imaging (MRI) techniques to evaluate BBB functionality. Variations in BBB Kw across the Alzheimer's disease (AD) continuum remain uncertain. METHODS The study encompassed 38 cognitively normal individuals without AD biomarkers (CN_A-), 30 cognitively normal (CN_A+), and 31 cognitively impaired individuals (CI_A+) with positive AD biomarkers. Participants underwent clinical assessments, MRI/positron emission tomography scans, and assays of plasma biomarkers. RESULTS Significantly lower Kw was observed in multiple brain regions throughout the AD continuum. This alteration in Kw correlated with plasma biomarkers and neuropsychological performance. Elevated levels of phosphorylated tau 217 intensified the inverse relationship between Kw and neuropsychological performance. The integration of Kw, brain volume, and plasma biomarkers demonstrated potential in distinguishing stages within the AD continuum. DISCUSSION Consistently lower Kw was evident across the AD continuum and may act as a diagnostic tool for early AD screening. HIGHLIGHTS Observations revealed a decline in water exchange rate (Kw) across multiple brain regions within the Alzheimer's disease (AD) continuum, notably in the hippocampus, parahippocampal gyrus, and deep brain nuclei during the preclinical stage of AD. Strong correlations were established between Kw levels in various brain regions and plasma biomarkers, as well as neuropsychological performance in the AD continuum. Interaction between plasma phosphorylated tau (p-tau)217 and Kw in the hippocampus was linked to executive function, indicating a combined detrimental impact on cognitive abilities stemming from both blood-brain barrier Kw and p-tau 217. The combined use of Kw, brain volume, and plasma biomarkers-neurofilament light chain and glial fibrillary acidic protein-demonstrated potential for distinguishing individuals within the AD continuum.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of NeurologyBeijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Hui Li
- Department of RadiologyBeijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT)USC Mark & Mary Stevens Neuroimaging and Informatics InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Danny J. J. Wang
- Laboratory of FMRI Technology (LOFT)USC Mark & Mary Stevens Neuroimaging and Informatics InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Wenli Hu
- Department of NeurologyBeijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Ying Han
- Department of NeurologyXuan Wu Hospital of Capital Medical UniversityBeijingChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryGaoke Innovation CenterShenzhenChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
- The Central Hospital of KaramayKaramayChina
| | - Qi Yang
- Department of RadiologyBeijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
42
|
Tan AYS, Martinez LCMG, Murray HC, Mehrabi NF, Tippett LJ, Turner CP, Curtis MA, Faull RLM, Dragunow M, Singh-Bains MK. Elucidating cortical neurovascular involvement in Huntington's disease using human brain tissue microarrays. Neurobiol Dis 2025; 206:106829. [PMID: 39909082 DOI: 10.1016/j.nbd.2025.106829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025] Open
Abstract
Although the genetic basis of Huntington's disease (HD) has been determined, the underlying pathophysiological mechanisms contributing to neurodegeneration remain largely unknown. In recent years, increasing evidence has posited vascular dysfunction as a significant early event in disease pathogenesis; however, these processes remain to be fully elucidated. High-content immunohistochemical screening studies were conducted on HD middle temporal gyrus (MTG) human brain tissue microarrays (TMAs) to investigate various components of the vascular system, including endothelial cells (UEA-1), pericytes (PDGFRβ), vascular smooth muscle cells (αSMA), extracellular matrix components (ECM; collagen IV and fibronectin), and leakage markers (haemoglobin and fibrinogen). Analyses of vascular markers revealed an increase in the number of vessels in the HD TMA cohort which was associated with advancing striatal pathology and earlier symptom onset. Furthermore, our findings highlight the preservation of pericytes, vascular smooth muscle cells, ECM components, and blood-brain barrier integrity in the HD MTG. Collectively, the TMA findings allude to mild vascular remodelling in the temporal cortex which is known to present with a lesser degree of neuronal degeneration in HD.
Collapse
Affiliation(s)
- Adelie Y S Tan
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Lance C M G Martinez
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Helen C Murray
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Nasim F Mehrabi
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland 1023, New Zealand
| | - Lynette J Tippett
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; School of Psychology, University of Auckland, Auckland 1023, New Zealand
| | - Clinton P Turner
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland 1023, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland 1023, New Zealand.
| | - Malvindar K Singh-Bains
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
43
|
Dutt S, Bachman SL, Dahl MJ, Li Y, Yew B, Jang JY, Ho JK, Nashiro K, Min J, Yoo HJ, Gaubert A, Nguyen A, Blanken AE, Sible IJ, Marshall AJ, Kapoor A, Alitin JPM, Hoang K, Rouanet J, Sordo L, Head E, Shao X, Wang DJJ, Mather M, Nation DA. Locus coeruleus MRI contrast, cerebral perfusion, and plasma Alzheimer's disease biomarkers in older adults. Neurobiol Aging 2025; 147:12-21. [PMID: 39637519 PMCID: PMC11781958 DOI: 10.1016/j.neurobiolaging.2024.11.008] [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: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
The locus coeruleus (LC) is among the first brain structures impacted by Alzheimer's disease (AD), and noradrenergic denervation may contribute to early neurovascular dysfunction in AD. Mechanistic links between the LC and cerebral perfusion have been demonstrated in rodents, but there have been no similar studies in aging humans. Community-dwelling older adults with no history of stroke or dementia (N=66) underwent structural (T1-MPRAGE; T1-FSE) and perfusion (resting pCASL) MRI. Plasma AD biomarkers levels were evaluated for Aβ42/40 ratio (n=56) and pTau181 (n=60). Higher rostral LC structural MRI contrast was associated with lower perfusion in entorhinal and limbic regions but higher perfusion in lateral and medial orbitofrontal cortices. Relationships between LC structure and regional cerebral perfusion were attenuated in older adults with higher plasma pTau levels and lower plasma Aβ42/40 ratios. Previously unstudied links between LC structure and cerebral perfusion are detectible in older adults using MRI and are attenuated in those showing greater AD pathophysiologic change, suggesting an uncoupling of LC-cerebral perfusion relationships in older adults with aggregating AD-related pathophysiology.
Collapse
Affiliation(s)
- Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Shelby L Bachman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Martin J Dahl
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Yanrong Li
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Belinda Yew
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jung Yun Jang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jean K Ho
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Kaoru Nashiro
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jungwon Min
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hyun Joo Yoo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Aimée Gaubert
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Nguyen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Anna E Blanken
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA; San Francisco Veterans Affairs Health Care System, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa J Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, CA, USA
| | - John Paul M Alitin
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kim Hoang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jeremy Rouanet
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Lorena Sordo
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Xingfeng Shao
- Laboratory of FMRI Technology, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA.
| |
Collapse
|
44
|
McDonough A, Weinstein JR. Glial 'omics in ischemia: Acute stroke and chronic cerebral small vessel disease. Glia 2025; 73:495-518. [PMID: 39463002 PMCID: PMC11785505 DOI: 10.1002/glia.24634] [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: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Vascular injury and pathologies underlie common diseases including ischemic stroke and cerebral small vessel disease (CSVD). Prior work has identified a key role for glial cells, including microglia, in the multifaceted and temporally evolving neuroimmune response to both stroke and CSVD. Transcriptional profiling has led to important advances including identification of distinct gene expression signatures in ischemia-exposed, flow cytometrically sorted microglia and more recently single cell RNA sequencing-identified microglial subpopulations or clusters. There is a reassuring degree of overlap in the results from these two distinct methodologies with both identifying a proliferative and a separate type I interferon responsive microglial element. Similar patterns were later seen using multimodal and spatial transcriptomal profiling in ischemia-exposed microglia and astrocytes. Methodological advances including enrichment of specific neuroanatomic/functional regions (such as the neurovascular unit) prior to single cell RNA sequencing has led to identification of novel cellular subtypes and generation of new credible hypotheses as to cellular function based on the enhanced cell sub-type specific gene expression patterns. A ribosomal tagging strategy focusing on the cellular translatome analyses carried out in the acute phases post stroke has revealed distinct inflammation-regulating roles for microglia and astrocytes in this setting. Early spatial transcriptomics experiments using cerebral ischemia models have identified regionally distinct microglial cell clusters in ischemic core versus penumbra. There is great potential for combination of these methods for multi-omics approaches to further elucidate glial responses in the context of both acute ischemic stroke and chronic CSVD.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington 98195-6465
| | - Jonathan R. Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington 98195-6465
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington 98195-6465
| |
Collapse
|
45
|
Liu H, Yu R, Zhang M, Zheng X, Zhong L, Yang W, Luo Y, Huang Z, Zheng J, Zhong H, Wei X, Zheng W, Yu Y, Wang Q. Fibrinogen degradation products exacerbate alpha-synuclein aggregation by inhibiting autophagy via downregulation of Beclin1 in multiple system atrophy. Neurotherapeutics 2025; 22:e00538. [PMID: 39904669 PMCID: PMC12014411 DOI: 10.1016/j.neurot.2025.e00538] [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: 04/19/2024] [Revised: 11/24/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disease arising from accumulation of the α-synuclein and aberrant protein clearance in oligodendrocytes. The mechanisms of autophagy involved in the progression of MSA remain poorly understood. It is reported that MSA patients have blood-brain barrier impairments, which may increase the entry of fibrinogen into the brain. However, the roles of fibrinogen and its degradation products (FDPs) on autophagy and α-synuclein accumulation in MSA remain unknown. Here, we established the MSA animal model by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) and 3-nitropropionic acid (3-NP), and cellular models by adding fibrillar α-syn into oligodendrocytes to investigate the mechanisms of FDPs on autophagy and accumulation of α-synuclein in oligodendrocytes. We found that FDPs inhibit the entry of α-synuclein into lysosomes for degradation, increasing aggregation of α-synuclein in oligodendrocytes (OLN-93). Our findings indicated that in OLN-93, FDPs inhibited the expressions of Beclin1 and Bif-1, which could promote the fusion of autophagosomes with lysosomes. Furthermore, the expression of α-synuclein was elevated in FDPs-injected mice, accompanied by an increase in the protein level of p62. We detected elevated expression of FDPs in the striatum of MSA mice. Finally, FDPs inhibited the expression of Beclin1 and Bif-1, which led to aberrant autophagic degradation and increased aggregation of α-synuclein and phospho-α-synuclein in MSA mice. Our study illustrates that FDPs can cause aggregation of α-synuclein in MSA by inhibiting Beclin1-mediated autophagy, which may exacerbate disease progression. These results provide a new therapeutic approach for MSA, that targets the inhibitory effect of FDPs on oligodendrocyte autophagy.
Collapse
Affiliation(s)
- Huanzhu Liu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Ruoyang Yu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Muwei Zhang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Lizi Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hui Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wenhua Zheng
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, 999078, Macao, China
| | - Yinghua Yu
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, Australia.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
46
|
Wen C, Gan JH, Liu S, Lu H, Wang LC, Wu H, Shi ZH, Ji Y. Enlarged perivascular spaces correlate with blood-brain barrier leakage and cognitive impairment in Alzheimer's disease. J Alzheimers Dis 2025; 104:382-392. [PMID: 39924914 DOI: 10.1177/13872877251317220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
BackgroundThe clinical significance of enlarged perivascular spaces (EPVS) in Alzheimer' s disease (AD) was ambiguous.ObjectiveTo investigate whether EPVS contribute to blood-brain barrier (BBB) leakage and cognition in AD.MethodsThe study included a total of 64 participants (26 healthy controls and 38 patients with AD). The evaluation of EPVS and BBB permeability was performed in specific anatomical locations: the centrum semiovale (CSO), basal ganglia, and hippocampus. The EPVS ratings were performed according to Potter's instructions. BBB permeability was evaluated using dynamic contrast-enhanced-MRI. The relationship between EPVS and global cognition (Mini-Mental State Examination and Montreal Cognitive Assessment), cognitive subdomains, and BBB permeability were examined in both groups. Finally, the relationship between CSO BBB permeability and cognition in AD patients was investigated.ResultsHigh-grade CSO EPVS was found associated with AD (OR: 3.40, 95% CI: 1.11-11.90, p = 0.04). In the AD group, a significant correlation was observed between high-grade CSO EPVS and lower MMSE score (r = -0.36, p = 0.03) and verbal fluency (r = -0.44, p = 0.01). High-grade CSO EPVS positively correlated with BBB leakage (r = 0.58, p < 0.001). The BBB permeability of CSO negatively correlated with verbal fluency (r = -0.52, p < 0.001) and attention (r = -0.40, p = 0.01).ConclusionsHigh-grade CSO EPVS is related to BBB leakage, which contributes to cognitive impairment in AD patients, especially verbal frequency. CSO EPVS can function as a convenient AD marker for intervention and therapy.
Collapse
Affiliation(s)
- Chen Wen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jing-Huan Gan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Hao Lu
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Li-Chen Wang
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Hao Wu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Zhi-Hong Shi
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| |
Collapse
|
47
|
Manfredi JN, Gupta SK, Vyavahare S, Deak F, Lu X, Buddha L, Wankhade U, Lohakare J, Isales C, Fulzele S. Gut microbiota dysbiosis in Alzheimer's disease (AD): Insights from human clinical studies and the mouse AD models. Physiol Behav 2025; 290:114778. [PMID: 39672482 DOI: 10.1016/j.physbeh.2024.114778] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Alzheimer's Disease (AD) is a debilitating neurocognitive disorder with an unclear underlying mechanism. Recent studies have implicated gut microbiota dysbiosis with the onset and progression of AD. The connection between gut microbiota and AD can significantly affect the prevention and treatment of AD patients. This systematic review summarizes primary outcomes of human and mouse AD models concerning gut microbiota alterations. A systematic literature search in February through March 2023 was conducted on PubMed, Embase, and Web of Science. We identified 711 as potential manuscripts of which 672 were excluded because of irrelevance to the identified search criteria. Primary outcomes include microbiota compositions of control and AD models in humans and mice. In total, 39 studies were included (19 mouse and 20 human studies), published between 2017 and 2023. We included studies involving well-established mice models of AD (5xFAD, 3xTg-AD, APP/PS1, Tg2576, and APPPS2) which harbor mutations and genes that drive the formation of Aß plaques. All human studies were included on those with AD or mild cognitive impairment. Among alterations in gut microbiota, most studies found a decreased abundance of the phyla Firmicutes and Bifidobacteria, a genus of the phylum Actinomycetota. An increased abundance of the phyla Bacteroidetes and Proteobacteria were identified in animal and human studies. Studies indicated that gut microbiota alter the pathogenesis of AD through its impact on neuroinflammation and permeability of the gastrointestinal tract. The ensuing increase in blood-brain barrier permeability may accelerate Aβ penetrance and formation of neuritic plaques that align with the amyloid hypothesis of AD pathogenesis. Further studies should assess the relationship between gut microbiota and AD progression and therapy preserving beneficial gut microbiota.
Collapse
Affiliation(s)
- John N Manfredi
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ferenc Deak
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Xinyun Lu
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Lasya Buddha
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Umesh Wankhade
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jayant Lohakare
- College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
48
|
Padrela BE, Slivka M, Sneve MH, Garrido PF, Dijsselhof MBJ, Hageman T, Geier O, Grydeland H, Mahroo A, Kuijer JPA, Konstandin S, Eickel K, Barkhof F, Günther M, Walhovd KB, Fjell AM, Mutsaerts HJMM, Petr J. Blood-brain barrier water permeability across the adult lifespan: A multi-echo ASL study. Neurobiol Aging 2025; 147:176-186. [PMID: 39798256 DOI: 10.1016/j.neurobiolaging.2024.12.012] [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: 08/13/2024] [Revised: 11/28/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
An emerging biomarker of blood-brain barrier (BBB) permeability is the time of exchange (Tex) of water from the blood to tissue, as measured by multi-echo arterial spin labeling (ASL) MRI. This new non-invasive sequence, already tested in mice, has recently been adapted to humans and optimized for clinical scanning time. In this study, we studied the normal variability of Tex over age and sex, which needs to be established as a reference for studying changes in neurological disease. We evaluated Tex, cerebral blood flow (CBF) and arterial transit time (ATT) in 209 healthy adults between 26 and 87 years, over age and sex, using general linear models in gray matter, white matter, and regionally in cerebral lobes. After QC, 194 participants were included in the main analysis, and the results demonstrated that both gray matter (GM) and white matter (WM) BBB permeability was higher with higher age (Tex lower by 0.47 ms per year in GM [p < 0.05], and by 0.49 ms in WM, for females; no significant for males), with the largest Tex difference in the frontal lobes (0.64 ms decrease per year, p = 0.011, population average). CBF was lower with higher age in the GM (-0.71 mL/min/100g per year, p < 0.001, for females; -0.31 mL/min/100g per year, p < 0.05, for males). When correcting Tex models for CBF and ATT, effect of age on Tex disappears in the GM, but not in the WM (β=-0.28, p = 0.08). The CBF findings of this study are in line with previous studies, demonstrating the validity of the new sequence. The BBB water permeability variation over age and sex described in this study provides a reference for future BBB research.
Collapse
Affiliation(s)
- Beatriz E Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands.
| | - Maksim Slivka
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Markus H Sneve
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Pablo F Garrido
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mathijs B J Dijsselhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Tamara Hageman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
| | - Oliver Geier
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Håkon Grydeland
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Amnah Mahroo
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Joost P A Kuijer
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Simon Konstandin
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Klaus Eickel
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; mediri GmbH, Heidelberg, Germany; Faculty 1 - Physics / Electrical Engineering, University Bremen, Bremen, Germany
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Henk J M M Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| |
Collapse
|
49
|
Bohannon DG, Long D, Okhravi HR, Lee SC, De Jesus CL, Neubert TA, Rostagno AA, Ghiso JA, Kim W. Functionally distinct pericyte subsets differently regulate amyloid-β deposition in patients with Alzheimer's disease. Brain Pathol 2025; 35:e13282. [PMID: 38932696 PMCID: PMC11835444 DOI: 10.1111/bpa.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Although the concept that the blood-brain barrier (BBB) plays an important role in the etiology and pathogenesis of Alzheimer's disease (AD) has become increasingly accepted, little is known yet about how it actually contributes. We and others have recently identified a novel functionally distinct subset of BBB pericytes (PCs). In the present study, we sought to determine whether these PC subsets differentially contribute to AD-associated pathologies by immunohistochemistry and amyloid beta (Aβ) peptidomics. We demonstrated that a disease-associated PC subset (PC2) expanded in AD patients compared to age-matched, cognitively unimpaired controls. Surprisingly, we found that this increase in the percentage of PC2 (%PC2) was correlated negatively with BBB breakdown in AD patients, unlike in natural aging or other reported disease conditions. The higher %PC2 in AD patients was also correlated with a lower Aβ42 plaque load and a lower Aβ42:Aβ40 ratio in the brain as determined by immunohistochemistry. Colocalization analysis of multicolor confocal immunofluorescence microscopy images suggests that AD patient with low %PC2 have higher BBB breakdown due to internalization of Aβ42 by the physiologically normal PC subset (PC1) and their concomitant cell death leading to more vessels without PCs and increased plaque load. On the contrary, it appears that PC2 can secrete cathepsin D to cleave and degrade Aβ built up outside of PC2 into more soluble forms, ultimately contributing to less BBB breakdown and reducing Aβ plaque load. Collectively our data shows functionally distinct mechanisms for PC1 and PC2 in high Aβ conditions, demonstrating the importance of correctly identifying these populations when investigating the contribution of neurovascular dysfunction to AD pathogenesis.
Collapse
Affiliation(s)
- Diana G. Bohannon
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Danielle Long
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Hamid R. Okhravi
- Glennan Center for Geriatrics and GerontologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Integrated Neurodegenerative Disorders CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | | | | | - Thomas A. Neubert
- Department of Neuroscience and PhysiologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Agueda A. Rostagno
- Department of PathologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Jorge A. Ghiso
- Department of PathologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
- Department of PsychiatryNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Woong‐Ki Kim
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Integrated Neurodegenerative Disorders CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Division of MicrobiologyTulane National Primate Research CenterCovingtonLouisianaUSA
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
50
|
Xu M, Chen Y, Lin Y, Wang D, Zheng X. Serum-derived exosomal microRNAs as biomarkers for postoperative delirium. Front Neurosci 2025; 19:1525230. [PMID: 40092071 PMCID: PMC11906430 DOI: 10.3389/fnins.2025.1525230] [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: 11/09/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Postoperative delirium (POD) is a frequent and challenging complication in elderly surgical patients, marked by abrupt cognitive and attentional disturbances. Current POD diagnosis depends on clinical assessments that are time-intensive and lack predictive accuracy before surgery. Although previous research has explored biomarkers such as neuroinflammatory factors and Alzheimer's-related proteins to enhance POD prediction, single molecular markers have proven insufficient for reliable prognosis. Methods This study investigated serum exosomal miRNA expression profiles in postoperative patients to assess their association with POD. We compared miRNA expression between POD and non-POD groups through cognitive assessments and serum analyses. Additionally, enrichment analysis was conducted to determine the biological pathways regulated by differentially expressed miRNAs. Results Our analysis identified 57 miRNAs with significantly altered expression between POD and non-POD patients, including 16 upregulated and 41 downregulated miRNAs in the POD group. Enrichment analysis revealed that these miRNAs are involved in genes regulating neurotrophin signaling, neuroactive ligand-receptor interactions, and pathways that influence neuronal plasticity and cell viability. Discussion This study highlights specific miRNAs as potential biomarkers for POD and suggests their involvement in the underlying mechanisms of cognitive decline following surgery. By enhancing diagnostic capabilities and identifying potential therapeutic targets, our findings could lead to more effective POD management strategies for elderly patients. Further research is recommended to validate these miRNAs and evaluate their clinical utility for predictive screening and therapeutic interventions.
Collapse
Affiliation(s)
- Maokai Xu
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yingjie Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yujun Lin
- Department of Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Danfeng Wang
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| |
Collapse
|