1
|
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
|
2
|
Lim C, Lee H, Moon Y, Han S, Kim HJ, Chung HW, Moon W. Volume and Permeability of White Matter Hyperintensity on Cognition: A DCE Imaging Study of an Older Cohort With and Without Cognitive Impairment. J Magn Reson Imaging 2025; 61:2260-2270. [PMID: 39425583 PMCID: PMC11987793 DOI: 10.1002/jmri.29631] [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: 10/21/2023] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND The impact of blood-brain barrier (BBB) leakage on white matter hyperintensity (WMH) subtypes (location) and its association with clinical factors and cognition remains unclear. PURPOSE To investigate the relationship between WMH volume, permeability, clinical factors, and cognition in older individuals across the cognitive spectrum. STUDY TYPE Prospective, cross-sectional. SUBJECTS A total of 193 older adults with/without cognitive impairment; 128 females; mean age 70.1 years (standard deviation 6.8). FIELD STRENGTH/SEQUENCE 3 T, GE Dynamic contrast-enhanced, three-dimensional (3D) Magnetization-prepared rapid gradient-echo (MPRAGE T1WI), 3D fluid-attenuated inversion recovery (FLAIR). ASSESSMENT Periventricular WMH (PWMH), deep WMH (DWMH), and normal-appearing white matter (NAWM) were segmented using FMRIB automatic segmentation tool algorithms on 3D FLAIR. Hippocampal volume and cortex volume were segmented on 3D T1WI. BBB permeability (Ktrans) and blood plasma volume (Vp) were determined using the Patlak model. Vascular risk factors and cognition were assessed. STATISTICAL TESTS Univariate and multivariate analyses were performed to identify factors associated with WMH permeability. Logistic regression analysis assessed the association between WMH imaging features and cognition, adjusting for age, sex, apolipoprotein E4 status, education, and brain volumes. A P-value <0.05 was considered significant. RESULTS PWMH exhibited higher Ktrans (0.598 ± 0.509 × 10-3 minute-1) compared to DWMH (0.496 ± 0.478 × 10-3 minute-1) and NAWM (0.476 ± 0.398 × 10-3 minute-1). Smaller PWMH volume and cardiovascular disease (CVD) history were significantly associated with higher Ktrans in PWMH. In DWMH, higher Ktrans were associated with CVD history and cortical volume. In NAWM, it was linked to CVD history and dyslipidemia. Larger PWMH volume (odds ratio [OR] 1.106, confidence interval [CI]: 1.021-1.197) and smaller hippocampal volume (OR 0.069; CI: 0.019-0.253) were independently linked to worse global cognition after covariate adjustment. DATA CONCLUSION Elevated BBB leakage in PWMH was associated with lower PWMH volume and prior CVD history. Notably, PWMH volume, rather than permeability, was correlated with cognitive decline, suggesting that BBB leakage in WMH may be a consequence of CVD rather than indicate disease progression. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Changmok Lim
- Department of Radiology, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
| | - Hunwoo Lee
- Department of Radiology, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
| | - Yeonsil Moon
- Department of Neurology, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
- Research Institute of Medical ScienceKonkuk University of MedicineSeoulRepublic of Korea
| | - Seol‐Heui Han
- Department of Neurology, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
- Research Institute of Medical ScienceKonkuk University of MedicineSeoulRepublic of Korea
| | - Hee Jin Kim
- Department of Neurology, Hanyang University Medical CenterHanyang University School of MedicineSeoulRepublic of Korea
| | - Hyun Woo Chung
- Department of Nuclear Medicine, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
| | - Won‐Jin Moon
- Department of Radiology, Konkuk University Medical CenterKonkuk University School of MedicineSeoulRepublic of Korea
- Research Institute of Medical ScienceKonkuk University of MedicineSeoulRepublic of Korea
| |
Collapse
|
3
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [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: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|
4
|
Ringstad G, Eide PK, Naganawa S, Agarwal N. Gadolinium-Based Imaging and the Study of Neurofluid Dynamics. Neuroimaging Clin N Am 2025; 35:191-209. [PMID: 40210377 DOI: 10.1016/j.nic.2024.12.008] [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
Gadolinium-based MR imaging has significantly advanced our understanding of neurofluid dynamics within the brain and spinal cord. Both intrathecal and intravenous MR imaging techniques hold promise for identifying surrogate markers of altered neurofluid dynamics in the brain. These insights can shed light on the pathophysiology of various neurologic disorders and aid in developing improved treatment strategies.
Collapse
Affiliation(s)
- Geir Ringstad
- Department of Radiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Geriatric Medicine, Sørlandet Hospital Trust, Arendal, Norway; KG Jebsen Centre for Brain Fluid Research, University of Oslo, Oslo, Norway.
| | - Per Kristian Eide
- KG Jebsen Centre for Brain Fluid Research, University of Oslo, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Sognsvannsveien 20, 0372 Oslo, Norway; Department of Neurosurgery, University of Oslo, Oslo, Norway
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Shouwa-ku, Nagoya, 466-8550 Japan
| | - Nivedita Agarwal
- Diagnostic Imaging and Neuroradiology Unit, Scientific Institute, IRCCS "Eugenio Medea" Via Don Luigi Monza 20, 23842 Bosisio Parini (LC), Italy
| |
Collapse
|
5
|
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
|
6
|
Carnevale L, Lembo G. Imaging the cerebral vasculature at different scales: translational tools to investigate the neurovascular interfaces. Cardiovasc Res 2025; 120:2373-2384. [PMID: 39082279 DOI: 10.1093/cvr/cvae165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/26/2024] [Accepted: 05/23/2024] [Indexed: 04/09/2025] Open
Abstract
The improvements in imaging technology opened up the possibility to investigate the structure and function of cerebral vasculature and the neurovascular unit with unprecedented precision and gaining deep insights not only on the morphology of the vessels but also regarding their function and regulation related to the cerebral activity. In this review, we will dissect the different imaging capabilities regarding the cerebrovascular tree, the neurovascular unit, the haemodynamic response function, and thus, the vascular-neuronal coupling. We will discuss both clinical and preclinical setting, with a final discussion on the current scenery in cerebrovascular imaging where magnetic resonance imaging and multimodal microscopy emerge as the most potent and versatile tools, respectively, in the clinical and preclinical context.
Collapse
Affiliation(s)
- Lorenzo Carnevale
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Giuseppe Lembo
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, 'Sapienza' University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Jahng GH, Lee MB, Kwon OI. Gadolinium based contrast agent induced electrical conductivity heterogeneity analysis in the brain of Alzheimer's disease. Sci Rep 2025; 15:10832. [PMID: 40155644 PMCID: PMC11953297 DOI: 10.1038/s41598-025-92966-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] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Magnetic resonance imaging (MRI) often uses gadolinium-based contrast agents (GBCAs) to improve the characterization of imaging contrast, owing to their strong paramagnetic properties. Magnetic resonance electrical properties tomography (MREPT) visualizes the conductivity distribution of biological tissues at the Larmor frequency using the [Formula: see text] field phase signal. In this paper, we investigate the effect of GBCA on brain conductivity. To compare the differences of reconstructed noisy conductivity maps before and after the GBCA injection, we propose a method to remove the background low-frequency noise artifact based on an elliptic partial differential equation. By analyzing the relationship between electrical conductivity and magnetic permeability, the objective of this study is to develop a cost-effective and accessible initial screening imaging tool for diagnosing and monitoring the treatment of Alzheimer's disease (AD) pathophysiology. To investigate vascular damage in AD, we define a conductivity heterogeneity volume fraction (CHVF) caused by GBCA leakage. Using CHVF, we develop three indices to characterize mild cognitive impairment (MCI) and AD. To verify the proposed method, we studied a total of 42 participants, including 14 individuals diagnosed with AD, 18 participants with MCI, and 10 cognitively normal (CN) participants. Finally, we designed a radar chart informed by the CHVF analysis, to exhibit the pertinent parameters for MCI and AD patients, facilitating the evaluation and ongoing monitoring of each patient's diagnosis and treatment regimen.
Collapse
Affiliation(s)
- Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, 05278, Korea
| | - Mun Bae Lee
- Department of Mathematics, College of Basic Science, Konkuk University, Seoul, 05029, Korea
| | - Oh-In Kwon
- Department of Mathematics, College of Basic Science, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Stringer MS, Blair GW, Kopczak A, Kerkhofs D, Thrippleton MJ, Chappell FM, Maniega SM, Brown R, Shuler K, Hamilton I, Garcia DJ, Doubal FN, Clancy U, Sakka E, Poliakova T, Janssen E, Duering M, Ingrisch M, Staals J, Backes WH, van Oostenbrugge R, Biessels GJ, Dichgans M, Wardlaw JM. Cerebrovascular Function in Sporadic and Genetic Cerebral Small Vessel Disease. Ann Neurol 2025; 97:483-498. [PMID: 39552538 PMCID: PMC11831873 DOI: 10.1002/ana.27136] [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: 06/08/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVE Cerebral small vessel diseases (SVDs) are associated with cerebrovascular dysfunction, such as increased blood-brain barrier leakage (permeability surface area product), vascular pulsatility, and decreased cerebrovascular reactivity (CVR). No studies assessed all 3 functions concurrently. We assessed 3 key vascular functions in sporadic and genetic SVD to determine associations with SVD severity, subtype, and interrelations. METHODS In this prospective, cross-sectional, multicenter INVESTIGATE-SVDs study, we acquired brain magnetic resonance imaging in patients with sporadic SVD/cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), including structural, quantitative microstructural, permeability surface area product, blood plasma volume fraction, vascular pulsatility, and CVR (in response to CO2) scans. We determined vascular function and white matter hyperintensity (WMH) associations, using covariate-adjusted linear regression; normal-appearing white matter and WMH differences, interrelationships between vascular functions, using linear mixed models; and major sources of variance using principal component analyses. RESULTS We recruited 77 patients (45 sporadic/32 CADASIL) at 3 sites. In adjusted analyses, patients with worse WMH had lower CVR (B = -1.78, 95% CI -3.30, -0.27) and blood plasma volume fraction (B = -0.594, 95% CI -0.987, -0.202). CVR was worse in WMH than normal-appearing white matter (eg, CVR: B = -0.048, 95% CI -0.079, -0.017). Adjusting for WMH severity, SVD subtype had minimal influence on vascular function (eg, CVR in CADASIL vs sporadic: B = 0.0169, 95% CI -0.0247, 0.0584). Different vascular function mechanisms were not generally interrelated (eg, permeability surface area product~CVR: B = -0.85, 95% CI -4.72, 3.02). Principal component analyses identified WMH volume/quantitative microstructural metrics explained most variance in CADASIL and arterial pulsatility in sporadic SVD, but similar main variance sources. INTERPRETATION Vascular function was worse with higher WMH, and in WMH than normal-appearing white matter. Sporadic SVD-CADASIL differences largely reflect disease severity. Limited vascular function interrelations may suggest disease stage-specific differences. ANN NEUROL 2025;97:483-498.
Collapse
Affiliation(s)
- Michael S. Stringer
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Gordon W. Blair
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Anna Kopczak
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
| | - Danielle Kerkhofs
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Michael J. Thrippleton
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Francesca M. Chappell
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Susana Muñoz Maniega
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Rosalind Brown
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Kirsten Shuler
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Iona Hamilton
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Daniela Jaime Garcia
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Fergus N. Doubal
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Una Clancy
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Eleni Sakka
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Tetiana Poliakova
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Esther Janssen
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
- Medical Image Analysis Center (MIAC AG) and Department of Biomedical EngineeringUniversity of BaselBaselSwitzerland
| | | | - Julie Staals
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Walter H. Backes
- Department of Radiology & Nuclear MedicineMaastricht University Medical Center, Schools for Mental Health & Neuroscience and Cardiovascular DiseaseMaastrichtthe Netherlands
| | - Robert van Oostenbrugge
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtNetherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Joanna M. Wardlaw
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | | |
Collapse
|
14
|
Fuller OK, McLennan ED, Egan CL, Perera N, Terry LV, Pyun J, de Mendonca M, Telles GD, Smeuninx B, Burrows EL, Siddiqui G, Creek DJ, Scott JW, Pearen MA, Fonseka P, Nicolazzo JA, Mathivanan S, Hannan AJ, Ramm GA, Whitham M, Febbraio MA. Extracellular vesicles contribute to the beneficial effects of exercise training in APP/PS1 mice. iScience 2025; 28:111752. [PMID: 39898049 PMCID: PMC11787611 DOI: 10.1016/j.isci.2025.111752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Exercise improves cognitive function in Alzheimer's disease (AD) via mechanism that are not fully clear. Here, we first examined the effect of voluntary exercise training (VET) on energy metabolism and cognitive function in the APP/PS1 transgenic mouse (Tg) model of familial AD. Next, we profiled extracellular vesicles (EVs) and examined whether they may play a role in the protective effects of VET via intranasal administration of EVs, purified from the blood of sedentary (sEV) and/or acutely exercised (eEV) donor wild-type mice into APP/PS1Tg mice. We show that VET reduced resting energy expenditure (REE) and improved cognition in APP/PS1 Tg mice. Administration of eEV, but not sEV, also reduced REE, but had no effect on cognition. Taken together, these data show that exercise is effective intervention to improve symptoms of AD in APP/PS1Tg mice. In addition, eEVs mediate some of these effects, implicating EVs in the treatment of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver K. Fuller
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma D. McLennan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Casey L. Egan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren V. Terry
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jae Pyun
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Mariana de Mendonca
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Benoit Smeuninx
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma L. Burrows
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Ghizal Siddiqui
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Darren J. Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - John W. Scott
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Pamali Fonseka
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joseph A. Nicolazzo
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Suresh Mathivanan
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Grant A. Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Martin Whitham
- School of Sport, Exercise & Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Taoka T, Iwamoto K, Miyata S, Ito R, Nakamichi R, Nakane T, Okada I, Ichikawa K, Kan H, Kamagata K, Kikuta J, Aoki S, Fujimoto A, Kogo Y, Ichinose N, Naganawa S, Ozaki N. MR Imaging Indices for Brain Interstitial Fluid Dynamics and the Effects of Orexin Antagonists on Sleep. Magn Reson Med Sci 2025:mp.2024-0176. [PMID: 39924214 DOI: 10.2463/mrms.mp.2024-0176] [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: 02/11/2025] Open
Abstract
PURPOSE The purpose of this study was to assess the extent to which improvement in sleep with lemborexant contributed to changes in interstitial fluid dynamics. METHODS The 3 methods including diffusion tensor image analysis along the perivascular space (DTI-ALPS), dynamic contrast-enhanced method to assess tissue vascular permeability (Ktrans), and choroid plexus volume (CPV) were used. Correlations between these imaging indices and sleep parameters (latency to persistent sleep [LPS], wake after sleep onset [WASO], total sleep time [TST], and sleep efficiency [SE]) were evaluated using Pearson correlation analysis. Additionally, multiple regression analysis and linear mixed model analysis were employed to assess the relationship between baseline sleep status and imaging parameter changes. MRI and sleep assessments were performed before treatment initiation (week 0, w0) and at 12 weeks after lemborexant administration (week 12, w12). RESULTS The ALPS-index was inversely correlated with LPS and positively correlated with TST and SE at w0. In multiple regression analysis, ALPS-index was lower when sleep parameters other than LPS were poor at w0. A linear mixed model analysis suggested that poor sleep status in LPS and SE at w0 may have an effect on greater ALPS-index. In the evaluation of Ktrans measurement, the single regression analysis showed a statistically significant correlation between the reduction in Ktrans and the shortening in LPS. Examination of CPV and sleep parameters showed a significant negative correlation between TST and CPV at w0 and w12. Multiple regression analysis also showed that TST of w12 had a significant effect on CPV at w12. CONCLUSION Our results suggested that poor sleep status is related to the greater change of ALPS-index and CPV improvement after lemborexant administration may be related to in part to sleep parameter improvement.
Collapse
Affiliation(s)
- Toshiaki Taoka
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kunihiro Iwamoto
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Seiko Miyata
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Rintaro Ito
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Rei Nakamichi
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshiki Nakane
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Ippei Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazushige Ichikawa
- Department of Radiological Technology, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University of Medicine, Tokyo, Japan
| | - Junko Kikuta
- Department of Radiology, Juntendo University of Medicine, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University of Medicine, Tokyo, Japan
| | | | - Yuki Kogo
- Medical Headquarters, Eisai Co., Ltd., Tokyo, Japan
| | - Nobuyasu Ichinose
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of CT-MR Solution Planning, Canon Medical Systems Corporation, Otawara, Tochigi, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Norio Ozaki
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
16
|
Denkinger M, Baker S, Harrison TM, Chadwick T, Jagust WJ. Cross-sectional and longitudinal relationships among blood-brain barrier disruption, Alzheimer's disease biomarkers, and cognition in cognitively normal older adults. Neurobiol Aging 2025; 146:15-23. [PMID: 39571410 DOI: 10.1016/j.neurobiolaging.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Blood-brain barrier disruption (BBBd) occurs in aging, particularly in regions vulnerable to Alzheimer's disease (AD) pathology. However, its relationship to pathological protein accumulation, neurodegeneration, and cognitive impairment in normal aging is unclear. We used dynamic contrast-enhanced MRI (DCE-MRI) and positron emission tomography (PET) imaging in cognitively normal older adults to explore how BBBd correlates with brain atrophy and cognitive function, and whether these relationships are influenced by Aβ or tau. We found that greater BBBd in the hippocampus (HC) and an averaged BBBd-susceptible ROI were linked to worse episodic memory, with interactions between BBBd and atrophy influencing this relationship, independent of Aβ and tau. However, there were no significant relationships between BBBd and non-memory cognitive performance. In participants with longitudinal AD biomarker and cognitive data acquired prior to DCE-MRI, faster longitudinal entorhinal cortex (EC) tau accumulation and episodic memory decline were associated with greater HC BBBd, independent of global Aβ changes and regional atrophy. These findings enhance our understanding of the complex relationships between AD biomarkers, cognitive decline, and BBBd.
Collapse
Affiliation(s)
- Marisa Denkinger
- Department of Neuroscience, University of California, Berkeley, CA, United States.
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Theresa M Harrison
- Department of Neuroscience, University of California, Berkeley, CA, United States
| | - Trevor Chadwick
- Department of Neuroscience, University of California, Berkeley, CA, United States
| | - William J Jagust
- Department of Neuroscience, University of California, Berkeley, CA, United States; Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
17
|
Ishizaka H, Sekine A, Naka M, Nakano S, Nagase H, Tsushima Y. Hyperintensity of the left piriform cortex and amygdala on T2-weighted FLAIR images in patients with probable Alzheimer's disease correlates with cerebral cortical atrophy. Acta Radiol Open 2025; 14:20584601251317629. [PMID: 39916994 PMCID: PMC11795602 DOI: 10.1177/20584601251317629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/27/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
Background The left piriform cortex and amygdala (PC&A) tend to be slightly hyperintense relative to the right PC&A on T2-weighted fluid-attenuated inversion recovery (T2W-FLAIR) images in patients with probable Alzheimer's disease (pAD). This likely represents the antecedent and thus advanced degeneration of the left PC&A. Purpose To investigate the relationship between left PC&A hyperintensities and cerebral cortical atrophy on magnetic resonance (MR) voxel-based morphometry in patients with pAD and discuss how this finding could relate to AD progression. Material and Methods Patients with pAD (n = 47; age range = 68-93 years, mean = 80.8 ± 6.7 years; 14 men and 33 women) who underwent T2W-FLAIR imaging and MR morphometric study using a voxel-based specific regional analysis system for AD (VSRAD) were retrospectively examined. To measure signal intensity ratios of the left to right PC&A (L-PC&A/R-PC&A), regions of interest (ROIs) were set on the transaxial images in which both PC&As were most broadly depicted; the ROIs were defined as large as possible. Correlations between the L-PC&A/R-PC&A and medial temporal lobe cortical atrophy (MTLCA) as well as whole cerebral cortical atrophy (WCCA) on VSRAD were determined. Correlation between the L-PC&A/R-PC&A and age was also determined. Results The L-PC&A/R-PC&A correlated with both MTLCA (r = 0.375, p = .010, 95% confidence interval [CI] = 0.095-0.600) and WCCA (r = 0.576, p < .001, 95% CI = 0.343-0.742). The L-PC&A/R-PC&A did not correlate with age (r = 0.013, p = .932, 95% CI = -0.282-0.305). Conclusion Left-sided dominance of PC&A degeneration appeared to accelerate with the progression of AD stages.
Collapse
Affiliation(s)
- Hiroshi Ishizaka
- Department of Radiology, Maebashi Red Cross Hospital, Gunma, Japan
| | - Akiko Sekine
- Department of Neurology, Maebashi Red Cross Hospital, Gunma, Japan
| | - Minoru Naka
- Department of Radiology, Maebashi Red Cross Hospital, Gunma, Japan
| | - Saeki Nakano
- Department of Radiology, Maebashi Red Cross Hospital, Gunma, Japan
| | - Hiroyuki Nagase
- Department of Radiology, Maebashi Red Cross Hospital, Gunma, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Graduate School of Medicine, Gunma University, Gunma, Japan
| |
Collapse
|
18
|
van Setten A, Uleman JF, Melis RJF, Lawlor B, Riksen NP, Claassen JAHR, de Heus RAA. No association between markers of systemic inflammation and endothelial dysfunction with Alzheimer's disease progression: a longitudinal study. GeroScience 2025; 47:1093-1104. [PMID: 39085534 PMCID: PMC11872860 DOI: 10.1007/s11357-024-01294-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] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
INTRODUCTION Systemic inflammation and endothelial dysfunction are potentially modifiable factors implicated in Alzheimer's disease (AD), which offer potential therapeutic targets to slow disease progression. METHODS We investigated the relationship between baseline circulating levels of inflammatory (TNF-α, IL-1ß) and endothelial cell markers (VCAM-1, ICAM-1, E-selectin) and 18-month cognitive decline (ADAS-cog12) in 266 mild-to-moderate AD patients from the NILVAD study. We employed individual growth models to examine associations, potential mediation, and interaction effects while adjusting for confounders. RESULTS The average increase in ADAS-cog12 scores over all patients was 8.1 points in 18 months. No significant association was found between the markers and the rate of cognitive decline. Mediation analysis revealed no mediating role for endothelial cell markers, and interaction effects were not observed. DISCUSSION Our results do not support the role of systemic inflammation or endothelial dysfunction in progression in persons with AD.
Collapse
Affiliation(s)
- Arne van Setten
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen F Uleman
- Copenhagen Health Complexity Center, Department of Public Health, University of Copenhagen, Oster Farimagsgade 5, 1353, Copenhagen K, Denmark.
| | - René J F Melis
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College, Dublin, Ireland
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Rianne A A de Heus
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Primary and Community Care, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Janus A, Dumas D, Le Douce J, Marie S, Pasculli G, Bambury P, Lemarchant S, Kremer P, Godfrin Y. Safety, Tolerability and Pharmacokinetic-Pharmacodynamic Relationship of NX210c Peptide in Healthy Elderly Volunteers: Randomized, Placebo-Controlled, Double-Blind, Multiple Ascending Dose Study. Neurol Ther 2025; 14:357-377. [PMID: 39708220 PMCID: PMC11762061 DOI: 10.1007/s40120-024-00691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Blood-brain barrier (BBB) integrity is fundamental to brain homeostasis, enabling control of substance exchange and safeguarding neurons against harmful toxins, pathogens, and immune cells that lead to dysregulation and inflammation involved in ageing and neurodegenerative diseases (NDD). The cyclized peptide NX210c is a thrombospondin type 1 repeat analogue derived from subcommissural organ-spondin. It exerts beneficial effects in animal models of NDD owing to its effects on neurons and endothelial cells. NX210c demonstrated a good safety profile in a single ascending dose phase 1a clinical study. The present multiple ascending dose phase 1b study was performed to evaluate the tolerability and pharmacological effects of repeated doses of NX210c in healthy elderly (age: > 55 years) volunteers. METHODS This was a randomized, placebo-controlled, double-blind study (EudraCT No. 2022-002868-76), investigating safety/tolerability, pharmacokinetics, and pharmacodynamics (including blood and cerebrospinal fluid biomarkers). Participants received 5 or 10 mg/kg NX210c or placebo (10-min infusion) thrice weekly for 4 weeks in an ascending dose fashion. Follow-up was conducted 2 weeks after last dosing. RESULTS The investigation included 29 participants. No serious adverse events were recorded and all adverse events were mild. Dedicated central nervous system testing did not reveal neurotoxicity. Biomarker evaluation showed a statistically significant reduction in blood claudin-5 and a trend toward reduction of blood homocysteine. In silico data modelling revealed salient pharmacokinetic-pharmacodynamic relationships, including reduction of claudin-5, neurofilament light chain, and SPARC-like protein 1 release, and degradation of homocysteine. CONCLUSION Multiple doses of NX210c exhibited a good safety profile, showed non-cumulative pharmacokinetics, and exerted pharmacodynamic effects on biomarkers linked to BBB integrity. The effects of NX210c on claudin-5 and biomarkers influencing BBB integrity-and the overarching brain protection it offers-provide a novel therapeutic strategy targeting an underlying driver of neurodegenerative conditions for which disease-modifying treatments are limited or not available.
Collapse
Affiliation(s)
- Annette Janus
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France.
| | - Daniël Dumas
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Juliette Le Douce
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Sébastien Marie
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | | | | | - Sighild Lemarchant
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Philip Kremer
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Yann Godfrin
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
- Godfrin Life-Sciences, Caluire-Et-Cuire, France
| |
Collapse
|
20
|
Chaudhuri S, Cho M, Stumpff JC, Bice PJ, İş Ö, Ertekin-Taner N, Saykin AJ, Nho K. Cell-specific transcriptional signatures of vascular cells in Alzheimer's disease: perspectives, pathways, and therapeutic directions. Mol Neurodegener 2025; 20:12. [PMID: 39876020 PMCID: PMC11776188 DOI: 10.1186/s13024-025-00798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disease that is marked by profound neurovascular dysfunction and significant cell-specific alterations in the brain vasculature. Recent advances in high throughput single-cell transcriptomics technology have enabled the study of the human brain vasculature at an unprecedented depth. Additionally, the understudied niche of cerebrovascular cells, such as endothelial and mural cells, and their subtypes have been scrutinized for understanding cellular and transcriptional heterogeneity in AD. Here, we provide an overview of rich transcriptional signatures derived from recent single-cell and single-nucleus transcriptomic studies of human brain vascular cells and their implications for targeted therapy for AD. We conducted an in-depth literature search using Medline and Covidence to identify pertinent AD studies that utilized single-cell technologies in human post-mortem brain tissue by focusing on understanding the transcriptional differences in cerebrovascular cell types and subtypes in AD and cognitively normal older adults. We also discuss impaired cellular crosstalk between vascular cells and neuroglial units, as well as astrocytes in AD. Additionally, we contextualize the findings from single-cell studies of distinct endothelial cells, smooth muscle cells, fibroblasts, and pericytes in the human AD brain and highlight pathways for potential therapeutic interventions as a concerted multi-omic effort with spatial transcriptomics technology, neuroimaging, and neuropathology. Overall, we provide a detailed account of the vascular cell-specific transcriptional signatures in AD and their crucial cellular crosstalk with the neuroglial unit.
Collapse
Affiliation(s)
- Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Minyoung Cho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Julia C Stumpff
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula J Bice
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Özkan İş
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
21
|
Ferreira da Silva A, Gomes A, Gonçalves LMD, Fernandes A, Almeida AJ. Exploring the Link Between Periodontitis and Alzheimer's Disease-Could a Nanoparticulate Vaccine Break It? Pharmaceutics 2025; 17:141. [PMID: 40006510 PMCID: PMC11858903 DOI: 10.3390/pharmaceutics17020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, as approximately 55 million people worldwide are affected, with a significant tendency to increase. It reveals three main pathological features: amyloid plaques, neurofibrillary tangles, and neuroinflammation, responsible for the neurodegenerative changes that slowly lead to deterioration of personality and cognitive control. Over a century after the first case report, effective treatments remain elusive, likely due to an incomplete understanding of the precise mechanisms driving its pathogenesis. Recent studies provide growing evidence of an infectious aetiology for AD, a hypothesis reinforced by findings that amyloid beta functions as an antimicrobial peptide. Among the microorganisms already associated with AD, Porphyromonas gingivalis (Pg), the keystone pathogen of periodontitis (PeD), has received particular attention as a possible aetiological agent for AD development. Herein, we review the epidemiological and genetic evidence linking PeD and Pg to AD, highlighting the identification of periodontal bacteria in post mortem analysis of AD patients' brains and identifying putative mechanistic links relevant to the biological plausibility of the association. With the focus on AD research shifting from cure to prevention, the proposed mechanisms linking PeD to AD open the door for unravelling new prophylactic approaches able to reduce the global burden of AD. As hypothesised in this review, these could include a bionanotechnological approach involving the development of an oral nanoparticulate vaccine based on Pg-specific antigens. Such a vaccine could prevent Pg antigens from progressing to the brain and triggering AD pathology, representing a promising step toward innovative and effective AD prevention.
Collapse
Affiliation(s)
| | | | | | | | - António J. Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal; (A.F.d.S.); (A.G.); (L.M.D.G.); (A.F.)
| |
Collapse
|
22
|
Liu X, Lv Z, Huang Q, Lei Y, Liu H, Xu P. The Role of Oligodendrocyte Lineage Cells in the Pathogenesis of Alzheimer's Disease. Neurochem Res 2025; 50:72. [PMID: 39751972 DOI: 10.1007/s11064-024-04325-3] [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: 07/29/2024] [Revised: 10/06/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is a central nervous system degenerative disease with a stealthy onset and a progressive course characterized by memory loss, cognitive dysfunction, and abnormal psychological and behavioral symptoms. However, the pathogenesis of AD remains elusive. An increasing number of studies have shown that oligodendrocyte progenitor cells (OPCs) and oligodendroglial lineage cells (OLGs), especially OPCs and mature oligodendrocytes (OLGs), which are derived from OPCs, play important roles in the pathogenesis of AD. OLGs function mainly by myelinating axons, transmitting electrical signals, and regulating neural development. In addition to myelin, OPCs and OLGs can also participate in AD pathogenesis in other ways. This review summarizes the mechanisms by which OPCs and OLGs affect myelin formation, oxidative stress, neuroinflammation, the blood-brain barrier, synaptic function, and amyloid-beta protein and further elucidates the mechanisms by which oligodendrocyte lineage cells participate in AD pathogenesis and treatment, which is highly important for clarifying the pathogenesis of AD, clinical treatment, and prevention.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Neurology, China Guihang Group 302 Hospital, Anshun, China
| | - Zhengxiang Lv
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Neurology, China Guihang Group 302 Hospital, Anshun, China
| | - Qin Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yihui Lei
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Haijun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
23
|
Maridaki Z, Syrros G, Gianna Delichatsiou S, Warsh J, Konstantinou GN. Claudin-5 and occludin levels in patients with psychiatric disorders - A systematic review. Brain Behav Immun 2025; 123:865-875. [PMID: 39500414 DOI: 10.1016/j.bbi.2024.11.006] [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: 05/13/2024] [Revised: 10/14/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Recent research has underscored the critical role of blood-brain barrier (BBB) integrity in psychiatric disorders, highlighting disruptions in tight junction (TJ) proteins, specifically claudin-5 and occludin. These proteins are pivotal in maintaining the BBB's selective permeability, which is essential forbrain homeostasis. Altered levels of the TJ proteins have been observed in various psychiatric conditions, suggesting potential as biomarkers for the pathophysiology of these disorders. This systematic review synthesizes existing research on the alterations of claudin-5 and occludin levels in the serum of individuals with psychiatric disorders, evaluating their correlation with BBB dysfunction and psychiatric pathophysiology. METHODS In adherence to the PRISMA guidelines, a comprehensive search strategy was employed, utilizing databases such as PubMed, Google Scholar, Web of Science, and Scopus. The review encompassed studies published between 2000 and 2024 that measured serum claudin-5 and occludin levels of psychiatric patients. Thorough data extraction and synthesis were conducted. RESULTS Seventeen studies met the inclusion criteria. Key findings include indications for increased claudin-5 levels in Schizophrenia, Bipolar Disorder, Depression, and Specific learning disorder, and increased occludin levels in ADHD and Autism Spectrum Disorder patients. No significant differences were found in studies of patients with Alcohol Use and Insomnia Disorder. CONCLUSIONS The review underscores the potential association between altered serum levels of claudin-5 and occludin and psychiatric disorders, supporting their utility as biomarkers for BBB integrity and psychiatric pathophysiology. Further research is essential to elucidate the mechanisms linking TJ protein alterations with pathophysiology and, potentially, neuroprogression in psychiatric disorders, which could lead to novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zinovia Maridaki
- 1(st) Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Greece
| | - Georgios Syrros
- 2(nd) Department of Psychiatry, Attikon Hospital, National and Kapodistrian University of Athens, Greece
| | | | - Jerry Warsh
- Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health, Toronto, Canada
| | - Gerasimos N Konstantinou
- Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health, Toronto, Canada; Poul Hansen Family Centre for Depression, Centre of Mental Health, University Health Network, Toronto, Canada.
| |
Collapse
|
24
|
Reas ET, Solders SK, Tsiknia A, Triebswetter C, Shen Q, Rivera CS, Andrews MJ, Alderson-Myers A, Brewer JB. APOE 𝜀4-related blood-brain barrier breakdown is associated with microstructural abnormalities. Alzheimers Dement 2024; 20:8615-8624. [PMID: 39411970 DOI: 10.1002/alz.14302] [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: 06/25/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Blood-brain barrier (BBB) dysfunction occurs in Alzheimer's disease (AD). Yet, the stage at which it appears along the AD time course and whether it contributes to neurodegeneration remain unclear. METHODS Older adults (61 to 90 years) from cognitively normal (CN) to mildly cognitively impaired (CI), enriched for APOE 𝜀4 and amyloid positivity, underwent dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) and diffusion MRI to measure BBB permeability and brain microstructure. Analysis of variance compared BBB permeability according to cognitive status, amyloid beta (Aβ), and APOE4. Linear regressions assessed associations of BBB permeability with brain microstructure and interactions with Aβ and APOE4. RESULTS BBB permeability was elevated for APOE4 carriers across the cortical gray matter, with the strongest differences among CN amyloid-negative individuals. Associations between entorhinal BBB permeability and microstructure interacted with Aβ and APOE4, with the strongest relationships in amyloid-positive individuals and APOE4 carriers. DISCUSSION APOE4 may drive widespread BBB dysfunction in preclinical AD, which may contribute to neurodegenerative changes early along the AD cascade. HIGHLIGHTS Gray matter blood-brain barrier (BBB) permeability is elevated for APOE4 carriers. APOE4-related BBB breakdown appears in the absence of cognitive decline or amyloid. BBB leakage correlates with entorhinal cortex microstructural injury. Associations with microstructure are strongest for amyloid-positive APOE4 carriers.
Collapse
Affiliation(s)
- Emilie T Reas
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Seraphina K Solders
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Amaryllis Tsiknia
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Marina Del Rey, California, USA
| | | | - Qian Shen
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Charlotte S Rivera
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Murray J Andrews
- Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Austin Alderson-Myers
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - James B Brewer
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
25
|
Morgan CA, Thomas DL, Shao X, Mahroo A, Manson TJ, Suresh V, Jansson D, Ohene Y, Günther M, Wang DJJ, Tippett LJ, Dragunow M. Measurement of blood-brain barrier water exchange rate using diffusion-prepared and multi-echo arterial spin labelling: Comparison of quantitative values and age dependence. NMR IN BIOMEDICINE 2024; 37:e5256. [PMID: 39252500 DOI: 10.1002/nbm.5256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
Water exchange rate (Kw) across the blood-brain barrier (BBB) is an important physiological parameter that may provide new insight into ageing and neurodegenerative disease. Recently, two non-invasive arterial spin labelling (ASL) MRI methods have been developed to measure Kw, but results from the different methods have not been directly compared. Furthermore, the association of Kw with age for each method has not been investigated in a single cohort. Thirty participants (70% female, 63.8 ± 10.4 years) were scanned at 3 T with Diffusion-Prepared ASL (DP-ASL) and Multi-Echo ASL (ME-ASL) using previously implemented acquisition and analysis protocols. Grey matter Kw, cerebral blood flow (CBF) and arterial transit time (ATT) were extracted. CBF values were consistent; approximately 50 ml/min/100 g for both methods, and a strong positive correlation in CBF from both methods across participants (r = 0.82, p < 0.001). ATT was significantly different between methods (on average 147.7 ms lower when measured with DP-ASL compared to ME-ASL) but was positively correlated across participants (r = 0.39, p < 0.05). Significantly different Kw values of 106.6 ± 19.7 min-1 and 306.8 ± 71.7 min-1 were measured using DP-ASL and ME-ASL, respectively, and DP-ASL Kw and ME-ASL Kw were negatively correlated across participants (r = -0.46, p < 0.01). Kw measured using ME-ASL had a significant linear relationship with age (p < 0.05). In conclusion, DP-ASL and ME-ASL provided estimates of Kw with significantly different quantitative values and inconsistent dependence with age. We propose future standardisation of modelling and fitting methods for DP-ASL and ME-ASL, to evaluate the effect on Kw quantification. Also, sensitivity and bias analyses should be performed for both approaches, to assess the effect of varying acquisition and fitting parameters. Lastly, comparison with independent measures of BBB water transport, and with physiological and clinical biomarkers known to be associated with changes in BBB permeability, are essential to validate the ASL methods, and to demonstrate their clinical utility.
Collapse
Affiliation(s)
- Catherine A Morgan
- School of Psychology and Centre for Brain Research, University of Auckland, New Zealand
- Centre for Advanced MRI, Auckland Uniservices Limited, University of Auckland, New Zealand
| | - David L Thomas
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, California, Los Angeles, USA
| | - Amnah Mahroo
- Imaging Physics, Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Tabitha J Manson
- School of Psychology and Centre for Brain Research, University of Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Vinod Suresh
- Auckland Bioengineering Institute, University of Auckland, New Zealand
- Department of Engineering Science and Biomedical Engineering, University of Auckland, New Zealand
| | - Deidre Jansson
- Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, WA, USA
- School of Biological Sciences, Faculty of Science, University of Auckland, New Zealand
| | - Yolanda Ohene
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Matthias Günther
- Imaging Physics, Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, California, Los Angeles, USA
| | - Lynette J Tippett
- School of Psychology and Centre for Brain Research, University of Auckland, New Zealand
- Dementia Prevention Research Clinic, University of Auckland, New Zealand
| | - Michael Dragunow
- Department of Pharmacology and Centre for Brain Research, University of Auckland, New Zealand
| |
Collapse
|
26
|
Qiu J, Peng S, Qu R, Wu L, Xing L, Zhang L, Sun J. New evidence of vascular defects in neurodegenerative diseases revealed by single cell RNA sequencing. Clin Sci (Lond) 2024; 138:1377-1394. [PMID: 39469930 DOI: 10.1042/cs20241658] [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: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Neurodegenerative diseases (NDs) involve the progressive loss of neuronal structure or function in the brain and spinal cord. Despite their diverse etiologies, NDs manifest similar pathologies. Emerging research identifies vascular defects as a previously neglected hallmark of NDs. The development and popularization of single-cell RNA sequencing (scRNA-seq) technologies have significantly advanced our understanding of brain vascular cell types and their molecular characteristics, including gene expression changes at the single-cell level in NDs. These unprecedented insights deepen our understanding of the pathogenic mechanisms underlying NDs. However, the occurrence and role of vascular defects in disease progression remain largely unexplored. In this paper, we systematically summarize recent advances in the structure and organization of the central nervous system vasculature in mice, healthy individuals, and patients with NDs, focussing primarily on disease-specific alterations in vascular cell types or subtypes. Combining scRNA-seq with pathology evidence, we propose that vascular defects, characterized by disruptions in cell types and structural integrity, may serve as common early features of NDs. Finally, we discuss several pathways through which vascular defects in NDs lead to neuronal degeneration. A deeper understanding of the causes and contributions of vascular defects to NDs aids in elucidating the pathogenic mechanisms and developing meaningful therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
27
|
Kim TK, Hong JM, Kim J, Kim KH, Han SJ, Kim IC, Oh H, Jo DG, Yim JH. Therapeutic Potential of Ramalin Derivatives with Enhanced Stability in the Treatment of Alzheimer's Disease. Molecules 2024; 29:5223. [PMID: 39598614 PMCID: PMC11597085 DOI: 10.3390/molecules29225223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) remains a significant public health challenge with limited effective treatment options. Ramalin, a compound derived from Antarctic lichens, has shown potential in the treatment of AD because of its strong antioxidant and anti-inflammatory properties. However, its instability and toxicity have hindered the development of Ramalin as a viable therapeutic agent. The primary objective of this study was to synthesize and evaluate novel Ramalin derivatives with enhanced stabilities and reduced toxic profiles, with the aim of retaining or improving their therapeutic potential against AD. The antioxidant, anti-inflammatory, anti-BACE-1, and anti-tau activities of four synthesized Ramalin derivatives (i.e., RA-Hyd-Me, RA-Hyd-Me-Tol, RA-Sali, and RA-Benzo) were evaluated. These derivatives demonstrated significantly improved stabilities compared to the parent compound, with RA-Sali giving the most promising results. More specifically, RA-Sali exhibited a potent BACE-1 inhibitory activity and effectively reduced tau phosphorylation, a critical factor in AD pathology. Despite exhibiting reduced antioxidant activities compared to the parent compound, these derivatives represent a potential multi-targeted approach for AD treatment, marking a significant step forward in the development of stable and effective AD therapeutics.
Collapse
Affiliation(s)
- Tai Kyoung Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Jaewon Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
- Department of Plant Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Hee Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
- Department of Chemistry, Hanseo University, Seosan 31962, Republic of Korea
| | - Se Jong Han
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Il-Chan Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea;
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Joung Han Yim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| |
Collapse
|
28
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
29
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
30
|
Chetty CA, Bhardwaj H, Kumar GP, Devanand T, Sekhar CSA, Aktürk T, Kiyi I, Yener G, Güntekin B, Joseph J, Adaikkan C. EEG biomarkers in Alzheimer's and prodromal Alzheimer's: a comprehensive analysis of spectral and connectivity features. Alzheimers Res Ther 2024; 16:236. [PMID: 39449097 PMCID: PMC11515355 DOI: 10.1186/s13195-024-01582-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Biomarkers of Alzheimer's disease (AD) and mild cognitive impairment (MCI, or prodromal AD) are highly significant for early diagnosis, clinical trials and treatment outcome evaluations. Electroencephalography (EEG), being noninvasive and easily accessible, has recently been the center of focus. However, a comprehensive understanding of EEG in dementia is still needed. A primary objective of this study is to investigate which of the many EEG characteristics could effectively differentiate between individuals with AD or prodromal AD and healthy individuals. METHODS We collected resting state EEG data from individuals with AD, prodromal AD, and normal cognition. Two distinct preprocessing pipelines were employed to study the reliability of the extracted measures across different datasets. We extracted 41 different EEG features. We have also developed a stand-alone software application package, Feature Analyzer, as a comprehensive toolbox for EEG analysis. This tool allows users to extract 41 EEG features spanning various domains, including complexity measures, wavelet features, spectral power ratios, and entropy measures. We performed statistical tests to investigate the differences in AD or prodromal AD from age-matched cognitively normal individuals based on the extracted EEG features, power spectral density (PSD), and EEG functional connectivity. RESULTS Spectral power ratio measures such as theta/alpha and theta/beta power ratios showed significant differences between cognitively normal and AD individuals. Theta power was higher in AD, suggesting a slowing of oscillations in AD; however, the functional connectivity of the theta band was decreased in AD individuals. In contrast, we observed increased gamma/alpha power ratio, gamma power, and gamma functional connectivity in prodromal AD. Entropy and complexity measures after correcting for multiple electrode comparisons did not show differences in AD or prodromal AD groups. We thus catalogued AD and prodromal AD-specific EEG features. CONCLUSIONS Our findings reveal that the changes in power and connectivity in certain frequency bands of EEG differ in prodromal AD and AD. The spectral power, power ratios, and the functional connectivity of theta and gamma could be biomarkers for diagnosis of AD and prodromal AD, measure the treatment outcome, and possibly a target for brain stimulation.
Collapse
Affiliation(s)
| | - Harsha Bhardwaj
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - G Pradeep Kumar
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India
| | - T Devanand
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India
| | - C S Aswin Sekhar
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India
| | - Tuba Aktürk
- Neuroscience Research Center, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Ilayda Kiyi
- Department of Neuroscience, Health Sciences Institute, Dokuz Eylül University, Izmir, 35330, Turkey
| | - Görsev Yener
- Faculty of Medicine, Izmir University of Economics, Izmir, 35330, Turkey
- Brain Dynamics Research Center, Dokuz Eylül University, Izmir, 35330, Turkey
- Biomedicine and Genome Center, Izmir, 35340, Turkey
| | - Bahar Güntekin
- Neuroscience Research Center, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Biophysics, School of Medicine, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Justin Joseph
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India
| | - Chinnakkaruppan Adaikkan
- Centre for Brain Research, Indian Institute of Science, CV Raman Avenue, Bangalore, 560 012, India.
| |
Collapse
|
31
|
Dan YR, Chiam KH. Discovery of plasma biomarkers related to blood-brain barrier dysregulation in Alzheimer's disease. FRONTIERS IN BIOINFORMATICS 2024; 4:1463001. [PMID: 39429696 PMCID: PMC11487119 DOI: 10.3389/fbinf.2024.1463001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Blood-based biomarkers are quantitative, non-invasive diagnostic tools. This study aimed to identify candidate biomarkers for Alzheimer's disease (AD) using publicly available omics datasets, using the hypothesis that with blood-brain barrier dysfunction in AD, brain-synthesized proteins can leak into plasma for detection. Methods Differential abundance results of plasma and brain proteomic datasets were integrated to obtain a list of potential biomarkers. Biological validity was investigated with intercellular communication and gene regulatory analyses on brain single-cell transcriptomics data. Results Five proteins (APOD, B2M, CFH, CLU, and C3) fit biomarker criteria. 4 corresponding transcripts (APOD, B2M, CLU, and C3) were overexpressed in AD astrocytes, mediated by AD-related dysregulations in transcription factors regulating neuroinflammation. Additionally, CLU specifically induced downstream expression of neuronal death genes. Discussion In conclusion, a 5-protein panel is shown to effectively identify AD patients, with evidence of disease specificity and biological validity. Future research should investigate the mechanism of protein leakage through the blood-brain barrier.
Collapse
Affiliation(s)
- Yuet Ruh Dan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| | - Keng-Hwee Chiam
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| |
Collapse
|
32
|
Gulej R, Nyúl-Tóth Á, Csik B, Patai R, Petersen B, Negri S, Chandragiri SS, Shanmugarama S, Mukli P, Yabluchanskiy A, Conley S, Huffman D, Tarantini S, Csiszar A, Ungvari Z. Young blood-mediated cerebromicrovascular rejuvenation through heterochronic parabiosis: enhancing blood-brain barrier integrity and capillarization in the aged mouse brain. GeroScience 2024; 46:4415-4442. [PMID: 38727872 PMCID: PMC11336025 DOI: 10.1007/s11357-024-01154-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/05/2024] [Indexed: 06/15/2024] Open
Abstract
Age-related cerebromicrovascular changes, including blood-brain barrier (BBB) disruption and microvascular rarefaction, play a significant role in the development of vascular cognitive impairment (VCI) and neurodegenerative diseases. Utilizing the unique model of heterochronic parabiosis, which involves surgically joining young and old animals, we investigated the influence of systemic factors on these vascular changes. Our study employed heterochronic parabiosis to explore the effects of young and aged systemic environments on cerebromicrovascular aging in mice. We evaluated microvascular density and BBB integrity in parabiotic pairs equipped with chronic cranial windows, using intravital two-photon imaging techniques. Our results indicate that short-term exposure to young systemic factors leads to both functional and structural rejuvenation of cerebral microcirculation. Notably, we observed a marked decrease in capillary density and an increase in BBB permeability to fluorescent tracers in the cortices of aged mice undergoing isochronic parabiosis (20-month-old C57BL/6 mice [A-(A)]; 6 weeks of parabiosis), compared to young isochronic parabionts (6-month-old, [Y-(Y)]). However, aged heterochronic parabionts (A-(Y)) exposed to young blood exhibited a significant increase in cortical capillary density and restoration of BBB integrity. In contrast, young mice exposed to old blood from aged parabionts (Y-(A)) rapidly developed cerebromicrovascular aging traits, evidenced by reduced capillary density and increased BBB permeability. These findings underscore the profound impact of systemic factors in regulating cerebromicrovascular aging. The rejuvenation observed in the endothelium, following exposure to young blood, suggests the existence of anti-geronic elements that counteract microvascular aging. Conversely, pro-geronic factors in aged blood appear to accelerate cerebromicrovascular aging. Further research is needed to assess whether the rejuvenating effects of young blood factors could extend to other age-related cerebromicrovascular pathologies, such as microvascular amyloid deposition and increased microvascular fragility.
Collapse
Affiliation(s)
- Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Benjamin Petersen
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sharon Negri
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Siva Sai Chandragiri
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Santny Shanmugarama
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Shannon Conley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Derek Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
33
|
El-Ghazawi K, Eyo UB, Peirce SM. Brain Microvascular Pericyte Pathology Linking Alzheimer's Disease to Diabetes. Microcirculation 2024; 31:e12877. [PMID: 39222475 PMCID: PMC11471384 DOI: 10.1111/micc.12877] [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: 03/13/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 09/04/2024]
Abstract
The brain microvasculature, which delivers oxygen and nutrients and forms a critical barrier protecting the central nervous system via capillaries, is deleteriously affected by both Alzheimer's disease (AD) and type 2 diabetes (T2D). T2D patients have an increased risk of developing AD, suggesting potentially related microvascular pathological mechanisms. Pericytes are an ideal cell type to study for functional links between AD and T2D. These specialized capillary-enwrapping cells regulate capillary density, lumen diameter, and blood flow. Pericytes also maintain endothelial tight junctions to ensure blood-brain barrier integrity, modulation of immune cell extravasation, and clearance of toxins. Changes in these phenomena have been observed in both AD and T2D, implicating "pericyte pathology" as a common feature of AD and T2D. This review examines the mechanisms of AD and T2D from the perspective of the brain microvasculature, highlighting how pericyte pathology contributes to both diseases. Our review identifies voids in understanding how AD and T2D negatively impact the brain microvasculature and suggests future studies to examine the intersections of these diseases.
Collapse
Affiliation(s)
- Kareem El-Ghazawi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Department of Neuroscience, University of Virginia Center for Brain Immunology and Glia School of Medicine, Charlottesville, VA, USA
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
34
|
Zedde M, Pascarella R. The Cerebrovascular Side of Plasticity: Microvascular Architecture across Health and Neurodegenerative and Vascular Diseases. Brain Sci 2024; 14:983. [PMID: 39451997 PMCID: PMC11506257 DOI: 10.3390/brainsci14100983] [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: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The delivery of nutrients to the brain is provided by a 600 km network of capillaries and microvessels. Indeed, the brain is highly energy demanding and, among a total amount of 100 billion neurons, each neuron is located just 10-20 μm from a capillary. This vascular network also forms part of the blood-brain barrier (BBB), which maintains the brain's stable environment by regulating chemical balance, immune cell transport, and blocking toxins. Typically, brain microvascular endothelial cells (BMECs) have low turnover, indicating a stable cerebrovascular structure. However, this structure can adapt significantly due to development, aging, injury, or disease. Temporary neural activity changes are managed by the expansion or contraction of arterioles and capillaries. Hypoxia leads to significant remodeling of the cerebrovascular architecture and pathological changes have been documented in aging and in vascular and neurodegenerative conditions. These changes often involve BMEC proliferation and the remodeling of capillary segments, often linked with local neuronal changes and cognitive function. Cerebrovascular plasticity, especially in arterioles, capillaries, and venules, varies over different time scales in development, health, aging, and diseases. Rapid changes in cerebral blood flow (CBF) occur within seconds due to increased neural activity. Prolonged changes in vascular structure, influenced by consistent environmental factors, take weeks. Development and aging bring changes over months to years, with aging-associated plasticity often improved by exercise. Injuries cause rapid damage but can be repaired over weeks to months, while neurodegenerative diseases cause slow, varied changes over months to years. In addition, if animal models may provide useful and dynamic in vivo information about vascular plasticity, humans are more complex to investigate and the hypothesis of glymphatic system together with Magnetic Resonance Imaging (MRI) techniques could provide useful clues in the future.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
35
|
Greene C, Rebergue N, Fewell G, Janigro D, Godfrin Y, Campbell M, Lemarchant S. NX210c drug candidate peptide strengthens mouse and human blood-brain barriers. Fluids Barriers CNS 2024; 21:76. [PMID: 39334382 PMCID: PMC11438064 DOI: 10.1186/s12987-024-00577-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Alterations of blood-brain barrier (BBB) and blood-spinal cord barrier have been documented in various animal models of neurodegenerative diseases and in patients. Correlations of these alterations with functional deficits suggest that repairing barriers integrity may represent a disease-modifying approach to prevent neuroinflammation and neurodegeneration induced by the extravasation of blood components into the parenchyma. Here, we screened the effect of a subcommissural organ-spondin-derived peptide (NX210c), known to promote functional recovery in several models of neurological disorders, on BBB integrity in vitro and in vivo. METHODS In vitro, bEnd.3 endothelial cell (EC) monolayers and two different primary human BBB models containing EC, astrocytes and pericytes, in static and microfluidic conditions, were treated with NX210c (1-100 µM), or its vehicle, for 4 h and up to 5 days. Tight junction (TJ) protein levels, permeability to dextrans and transendothelial electrical resistance (TEER) were evaluated. In vivo, young and old mice (3- and 21-month-old, respectively) were treated daily intraperitoneally with NX210c at 10 mg/kg or its vehicle for 5 days and their brains collected at day 6 to measure TJ protein levels by immunohistochemistry. RESULTS NX210c induced an increase in claudin-5 protein expression after 24-h and 72-h treatments in mouse EC. Occludin level was also increased after a 24-h treatment. Accordingly, NX210c decreased by half the permeability of EC to a 40-kDa FITC-dextran and increased TEER. In the human static BBB model, NX210c increased by ∼ 25% the TEER from 3 to 5 days. NX210c also increased TEER in the human 3D dynamic BBB model after 4 h, which was associated with a reduced permeability to a 4-kDa FITC-dextran. In line with in vitro results, after only 5 days of daily treatments in mice, NX210c restored aging-induced reduction of claudin-5 and occludin levels in the hippocampus, and also in the cortex for occludin. CONCLUSIONS In summary, we have gathered preclinical data showing the capacity of NX210c to strengthen BBB integrity. Through this property, NX210c holds great promises of being a disease-modifying treatment for several neurological disorders with high unmet medical needs.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | | | | | | - Yann Godfrin
- Axoltis Pharma, 60 avenue Rockefeller, Lyon, 69008, France
- Godfrin Life-Sciences, Caluire-et-Cuire, 69300, France
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | |
Collapse
|
36
|
Zheng Y, Cai X, Wang D, Chen X, Wang T, Xie Y, Li H, Wang T, He Y, Li J, Li J. Exploring the relationship between lipid metabolism and cognition in individuals living with stable-phase Schizophrenia: a small cross-sectional study using Olink proteomics analysis. BMC Psychiatry 2024; 24:593. [PMID: 39227832 PMCID: PMC11370234 DOI: 10.1186/s12888-024-06054-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Cognitive impairment is a core symptom of schizophrenia. Metabolic abnormalities impact cognition, and although the influence of blood lipids on cognition has been documented, it remains unclear. We conducted a small cross-sectional study to investigate the relationship between blood lipids and cognition in patients with stable-phase schizophrenia. Using Olink proteomics, we explored the potential mechanisms through which blood lipids might affect cognition from an inflammatory perspective. METHODS A total of 107 patients with stable-phase schizophrenia and cognitive impairment were strictly included. Comprehensive data collection included basic patient information, blood glucose, blood lipids, and body mass index. Cognitive function was assessed using the Montreal Cognitive Assessment (MoCA) and the MATRICS Consensus Cognitive Battery (MCCB). After controlling for confounding factors, we identified differential metabolic indicators between patients with mild and severe cognitive impairment and conducted correlation and regression analyses. Furthermore, we matched two small sample groups of patients with lipid metabolism abnormalities and used Olink proteomics to analyze inflammation-related differential proteins, aiming to further explore the association between lipid metabolism abnormalities and cognition. RESULTS The proportion of patients with severe cognitive impairment (SCI) was 34.58%. Compared to patients with mild cognitive impairment (MCI), those with SCI performed worse in the Attention/Alertness (t = 2.668, p = 0.009) and Working Memory (t = 2.496, p = 0.014) cognitive dimensions. Blood lipid metabolism indicators were correlated with cognitive function, specifically showing that higher levels of TG (r = -0.447, p < 0.001), TC (r = -0.307, p = 0.002), and LDL-C (r = -0.607, p < 0.001) were associated with poorer overall cognitive function. Further regression analysis indicated that TG (OR = 5.578, P = 0.003) and LDL-C (OR = 5.425, P = 0.001) may be risk factors for exacerbating cognitive impairment in individuals with stable-phase schizophrenia. Proteomics analysis revealed that, compared to individuals with stable-phase schizophrenia and normal lipid metabolism, those with hyperlipidemia had elevated levels of 10 inflammatory proteins and decreased levels of 2 inflammatory proteins in plasma, with these changes correlating with cognitive function. The differential proteins were primarily involved in pathways such as cytokine-cytokine receptor interaction, chemokine signaling pathway, and IL-17 signaling pathway. CONCLUSION Blood lipids are associated with cognitive function in individuals with stable-phase schizophrenia, with higher levels of TG, TC, and LDL-C correlating with poorer overall cognitive performance. TG and LDL-C may be risk factors for exacerbating cognitive impairment in these patients. From an inflammatory perspective, lipid metabolism abnormalities might influence cognition by activating or downregulating related proteins, or through pathways such as cytokine-cytokine receptor interaction, chemokine signaling pathway, and IL-17 signaling pathway.
Collapse
Affiliation(s)
- Yingkang Zheng
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaojun Cai
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China.
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China.
| | - Dezhong Wang
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Xinghai Chen
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Tao Wang
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Yanpeng Xie
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Haojing Li
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Tong Wang
- Department of Endocrinology, Heilongjiang Academy of Chinese Medicine, Harbin, China
| | - Yinxiong He
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiarui Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Juan Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
37
|
van Osch MJP, Wåhlin A, Scheyhing P, Mossige I, Hirschler L, Eklund A, Mogensen K, Gomolka R, Radbruch A, Qvarlander S, Decker A, Nedergaard M, Mori Y, Eide PK, Deike K, Ringstad G. Human brain clearance imaging: Pathways taken by magnetic resonance imaging contrast agents after administration in cerebrospinal fluid and blood. NMR IN BIOMEDICINE 2024; 37:e5159. [PMID: 38634301 DOI: 10.1002/nbm.5159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024]
Abstract
Over the last decade, it has become evident that cerebrospinal fluid (CSF) plays a pivotal role in brain solute clearance through perivascular pathways and interactions between the brain and meningeal lymphatic vessels. Whereas most of this fundamental knowledge was gained from rodent models, human brain clearance imaging has provided important insights into the human system and highlighted the existence of important interspecies differences. Current gold standard techniques for human brain clearance imaging involve the injection of gadolinium-based contrast agents and monitoring their distribution and clearance over a period from a few hours up to 2 days. With both intrathecal and intravenous injections being used, which each have their own specific routes of distribution and thus clearance of contrast agent, a clear understanding of the kinetics associated with both approaches, and especially the differences between them, is needed to properly interpret the results. Because it is known that intrathecally injected contrast agent reaches the blood, albeit in small concentrations, and that similarly some of the intravenously injected agent can be detected in CSF, both pathways are connected and will, in theory, reach the same compartments. However, because of clear differences in relative enhancement patterns, both injection approaches will result in varying sensitivities for assessment of different subparts of the brain clearance system. In this opinion review article, the "EU Joint Programme - Neurodegenerative Disease Research (JPND)" consortium on human brain clearance imaging provides an overview of contrast agent pharmacokinetics in vivo following intrathecal and intravenous injections and what typical concentrations and concentration-time curves should be expected. This can be the basis for optimizing and interpreting contrast-enhanced MRI for brain clearance imaging. Furthermore, this can shed light on how molecules may exchange between blood, brain, and CSF.
Collapse
Affiliation(s)
- Matthias J P van Osch
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Anders Wåhlin
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Paul Scheyhing
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ingrid Mossige
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, The Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lydiane Hirschler
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Anders Eklund
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Klara Mogensen
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Ryszard Gomolka
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Radbruch
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sara Qvarlander
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Andreas Decker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Katerina Deike
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Department of Geriatrics and Internal Medicine, Sorlandet Hospital, Arendal, Norway
| |
Collapse
|
38
|
Hossen F, Sun GY, Lee JC. Oligomeric Tau-induced oxidative damage and functional alterations in cerebral endothelial cells: Role of RhoA/ROCK signaling pathway. Free Radic Biol Med 2024; 221:261-272. [PMID: 38815773 PMCID: PMC11184584 DOI: 10.1016/j.freeradbiomed.2024.05.044] [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: 01/17/2024] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Despite of yet unknown mechanism, microvascular deposition of oligomeric Tau (oTau) has been implicated in alteration of the Blood-Brain Barrier (BBB) function in Alzheimer's disease (AD) brains. In this study, we employed an in vitro BBB model using primary mouse cerebral endothelial cells (CECs) to investigate the mechanism underlying the effects of oTau on BBB function. We found that exposing CECs to oTau induced oxidative stress through NADPH oxidase, increased oxidative damage to proteins, decreased proteasome activity, and expressions of tight junction (TJ) proteins including occludin, zonula occludens-1 (ZO-1) and claudin-5. These effects were suppressed by the pretreatment with Fasudil, a RhoA/ROCK signaling inhibitor. Consistent with the biochemical alterations, we found that exposing the basolateral side of CECs to oTau in the BBB model disrupted the integrity of the BBB, as indicated by an increase in FITC-dextran transport across the model, and a decrease in trans endothelial electrical resistance (TEER). oTau also increased the transmigration of peripheral blood mononuclear cells (PBMCs) in the BBB model. These functional alterations in the BBB induced by oTau were also suppressed by Fasudil. Taken together, our findings suggest that targeting the RhoA/ROCK pathway can be a potential therapeutic strategy to maintain BBB function in AD.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
39
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [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: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Rather MA, Khan A, Jahan S, Siddiqui AJ, Wang L. Influence of Tau on Neurotoxicity and Cerebral Vasculature Impairment Associated with Alzheimer's Disease. Neuroscience 2024; 552:1-13. [PMID: 38871021 DOI: 10.1016/j.neuroscience.2024.05.042] [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: 12/10/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's disease is a fatal chronic neurodegenerative condition marked by a gradual decline in cognitive abilities and impaired vascular function within the central nervous system. This affliction initiates its insidious progression with the accumulation of two aberrant protein entities including Aβ plaques and neurofibrillary tangles. These chronic elements target distinct brain regions, steadily erasing the functionality of the hippocampus and triggering the erosion of memory and neuronal integrity. Several assumptions are anticipated for AD as genetic alterations, the occurrence of Aβ plaques, altered processing of amyloid precursor protein, mitochondrial damage, and discrepancy of neurotropic factors. In addition to Aβ oligomers, the deposition of tau hyper-phosphorylates also plays an indispensable part in AD etiology. The brain comprises a complex network of capillaries that is crucial for maintaining proper function. Tau is expressed in cerebral blood vessels, where it helps to regulate blood flow and sustain the blood-brain barrier's integrity. In AD, tau pathology can disrupt cerebral blood supply and deteriorate the BBB, leading to neuronal neurodegeneration. Neuroinflammation, deficits in the microvasculature and endothelial functions, and Aβ deposition are characteristically detected in the initial phases of AD. These variations trigger neuronal malfunction and cognitive impairment. Intracellular tau accumulation in microglia and astrocytes triggers deleterious effects on the integrity of endothelium and cerebral blood supply resulting in further advancement of the ailment and cerebral instability. In this review, we will discuss the impact of tau on neurovascular impairment, mitochondrial dysfunction, oxidative stress, and the role of hyperphosphorylated tau in neuron excitotoxicity and inflammation.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States.
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail City, Saudi Arabia
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States
| |
Collapse
|
41
|
Le QV, Shim G. Biorobotic Drug Delivery for Biomedical Applications. Molecules 2024; 29:3663. [PMID: 39125066 PMCID: PMC11314275 DOI: 10.3390/molecules29153663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 08/12/2024] Open
Abstract
Despite extensive efforts, current drug-delivery systems face biological barriers and difficulties in bench-to-clinical use. Biomedical robotic systems have emerged as a new strategy for drug delivery because of their innovative diminutive engines. These motors enable the biorobots to move independently rather than relying on body fluids. The main components of biorobots are engines controlled by external stimuli, chemical reactions, and biological responses. Many biorobot designs are inspired by blood cells or microorganisms that possess innate swimming abilities and can incorporate living materials into their structures. This review explores the mechanisms of biorobot locomotion, achievements in robotic drug delivery, obstacles, and the perspectives of translational research.
Collapse
Affiliation(s)
- Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
- Integrative Institute of Basic Sciences, Soongsil University, Seoul 06978, Republic of Korea
| |
Collapse
|
42
|
Nam Y, Ji YJ, Shin SJ, Park HH, Yeon SH, Kim SY, Son RH, Jang GY, Kim HD, Moon M. Platycodon grandiflorum root extract inhibits Aβ deposition by breaking the vicious circle linking oxidative stress and neuroinflammation in Alzheimer's disease. Biomed Pharmacother 2024; 177:117090. [PMID: 38968796 DOI: 10.1016/j.biopha.2024.117090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease accompanied by irreversible cognitive impairment. A deleterious feedback loop between oxidative stress and neuroinflammation in early AD exacerbates AD-related pathology. Platycodon grandiflorum root extract (PGE) has antioxidant and anti-inflammatory effects in several organs. However, the mechanisms underlying the effects of PGE in the brain remain unclear, particularly regarding its impact on oxidative/inflammatory damage and Aβ deposition. Thus, we aim to identify the mechanism through which PGE inhibits Aβ deposition and oxidative stress in the brain by conducting biochemical and histological analyses. First, to explore the antioxidant mechanism of PGE in the brain, we induced oxidative stress in mice injected with scopolamine and investigated the effect of PGE on cognitive decline and oxidative damage. We also assessed the effect of PGE on reactive oxygen species (ROS) generation and the expressions of antioxidant enzymes and neurotrophic factor in H2O2- and Aβ-treated HT22 hippocampal cells. Next, we investigated whether PGE, which showed antioxidant effects, could reduce Aβ deposition by mitigating neuroinflammation, especially microglial phagocytosis. We directly verified the effect of PGE on microglial phagocytosis, microglial activation markers, and pro-inflammatory cytokines in Aβ-treated BV2 microglial cells. Moreover, we examined the effect of PGE on neuroinflammation, inducing microglial responses in Aβ-overexpressing 5XFAD transgenic mice. PGE exerts antioxidant effects in the brain, enhances microglial phagocytosis of Aβ, and inhibits neuroinflammation and Aβ deposition, ultimately preventing neuronal cell death in AD. Taken together, our findings indicate that the therapeutic potential of PGE in AD is mediated by its targeting of multiple pathological processes.
Collapse
Affiliation(s)
- Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea
| | - Yun-Jeong Ji
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science (NIHHS), Eumsung 27709, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea
| | - Hyun Ha Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea
| | - Sung-Hum Yeon
- Healthcare Research Division, HuonsGlobal Bldg., A-dong Pangyo I-Square, 17, Changeop-ro, Sujeong-gu, Seongnam-si, Gyeonggi-do 13449, Republic of Korea
| | - Sang-Yoon Kim
- Healthcare Research Division, HuonsGlobal Bldg., A-dong Pangyo I-Square, 17, Changeop-ro, Sujeong-gu, Seongnam-si, Gyeonggi-do 13449, Republic of Korea
| | - Rak Ho Son
- Healthcare Research Division, HuonsGlobal Bldg., A-dong Pangyo I-Square, 17, Changeop-ro, Sujeong-gu, Seongnam-si, Gyeonggi-do 13449, Republic of Korea
| | - Gwi Yeong Jang
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science (NIHHS), Eumsung 27709, Republic of Korea
| | - Hyung Don Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science (NIHHS), Eumsung 27709, Republic of Korea; Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea; Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea.
| |
Collapse
|
43
|
Boyd ED, Kaur J, Ding G, Chopp M, Jiang Q. Clinical magnetic resonance imaging evaluation of glymphatic function. NMR IN BIOMEDICINE 2024; 37:e5132. [PMID: 38465514 DOI: 10.1002/nbm.5132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 03/12/2024]
Abstract
The glymphatic system is a system of specialized perivascular spaces in the brain that facilitates removal of toxic waste solutes from the brain. Evaluation of glymphatic system function by means of magnetic resonance imaging (MRI) has thus far been largely focused on rodents because of the limitations of intrathecal delivery of gadolinium-based contrast agents to humans. This review discusses MRI methods that can be employed clinically for glymphatic-related measurements intended for early diagnosis, prevention, and the treatment of various neurological conditions. Although glymphatic system-based MRI research is in its early stages, recent studies have identified promising noninvasive MRI markers associated with glymphatic system alterations in neurological diseases. However, further optimization in data acquisition, validation, and modeling are needed to investigate the glymphatic system within the clinical setting.
Collapse
Affiliation(s)
- Edward D Boyd
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
44
|
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. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.26.24311027. [PMID: 39108503 PMCID: PMC11302722 DOI: 10.1101/2024.07.26.24311027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Blood-brain barrier (BBB) disruption is involved in the pathogenesis and progression of many neurological and systemic diseases. Non-invasive assessment of BBB permeability in humans has mainly been performed with dynamic contrast-enhanced magnetic resonance imaging, evaluating the BBB as a structural barrier. Here, we developed a novel non-invasive positron emission tomography (PET) method in humans to measure the BBB permeability of molecular radiotracers that cross the BBB through different transport mechanisms. Our method uses high-temporal resolution dynamic imaging and kinetic modeling to jointly estimate cerebral blood flow and tracer-specific BBB transport rate from a single dynamic PET scan and measure the molecular permeability-surface area (PS) product of the radiotracer. We show our method can resolve BBB PS across three PET radiotracers with greatly differing permeabilities, measure reductions in BBB PS of 18F-fluorodeoxyglucose (FDG) in healthy aging, and demonstrate a possible brain-body association between decreased FDG BBB PS in patients with metabolic dysfunction-associated steatotic liver inflammation. Our method opens new directions to efficiently study the molecular permeability of the human BBB in vivo using the large catalogue of available molecular PET tracers.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Quyen Tran
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
| | - Souvik Sarkar
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
| | - Valentina Medici
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA
| | - Victoria Lyo
- Department of Surgery, University of California Davis Health, Sacramento, CA
- Center for Alimentary and Metabolic Sciences, University of California Davis Health, Sacramento, CA
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, CA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California at Davis, Davis, CA
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA
| |
Collapse
|
45
|
Yin C, Zhang M, Jin S, Zhou Y, Ding L, Lv Q, Huang Z, Zhou J, Chen J, Wang P, Zhang S, You Q. Mechanism of Salvia miltiorrhiza Bunge extract to alleviate Chronic Sleep Deprivation-Induced cognitive dysfunction in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155725. [PMID: 38772181 DOI: 10.1016/j.phymed.2024.155725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/16/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Bidirectional communication between the gut microbiota and the brain may play an essential role in the cognitive dysfunction associated with chronic sleep deprivation(CSD). Salvia miltiorrhiza Bunge (Danshen, DS), a famous Chinese medicine and functional tea, is extensively used to protect learning and memory capacities, although the mechanism of action remains unknown. PURPOSE The purpose of this research was to explore the efficacy and the underlying mechanism of DS in cognitive dysfunction caused by CSD. METHODS DS chemical composition was analyzed by UPLC-QTOF-MS/MS. Forty rats were randomly assigned to five groups (n = 8): control (CON), model (MOD), low- (1.35 g/kg, DSL), high-dose (2.70 g/kg, DSH) DS group, and Melatonin(100 mg/kg, MT) group. A CSD rat model was established over 21 days. DS's effects and the underlying mechanism were explored using the open-field test(OFT), Morris water-maze(MWM), tissue staining(Hematoxylin and Eosin Staining, Nissl staining, Alcian blue-periodic acid SCHIFF staining, and Immunofluorescence), enzyme-linked immunosorbent assay, Western blot, quantitative real-time polymerase chain reaction(qPCR), and 16S rRNA sequencing. RESULTS We demonstrated that CSD caused gut dysbiosis and cognitive dysfunction. Furthermore, 16S rRNA sequencing demonstrated that Firmicutes and Proteobacteria were more in fecal samples from model group rats, whereas Bacteroidota and Spirochaetota were less. DS therapy, on the contrary hand, greatly restored the gut microbial community, consequently alleviating cognitive impairment in rats. Further research revealed that DS administration reduced systemic inflammation via lowering intestinal inflammation and barrier disruption. Following that, DS therapy reduced Blood Brain Barrier(BBB) and neuronal damage, further decreasing neuroinflammation in the hippocampus(HP). Mechanistic studies revealed that DS therapy lowered lipopolysaccharide (LPS) levels in the HP, serum, and colon, consequently blocking the TLR4/MyD88/NF-κB signaling pathway and its downstream pro-inflammatory products(IL-1β, IL-6, TNF-α, iNOS, and COX2) in the HP and colon. CONCLUSION DS treatment dramatically improved spatial learning and memory impairments in rats with CSD by regulating the composition of the intestinal flora, preserving gut and brain barrier function, and reducing inflammation mediated by the LPS-TLR4 signaling pathway. Our findings provide novel insight into the mechanisms by which DS treats cognitive dysfunction caused by CSD.
Collapse
Affiliation(s)
- Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Meiya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Shuna Jin
- Hubei Shizhen Laboratory, Wuhan 430065, PR China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuan Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Zixuan Huang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jiaqi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Ping Wang
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| | - Shunbo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Qiuyun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| |
Collapse
|
46
|
Chen T, Dai Y, Hu C, Lin Z, Wang S, Yang J, Zeng L, Li S, Li W. Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 2024; 21:60. [PMID: 39030617 PMCID: PMC11264766 DOI: 10.1186/s12987-024-00557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Maintaining the structural and functional integrity of the blood-brain barrier (BBB) is vital for neuronal equilibrium and optimal brain function. Disruptions to BBB performance are implicated in the pathology of neurodegenerative diseases. MAIN BODY Early indicators of multiple neurodegenerative disorders in humans and animal models include impaired BBB stability, regional cerebral blood flow shortfalls, and vascular inflammation associated with BBB dysfunction. Understanding the cellular and molecular mechanisms of BBB dysfunction in brain disorders is crucial for elucidating the sustenance of neural computations under pathological conditions and for developing treatments for these diseases. This paper initially explores the cellular and molecular definition of the BBB, along with the signaling pathways regulating BBB stability, cerebral blood flow, and vascular inflammation. Subsequently, we review current insights into BBB dynamics in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The paper concludes by proposing a unified mechanism whereby BBB dysfunction contributes to neurodegenerative disorders, highlights potential BBB-focused therapeutic strategies and targets, and outlines lessons learned and future research directions. CONCLUSIONS BBB breakdown significantly impacts the development and progression of neurodegenerative diseases, and unraveling the cellular and molecular mechanisms underlying BBB dysfunction is vital to elucidate how neural computations are sustained under pathological conditions and to devise therapeutic approaches.
Collapse
Affiliation(s)
- Tongli Chen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yan Dai
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Chenghao Hu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Zihao Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengzhe Wang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jing Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Shanshan Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Weiyun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
47
|
Hergert DC, Gaasedelen O, Ryman SG, Prestopnik J, Caprihan A, Rosenberg GA. Blood-Brain Barrier Permeability Is Associated With Cognitive Functioning in Normal Aging and Neurodegenerative Diseases. J Am Heart Assoc 2024; 13:e034225. [PMID: 38979810 PMCID: PMC11292768 DOI: 10.1161/jaha.124.034225] [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: 02/27/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The purpose of this study was to investigate the relationship between blood-brain barrier (BBB) permeability and cognitive functioning in healthy older adults and individuals with neurodegenerative diseases. METHODS AND RESULTS A total of 124 participants with Alzheimer disease, cerebrovascular disease, or a mix Alzheimer's and cerebrovascular diseases and 55 controlparticipants underwent magnetic resonance imaging and neuropsychological testing. BBB permeability was measured with dynamic contrast-enhanced magnetic resonance imaging and white matter injury was measured using a quantitative diffusion-tensor imaging marker of white matter injury. Structural equation modeling was used to examine the relationships between BBB permeability, vascular risk burden, white matter injury, and cognitive functioning. Vascular risk burden predicted BBB permeability (r=0.24, P<0.05) and white matter injury (r=0.38, P<0.001). BBB permeability predicted increased white matter injury (r=0.34, P<0.001) and increased white matter injury predicted lower cognitive functioning (r=-0.51, P<0.001). CONCLUSIONS The study provides empirical support for a vascular contribution to white matter injury and cognitive impairment, directly or indirectly via BBB permeability. This highlights the importance of targeting modifiable vascular risk factors to help mitigate future cognitive decline.
Collapse
Affiliation(s)
- Danielle C. Hergert
- US Department of Energy (Contractor), Kirtland Air Force BaseAlbuquerqueNMUSA
| | | | - Sephira G. Ryman
- The Mind Research Network/Lovelace Biomedical Research InstituteAlbuquerqueNMUSA
- Department of NeurologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Jillian Prestopnik
- Center for Memory & AgingUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Arvind Caprihan
- The Mind Research Network/Lovelace Biomedical Research InstituteAlbuquerqueNMUSA
| | - Gary A. Rosenberg
- Center for Memory & AgingUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of NeurologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| |
Collapse
|
48
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
49
|
Vikner T, Garpebring A, Björnfot C, Nyberg L, Malm J, Eklund A, Wåhlin A. Blood-brain barrier integrity is linked to cognitive function, but not to cerebral arterial pulsatility, among elderly. Sci Rep 2024; 14:15338. [PMID: 38961135 PMCID: PMC11222381 DOI: 10.1038/s41598-024-65944-y] [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: 12/09/2023] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Blood-brain barrier (BBB) disruption may contribute to cognitive decline, but questions remain whether this association is more pronounced for certain brain regions, such as the hippocampus, or represents a whole-brain mechanism. Further, whether human BBB leakage is triggered by excessive vascular pulsatility, as suggested by animal studies, remains unknown. In a prospective cohort (N = 50; 68-84 years), we used contrast-enhanced MRI to estimate the permeability-surface area product (PS) and fractional plasma volume ( v p ), and 4D flow MRI to assess cerebral arterial pulsatility. Cognition was assessed by the Montreal Cognitive Assessment (MoCA) score. We hypothesized that high PS would be associated with high arterial pulsatility, and that links to cognition would be specific to hippocampal PS. For 15 brain regions, PS ranged from 0.38 to 0.85 (·10-3 min-1) and v p from 0.79 to 1.78%. Cognition was related to PS (·10-3 min-1) in hippocampus (β = - 2.9; p = 0.006), basal ganglia (β = - 2.3; p = 0.04), white matter (β = - 2.6; p = 0.04), whole-brain (β = - 2.7; p = 0.04) and borderline-related for cortex (β = - 2.7; p = 0.076). Pulsatility was unrelated to PS for all regions (p > 0.19). Our findings suggest PS-cognition links mainly reflect a whole-brain phenomenon with only slightly more pronounced links for the hippocampus, and provide no evidence of excessive pulsatility as a trigger of BBB disruption.
Collapse
Affiliation(s)
- Tomas Vikner
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden.
- Department of Applied Physics and Electronics, Umeå University, 90187, Umeå, Sweden.
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA.
| | - Anders Garpebring
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden
| | - Cecilia Björnfot
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden
| | - Lars Nyberg
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden
- Department of Medical and Translational Biology, Umeå University, 90187, Umeå, Sweden
| | - Jan Malm
- Department of Clinical Science, Neurosciences, Umeå University, 90187, Umeå, Sweden
| | - Anders Eklund
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, 90187, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden
| | - Anders Wåhlin
- Department of Diagnostics and Intervention, Umeå University, 90187, Umeå, Sweden.
- Department of Applied Physics and Electronics, Umeå University, 90187, Umeå, Sweden.
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, 90187, Umeå, Sweden.
| |
Collapse
|
50
|
Taoka T, Ito R, Nakamichi R, Nakane T, Kawai H, Naganawa S. Diffusion Tensor Image Analysis ALong the Perivascular Space (DTI-ALPS): Revisiting the Meaning and Significance of the Method. Magn Reson Med Sci 2024; 23:268-290. [PMID: 38569866 PMCID: PMC11234944 DOI: 10.2463/mrms.rev.2023-0175] [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: 04/05/2024] Open
Abstract
More than 5 years have passed since the Diffusion Tensor Image Analysis ALong the Perivascular Space (DTI-ALPS) method was proposed with the intention of evaluating the glymphatic system. This method is handy due to its noninvasiveness, provision of a simple index in a straightforward formula, and the possibility of retrospective analysis. Therefore, the ALPS method was adopted to evaluate the glymphatic system for many disorders in many studies. The purpose of this review is to look back and discuss the ALPS method at this moment.The ALPS-index was found to be an indicator of a number of conditions related to the glymphatic system. Thus, although this was expected in the original report, the results of the ALPS method are often interpreted as uniquely corresponding to the function of the glymphatic system. However, a number of subsequent studies have pointed out the problems on the data interpretation. As they rightly point out, a higher ALPS-index indicates predominant Brownian motion of water molecules in the radial direction at the lateral ventricular body level, no more and no less. Fortunately, the term "ALPS-index" has become common and is now known as a common term by many researchers. Therefore, the ALPS-index should simply be expressed as high or low, and whether it reflects a glymphatic system is better to be discussed carefully. In other words, when a decreased ALPS-index is observed, it should be expressed as "decreased ALPS-index" and not directly as "glymphatic dysfunction". Recently, various methods have been proposed to evaluate the glymphatic system. It has become clear that these methods also do not seem to reflect the entirety of the extremely complex glymphatic system. This means that it would be desirable to use various methods in combination to evaluate the glymphatic system in a comprehensive manner.
Collapse
Affiliation(s)
- Toshiaki Taoka
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Rintaro Ito
- Department of Innovative Biomedical Visualization (iBMV), Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Rei Nakamichi
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Toshiki Nakane
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Hisashi Kawai
- Department of Radiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|