1
|
Pikus P, Turner RS, Rebeck GW. Mouse models of Anti-Aβ immunotherapies. Mol Neurodegener 2025; 20:57. [PMID: 40361247 PMCID: PMC12076828 DOI: 10.1186/s13024-025-00836-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/05/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The development of anti-amyloid-beta (Aβ) immunotherapies as the first disease modifying therapy for Alzheimer's Disease (AD) is a breakthrough of basic research and translational science. MAIN TEXT Genetically modified mouse models developed to study AD neuropathology and physiology were used for the discovery of Aβ immunotherapies and helped ultimately propel therapies to FDA approval. Nonetheless, the combination of modest efficacy and significant rates of an adverse side effect (amyloid related imaging abnormalities, ARIA), has prompted reverse translational research in these same mouse models to better understand the mechanism of the therapies. CONCLUSION This review considers the use of these mouse models in understanding the mechanisms of Aβ clearance, cerebral amyloid angiopathy (CAA), blood brain barrier breakdown, neuroinflammation, and neuronal dysfunction in response to Aβ immunotherapy.
Collapse
Affiliation(s)
- Philip Pikus
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd, NW, District of Columbia, Washington, 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, 3970 Reservoir Rd, NW, District of Columbia, Washington, 20007, USA
| | - R Scott Turner
- Department of Neurology, Georgetown University Medical Center, 3800 Reservoir Rd, NW, District of Columbia, Washington, 20007, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd, NW, District of Columbia, Washington, 20007, USA.
| |
Collapse
|
2
|
Wang W, Cao C, Pandian VD, Ye H, Chen H, Zhang L. Mac-1 regulates disease stage-specific immunosuppression via the nitric oxide pathway in autoimmune disease. SCIENCE ADVANCES 2025; 11:eads3728. [PMID: 40344054 PMCID: PMC12063669 DOI: 10.1126/sciadv.ads3728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Integrin Mac-1 plays a critical role in the development of multiple sclerosis (MS); however, the underlying mechanism is not fully understood. Here, we developed a myeloid-specific Mac-1-deficient mouse. Using an experimental autoimmune encephalomyelitis (EAE) mouse model of MS, we report that Mac-1 on myeloid cells is key to disease development. Our data reveal that myeloid-specific Mac-1 significantly increases EAE severity and hinders disease regression. Loss of Mac-1 increases Gr-1+ cells in peripheral tissues and the CNS and preferably accelerates the transition of Ly6Chi monocytes from a pro-inflammatory to an immunosuppressive phenotype in a disease stage-dependent manner. Mechanistically, our results demonstrate that Mac-1 suppresses interferon-γ production and prevents monocytes from acquiring immunosuppressive functions by reducing the expression of iNOS, IDO, and CD84. Administration of a NOS-specific inhibitor in Mac-1-deficient EAE mice abolishes disease regression. These insights could help develop Mac-1-targeting strategies for better treatment of MS.
Collapse
MESH Headings
- Animals
- Mice
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Nitric Oxide/metabolism
- Macrophage-1 Antigen/metabolism
- Macrophage-1 Antigen/genetics
- Disease Models, Animal
- Mice, Knockout
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Signal Transduction
- Monocytes/metabolism
- Monocytes/immunology
- Mice, Inbred C57BL
- Immune Tolerance
- Female
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
Collapse
Affiliation(s)
- Wei Wang
- Department of Physiology, Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Chunzhang Cao
- Department of Physiology, Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Vishnuprabu Durairaj Pandian
- Department of Physiology, Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Haofeng Ye
- Johns Hopkins Advanced Academic Programs, Johns Hopkins University of Arts and Sciences, Baltimore, MD, USA
| | - Hongxia Chen
- Department of Physiology, Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Li Zhang
- Department of Physiology, Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| |
Collapse
|
3
|
Casquero-Veiga M, Ceron C, Cortes-Canteli M. Alzheimer's disease and vascular biology - A focus on the procoagulant state. Curr Opin Cell Biol 2025; 95:102528. [PMID: 40347710 DOI: 10.1016/j.ceb.2025.102528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is characterized by a multifactorial pathophysiology. Beyond its classical hallmarks, growing evidence highlights vascular contributions, including hemostatic dysregulation and a prothrombotic state in AD. This review focuses on recent findings concerning two key blood clot components-fibrin(ogen) and platelets-and their roles in AD pathology, including fibrinogen's abnormal accumulation in the AD brain, its interaction with amyloid-β, together with the associated impacts on clot stability, vascular occlusion, and neuroinflammation; and the potential switch of platelets along the AD continuum from protective to deleterious. This review provides an update on the interplay between vascular dysfunction and AD, underscoring the need for comprehensive integrative research to address AD's complexity and advocating for personalized approaches to tackle this multifaceted disorder.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Ceron
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Cortes-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro Internacional de Neurociencia Cajal - Consejo Superior de Investigaciones Científicas (CINC - CSIC), Madrid, Spain.
| |
Collapse
|
4
|
Polykretis P, Bessi V, Banchelli M, de Angelis M, Cecchi C, Nacmias B, Chiti F, D'Andrea C, Matteini P. Fibrillar structures detected by AFM in the cerebrospinal fluid of patients affected by Alzheimer's disease and other neurological conditions. Arch Biochem Biophys 2025; 770:110462. [PMID: 40348257 DOI: 10.1016/j.abb.2025.110462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is a progressive debilitating neurodegenerative disorder that is usually diagnosed after irreversible brain damage has occurred. The detection and morphological characterization of amyloid-β fibrils, which are predominantly implicated in the pathogenic process of the disease, in the cerebrospinal fluid (CSF), emphasized the significance of examining such biofluid. In this work the crude CSF samples collected from patients with Alzheimer's disease and with other neurological conditions were analyzed by atomic force microscopy (AFM). This approach allowed for the identification of peculiar fibrillar structures, exhibiting the regular repetition of dimeric globular units along the longitudinal axis, which share morphological similarities with fibrin protofibrils. Several studies have consistently shown that the disruption of the blood-brain barrier (BBB) and the infiltration of fibrinogen and components of the coagulation cascade are linked with a wide range of neurological diseases, including AD. Therefore, the role of the fibrillar structures we identified must be clarified, in order to ascertain if they contribute to the etiology of the disease, and their use as possible biomarkers of brain damage and disease progression should be assessed.
Collapse
Affiliation(s)
- Panagis Polykretis
- Institute of Applied Physics "Nello Carrara", National Research Council, 50019 Sesto Fiorentino, Italy.
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134 Florence, Italy
| | - Martina Banchelli
- Institute of Applied Physics "Nello Carrara", National Research Council, 50019 Sesto Fiorentino, Italy
| | - Marella de Angelis
- Institute of Applied Physics "Nello Carrara", National Research Council, 50019 Sesto Fiorentino, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3, 50134 Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, 50143 Florence, Italy
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Biochemistry, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Cristiano D'Andrea
- Institute of Applied Physics "Nello Carrara", National Research Council, 50019 Sesto Fiorentino, Italy.
| | - Paolo Matteini
- Institute of Applied Physics "Nello Carrara", National Research Council, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
5
|
Liu XY, Jia HY, Wang G. Predictive value of the fibrinogen-to-albumin ratio for hemorrhagic transformation following intravenous thrombolysis in ischemic stroke: a retrospective propensity score-matched analysis. Front Neurol 2025; 16:1465508. [PMID: 40376152 PMCID: PMC12078177 DOI: 10.3389/fneur.2025.1465508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/18/2025] Open
Abstract
Objective This study aimed to explore the factors associated with hemorrhagic transformation (HT) in acute ischemic stroke patients after intravenous thrombolysis (IVT), with a specific focus on the relationship with the post-thrombolysis fibrinogen-to-albumin ratio (FAR). Methods The clinical records of 569 acute ischemic stroke (AIS) patients admitted to our department from 2020 to 2023 were retrospectively analyzed. All eligible patients were stratified into HT and non-HT (NHT) groups. Propensity score matching (PSM) was performed between the two groups. Receiver operating characteristic (ROC) curves were used to assess the predictive performance of the FAR, determining the optimal predictive value. Results Ultimately, 142 patients were included, with 71 in the HT group and 71 in the NHT group. After propensity score matching, a significant association was observed between the FAR and HT (OR = 1.40, 95% CI, 1.187-1.645; p <0.001). The ROC curve analysis indicated that the FAR predicted HT after intravenous thrombolysis, with an area under the curve (AUC) value of 0.751 (95% CI, 0.669-0.831; p <0.001) and an optimal cutoff value of 0.0918. The corresponding sensitivity and specificity were 78.9 and 60.9%, respectively. Conclusion In ischemic stroke patients undergoing IVT, the FAR may serve as a promising biochemical marker for predicting HT following treatment.
Collapse
Affiliation(s)
- Xiao-Yu Liu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hui-Yang Jia
- Department of Neurology, Panjin Central Hospital, Jinzhou Medical University, Panjin, China
| | - Gang Wang
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
6
|
Yu S, Shi J, Guo S, Guo Z, Xiao G. Stroke severity influences correlation of fibrinogen with early neurological deterioration after thrombolytic therapy. J Stroke Cerebrovasc Dis 2025; 34:108264. [PMID: 39988003 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/25/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND To investigate the relationships between fibrinogen levels at baseline and 24 hours after intravenous thrombolysis (IVT) and early neurological deterioration (END); and to investigate whether stroke severity influences the correlation between fibrinogen and END. METHODS The fibrinogen levels were measured at admission and 24 hours after intravenous thrombolysis (IVT) in 364 consecutive AIS patients. Regression analysis, stratified analyses and interaction tests were utilized to assess the relationship between fibrinogen and END and whether stroke severity (NIHSS<6 vs NIHSS≥6) influences the correlation. RESULTS Fibrinogen at admission was not independently associated with END after adjusting for potential confounders (OR, 1.00; 95 % CI, 0.66-1.53; P = 0.9874). However, increased fibrinogen levels after IVT were associated with increased risk of END (OR, 1.49; 95 % CI, 1.00-2.21; P = 0.0479). Fibrinogen after IVT was not independently associated with END (OR, 1.12; 95 % CI, 0.26-4.80; P = 0.8737) in patients with NIHSS<6, but was independently associated with END in patients with NIHSS≥6 (OR, 1.90; 95 % CI, 1.15-3.15; P = 0.0120). And the interaction test for NIHSS (NIHSS<6 vs NIHSS≥6) was statistically significant (P = 0.0366). Similar results were still found when we used the median NIHSS score of our study population (<10 vs ≥10) as a stratification criterion (Pinteraction = 0.0487). CONCLUSIONS Fibrinogen after IVT but not on admission was independently associated with END. And stroke severity influenced the correlation between fibrinogen after IVT and END.
Collapse
Affiliation(s)
- Shuhong Yu
- Department of Encephalopathy, Suzhou Integrated Traditional Chinese and Western Medicine Hospital, Suzhou 215101, China
| | - Jijun Shi
- Department of Neurology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou 215004, China
| | - Shiping Guo
- Department of Neurology, Qianshan Municipal Hospital, Anqing 246399, China
| | - Zhiliang Guo
- Department of Neurology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou 215004, China; Department of Neurology, Qianshan Municipal Hospital, Anqing 246399, China.
| | - Guodong Xiao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou 215004, China.
| |
Collapse
|
7
|
Jury‐Garfe N, Chimal‐Juárez E, Patel H, Martinez‐Pinto J, Vanderbosch K, Mardones MD, Perkins A, Prisco GVD, Marambio Y, Vidal R, Atwood BK, Lasagna‐Reeves CA. Tau depletion diminishes vascular amyloid-related deficits in a mouse model of cerebral amyloid angiopathy. Alzheimers Dement 2025; 21:e70238. [PMID: 40346706 PMCID: PMC12064415 DOI: 10.1002/alz.70238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Tau is essential for amyloid beta (Aβ)-induced synaptic and cognitive deficits in Alzheimer's disease (AD), making its downregulation a therapeutic target. Cerebral amyloid angiopathy (CAA), a major vascular contributor to cognitive decline, affects over 90% of patients with AD. This study explores the impact of tau downregulation on CAA pathogenesis. METHODS We crossed the Familial Danish Dementia mouse model (Tg-FDD), which develops vascular amyloid, with tau-null (mTau-/-) mice to generate a CAA model lacking endogenous tau (Tg-FDD/mTau-/-). Behavioral, electrophysiological, histological, and transcriptomic analyses were performed. RESULTS Tau depletion ameliorated motor and synaptic impairments, reduced vascular amyloid deposition, and prevented vascular damage. Tau ablation also mitigated astrocytic reactivity and neuroinflammation associated with vascular amyloid accumulation. CONCLUSION These findings provide the first in vivo evidence of the beneficial effects of tau downregulation in a CAA mouse model, supporting tau reduction as a potential therapeutic strategy for patients with parenchymal and vascular amyloid deposition. HIGHLIGHTS Tau ablation improves motor function and synaptic impair, reduces cerebrovascular amyloid deposits, and prevents vascular damage in a mouse model of cerebral amyloid angiopathy (CAA). Tau reduction decreases astrocytic reactivity, alters neuroinflammatory gene expression, and enhances oligodendrocyte function, suggesting a protective role against neuroinflammation in CAA. These findings highlight tau reduction as a potential therapeutic strategy to mitigate CAA-induced pathogenesis, with implications for treating patients with both parenchymal and vascular amyloid deposition.
Collapse
Affiliation(s)
- Nur Jury‐Garfe
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| | - Enrique Chimal‐Juárez
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Henika Patel
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| | - Jonathan Martinez‐Pinto
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Facultad de CienciasInstituto de FisiologíaUniversidad de ValparaísoValparaísoChile
| | - Kathryn Vanderbosch
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Muriel D. Mardones
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Abigail Perkins
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gonzalo Viana Di Prisco
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yamil Marambio
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ruben Vidal
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Brady K. Atwood
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Cristian A. Lasagna‐Reeves
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
8
|
Rust R, Sagare AP, Zhang M, Zlokovic BV, Kisler K. The blood-brain barrier as a treatment target for neurodegenerative disorders. Expert Opin Drug Deliv 2025; 22:673-692. [PMID: 40096820 DOI: 10.1080/17425247.2025.2480654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/14/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a vascular endothelial membrane which restricts entry of toxins, cells, and microorganisms into the brain. At the same time, the BBB supplies the brain with nutrients, key substrates for DNA and RNA synthesis, and regulatory molecules, and removes metabolic waste products from brain to blood. BBB breakdown and/or dysfunction have been shown in neurogenerative disorders including Alzheimer's disease (AD). Current data suggests that these BBB changes may initiate and/or contribute to neuronal, synaptic, and cognitive dysfunction, and possibly other aspects of neurodegenerative processes. AREAS COVERED We first briefly review recent studies uncovering molecular composition of brain microvasculature and examine the BBB as a possible therapeutic target in neurodegenerative disorders with a focus on AD. Current strategies aimed at protecting and/or restoring altered BBB functions are considered. The relevance of BBB-directed approaches to improve neuronal and synaptic function, and to slow progression of neurodegenerative processes are also discussed. Lastly, we review recent advancements in drug delivery across the BBB. EXPERT OPINION BBB breakdown and/or dysfunction can significantly affect neuronal and synaptic function and neurodegenerative processes. More attention should focus on therapeutics to preserve or restore BBB functions when considering treatments of neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mingzi Zhang
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
9
|
Cheemala A, Kimble AL, Burrage EN, Helming SB, Tyburski JD, Leclair NK, Omar OM, Zuberi AR, Murphy M, Jellison ER, Reese B, Hu X, Lutz CM, Yan R, Murphy PA. Amyotrophic lateral sclerosis and frontotemporal dementia mutation reduces endothelial TDP-43 and causes blood-brain barrier defects. SCIENCE ADVANCES 2025; 11:eads0505. [PMID: 40238886 PMCID: PMC12002129 DOI: 10.1126/sciadv.ads0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Mutations in the TARDBP gene encoding TDP-43 protein are linked to loss of function in neurons and familial frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). We recently identified reduced nuclear TDP-43 in capillary endothelial cells (ECs) of donors with ALS-FTD. Because blood-brain barrier (BBB) permeability increases in ALS-FTD, we postulated that reduced nuclear TDP-43 in ECs might contribute. Here, we show that nuclear TDP-43 is reduced in ECs of mice with an ALS-FTD-associated mutation in TDP-43 (TardbpG348C) and that this leads to cell-autonomous loss of junctional complexes and BBB integrity. Targeted excision of TDP-43 in brain ECs recapitulates BBB defects and loss of junctional complexes and ultimately leads to fibrin deposition, gliosis, phospho-Tau accumulation, and impaired memory and social interaction. Transcriptional changes in TDP-43-deficient ECs resemble diseased brain ECs. These data show that nuclear loss of TDP-43 in brain ECs disrupts the BBB and causes hallmarks of FTD.
Collapse
Affiliation(s)
- Ashok Cheemala
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Amy L. Kimble
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Emily N. Burrage
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stephen B. Helming
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Jordan D. Tyburski
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nathan K. Leclair
- MD/PhD Program, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Omar M. Omar
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Aamir R. Zuberi
- Rare Disease Translational Center and Technology Evaluation and Development Laboratory, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Melissa Murphy
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Evan R. Jellison
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Bo Reese
- Center for Genome Innovation, University of Connecticut, Storrs, CT, USA
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Cathleen M. Lutz
- Rare Disease Translational Center and Technology Evaluation and Development Laboratory, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Patrick A. Murphy
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
10
|
Bejarano L, Lourenco J, Kauzlaric A, Lamprou E, Costa CF, Galland S, Maas RR, Guerrero Aruffo P, Fournier N, Brouland JP, Hottinger AF, Daniel RT, Hegi ME, Joyce JA. Single-cell atlas of endothelial and mural cells across primary and metastatic brain tumors. Immunity 2025; 58:1015-1032.e6. [PMID: 40107274 DOI: 10.1016/j.immuni.2025.02.022] [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: 06/19/2024] [Revised: 11/06/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Central nervous system (CNS) malignancies include primary tumors, such as gliomas, and brain metastases (BrMs) originating from diverse extracranial cancers. The blood-brain barrier (BBB) is a key structural component of both primary and metastatic brain cancers. Here, we comprehensively analyzed the two major BBB cell types, endothelial and mural cells, across non-tumor brain tissue, isocitrate dehydrogenase (IDH) mutant (IDH mut) low-grade gliomas, IDH wild-type (IDH WT) high-grade glioblastomas (GBMs), and BrMs from various primary tumors. Bulk and single-cell RNA sequencing, integrated with spatial analyses, revealed that GBMs, but not low-grade gliomas, exhibit significant alterations in the tumor vasculature, including the emergence of diverse pathological vascular cell subtypes. However, these alterations are less pronounced in GBMs than in BrMs. Notably, the BrM vasculature shows higher permeability and more extensive interactions with distinct immune cell populations. This vascular atlas presents a resource toward understanding of tumor-specific vascular features in the brain, providing a foundation for developing vascular- and immune-targeting therapies.
Collapse
Affiliation(s)
- Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | - Joao Lourenco
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Annamaria Kauzlaric
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eleni Lamprou
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Catia F Costa
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sabine Galland
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roeltje R Maas
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Paola Guerrero Aruffo
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Centre, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy T Daniel
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monika E Hegi
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
11
|
Chen Y, Han L, Zhu DS, Guan YT. Fibrinogen and Neuroinflammation in the Neurovascular Unit in Stroke. J Inflamm Res 2025; 18:4567-4584. [PMID: 40191094 PMCID: PMC11971976 DOI: 10.2147/jir.s496433] [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: 09/21/2024] [Accepted: 02/13/2025] [Indexed: 04/09/2025] Open
Abstract
Stroke remains a leading cause of death and disability worldwide. Recent evidence suggests that stroke pathophysiology extends beyond vascular dysfunction to include complex interactions within the neurovascular unit (NVU), particularly involving fibrinogen. This blood-derived protein accumulates in the brain following blood-brain barrier (BBB) disruption and plays crucial roles in neuroinflammation and tissue repair. Through its unique structural domains, fibrinogen interacts with multiple cellular components, including astrocytes, microglia, and neural stem cells, thereby modulating inflammatory responses and neural repair mechanisms. This review examines fibrinogen's structure and its diverse functions in stroke pathophysiology, focusing on its interactions with vascular cells, glial cells, and peripheral immune cells. We also discuss emerging therapeutic strategies targeting fibrinogen-mediated pathways and the challenge of translating experimental results into effective clinical treatments.
Collapse
Affiliation(s)
- Yi Chen
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Lu Han
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - De-Sheng Zhu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Baoshan Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, People’s Republic of China
| | - Yang-Tai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Punan Hospital, Shanghai, 200125, People’s Republic of China
| |
Collapse
|
12
|
Xue X, Zhang Z, Yang Y, Hu L, Zhao M, He Q. Acute exposure of perchlorate on zebrafish larvae: Neurotoxicity during development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118111. [PMID: 40158377 DOI: 10.1016/j.ecoenv.2025.118111] [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: 10/18/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aimed to assess the acute developmental toxicity of perchlorate exposure and identify its manifestation of neurotoxicity in zebrafish (Danio rerio) model. METHODS Zebrafish larvae were exposed to sodium perchlorate at early developmental stages and the phenotypes of various developmental abnormalities were observed and recorded. Neurodevelopmental toxicity has been studied in particular in terms of central nervous system apoptosis, peripheral motor abnormalities, and abnormal autonomic motor behavior. Samples were processed for pathological, transcriptomic, and PCR analyses. RESULTS Acute exposure of zebrafish larvae to perchlorate resulted in developmental toxicity in a concentration-dependent manner. Heart rate abnormality (33 %), slow blood flow (37 %), abnormal size of liver (23 %), delayed yolk absorption (70 %), and abnormal body pigmentation (100 %) were observed in the 3.83 mg/mL group. Heart edema (10 %), heart rate abnormality (60 %), slow blood flow (67 %), abnormal size of liver (77 %), delayed yolk absorption (100 %), abnormalities in intestinal morphology (30 %), abnormal body pigmentation (100 %), and shorter body length (47 %) were observed in the 4.03 mg/mL group. Developmental neurotoxicity was characterized by brain apoptosis, damage to the central nervous system (P < 0.01) and peripheral motor neurons (P < 0.0001), and reduced autonomous motor distance (P < 0.01). Transcriptomic analysis revealed that c3a.1, c5, fga, fgb, and fgg were upregulated in the complement and coagulation cascades, and the mRNA levels of c3a.1, dusp1, slc2a6, purba, and klf9 were found upregulated in PCR. CONCLUSION Acute perchlorate exposure on zebrafish larvae caused various developmental toxicity in a concentration-dependent manner, notably neurotoxicity. We believed the immunological and inflammatory responses were involved.
Collapse
Affiliation(s)
- Xiaoran Xue
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Zaiqiu Zhang
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Yang Yang
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Lin Hu
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
13
|
Nencini F, Giurranna E, Borghi S, Taddei N, Fiorillo C, Becatti M. Fibrinogen Oxidation and Thrombosis: Shaping Structure and Function. Antioxidants (Basel) 2025; 14:390. [PMID: 40298646 PMCID: PMC12024030 DOI: 10.3390/antiox14040390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Fibrinogen, a pivotal plasma glycoprotein, plays an essential role in hemostasis by serving as the precursor to fibrin, which forms the structural framework of blood clots. Beyond coagulation, fibrinogen influences immune responses, inflammation, and tissue repair. Oxidative stress, characterized by an imbalance between reactive oxygen species (ROS) and antioxidants, induces fibrinogen oxidation, significantly altering its structure and function. This narrative review synthesizes findings from in vitro, ex vivo, and clinical studies, emphasizing the impact of fibrinogen oxidation on clot formation, architecture, and degradation. Oxidative modifications result in denser fibrin clots with thinner fibers, reduced permeability, and heightened resistance to fibrinolysis. These structural changes exacerbate prothrombotic conditions in cardiovascular diseases, diabetes, chronic inflammatory disorders and cancer. In contrast, "low-dose" oxidative stress may elicit protective adaptations in fibrinogen, preserving its function. The review also highlights discrepancies in experimental findings due to variability in oxidation protocols and patient conditions. Understanding the interplay between oxidation and fibrinogen function could unveil therapeutic strategies targeting oxidative stress. Antioxidant therapies or selective inhibitors of detrimental oxidation hold potential for mitigating thrombotic risks. However, further research is essential to pinpoint specific fibrinogen oxidation sites, clarify their roles in clot dynamics, and bridge the gap between basic research and clinical practice.
Collapse
|
14
|
Sun B, Li L, Harris OA, Luo J. Blood-brain barrier disruption: a pervasive driver and mechanistic link between traumatic brain injury and Alzheimer's disease. Transl Neurodegener 2025; 14:16. [PMID: 40140960 PMCID: PMC11938631 DOI: 10.1186/s40035-025-00478-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Traumatic brain injury (TBI) has emerged as a significant risk factor for Alzheimer's disease (AD), a complex and devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss. Both conditions share a common feature: blood‒brain barrier (BBB) dysfunction, which is believed to play a pivotal role in linking TBI to the development of AD. This review delves into the intricate relationship between TBI and AD, with a focus on BBB dysfunction and its critical role in disease mechanisms and therapeutic development. We first present recent evidence from epidemiological studies highlighting the increased incidence of AD among individuals with a history of TBI, as well as pathological and animal model studies that demonstrate how TBI can accelerate AD-like pathology. Next, we explore the mechanisms by which BBB dysfunction may mediate TBI-induced AD pathology. Finally, we investigate the shared molecular pathways associated with BBB dysfunction in both TBI and AD conditions and discuss the latest findings on how targeting these pathways and employing regenerative approaches, such as stem cell therapy and pharmacological interventions, can enhance BBB function and mitigate neurodegeneration.
Collapse
Affiliation(s)
- Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Odette A Harris
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
15
|
Yasunaga M, Takata F, Iwao T, Mizoguchi J, Tajima N, Dohgu S. Administration of Noggin Suppresses Fibrinogen Leakage into the Brain in the Acute Phase After Traumatic Brain Injury in Mice. Int J Mol Sci 2025; 26:3002. [PMID: 40243640 PMCID: PMC11988522 DOI: 10.3390/ijms26073002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Traumatic brain injury (TBI) causes neurovascular unit (NVU) dysfunction, including hyperpermeability of the blood-brain barrier to fibrinogen, glial activation, and neuronal damage, possibly leading to secondary brain damage. However, no known substance can inhibit its pathogenesis. In this study, we investigated noggin, a bone morphogenetic protein (BMP) 4 inhibitor, as a TBI pathogenesis-inhibiting substance. We induced acute TBI in C57BL/6J mice through a controlled cortical impact (CCI) and evaluated the effects of noggin on fibrinogen leakage into the brain and NVU-constituting cells, including pericytes, microglia, astrocytes, and neurons. CCI mice showed increased BMP4 levels and extravascular fibrinogen in the hippocampus. Noggin treatment significantly suppressed fibrinogen leakage four days post-CCI in a dose-dependent manner. Immunofluorescence staining revealed that noggin administration did not inhibit the activation of NVU cells such as pericytes, microglia, and astrocytes, which were characterized by increased PDGFRβ, Iba1, and GFAP expression levels, respectively. On postoperative day 4, CCI mice showed neuronal cell and myelinated neuronal fiber loss, which were not significantly affected by noggin administration. In conclusion, noggin administration suppresses fibrinogen leakage into the brain in the acute phase after TBI. However, the suppression of fibrinogen leakage through noggin administration did not alleviate neuronal damage and activation of NVU cells during the acute phase of TBI.
Collapse
Affiliation(s)
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (T.I.); (J.M.); (N.T.); (S.D.)
| | | | | | | | | |
Collapse
|
16
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
17
|
Liu H, Yu R, Zhang M, Zheng X, Zhong L, Yang W, Luo Y, Huang Z, Zheng J, Zhong H, Wei X, Zheng W, Yu Y, Wang Q. Fibrinogen degradation products exacerbate alpha-synuclein aggregation by inhibiting autophagy via downregulation of Beclin1 in multiple system atrophy. Neurotherapeutics 2025; 22:e00538. [PMID: 39904669 PMCID: PMC12014411 DOI: 10.1016/j.neurot.2025.e00538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/24/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disease arising from accumulation of the α-synuclein and aberrant protein clearance in oligodendrocytes. The mechanisms of autophagy involved in the progression of MSA remain poorly understood. It is reported that MSA patients have blood-brain barrier impairments, which may increase the entry of fibrinogen into the brain. However, the roles of fibrinogen and its degradation products (FDPs) on autophagy and α-synuclein accumulation in MSA remain unknown. Here, we established the MSA animal model by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) and 3-nitropropionic acid (3-NP), and cellular models by adding fibrillar α-syn into oligodendrocytes to investigate the mechanisms of FDPs on autophagy and accumulation of α-synuclein in oligodendrocytes. We found that FDPs inhibit the entry of α-synuclein into lysosomes for degradation, increasing aggregation of α-synuclein in oligodendrocytes (OLN-93). Our findings indicated that in OLN-93, FDPs inhibited the expressions of Beclin1 and Bif-1, which could promote the fusion of autophagosomes with lysosomes. Furthermore, the expression of α-synuclein was elevated in FDPs-injected mice, accompanied by an increase in the protein level of p62. We detected elevated expression of FDPs in the striatum of MSA mice. Finally, FDPs inhibited the expression of Beclin1 and Bif-1, which led to aberrant autophagic degradation and increased aggregation of α-synuclein and phospho-α-synuclein in MSA mice. Our study illustrates that FDPs can cause aggregation of α-synuclein in MSA by inhibiting Beclin1-mediated autophagy, which may exacerbate disease progression. These results provide a new therapeutic approach for MSA, that targets the inhibitory effect of FDPs on oligodendrocyte autophagy.
Collapse
Affiliation(s)
- Huanzhu Liu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Ruoyang Yu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Muwei Zhang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Lizi Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hui Zhong
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wenhua Zheng
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, 999078, Macao, China
| | - Yinghua Yu
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, Australia.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
18
|
Cheng X, Sun Y, Wang Y, Cheng W, Zhang H, Jiang Y. The Percentage of Neutrophils is Independently Associated with Blood-Brain Barrier(BBB) Disruption in Myelin Oligodendrocyte Glycoprotein Antibody Associated Disease (MOGAD). J Inflamm Res 2025; 18:2823-2836. [PMID: 40026312 PMCID: PMC11871905 DOI: 10.2147/jir.s501150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose This study aims to investigate the risk factors associated with blood-brain barrier(BBB) disruption in patients with myelin oligodendrocyte glycoprotein antibody associated disease(MOGAD). Patients and Methods We collected clinical data from 95 patients diagnosed with MOGAD at the Department of Neurology, the First Affiliated Hospital of Zhengzhou University from October 2018 to May 2024. Patients were classified into normal or damaged BBB groups based on cerebrospinal fluid (CSF) albumin/serum albumin (QAlb). Binary logistic regression analysis was used to evaluate the risk factors for BBB disruption in MOGAD patients. Results Our study revealed that in MOGAD patients with BBB damaged, there is a higher proportion of acute phase high EDSS scores, higher incidence of prodromal symptoms, and a higher rate of viral infections. Myelitis is the main clinical phenotype, with clinical manifestations primarily including limb weakness and bladder/bowel dysfunction. Laboratory tests showed higher levels of CSF protein, immunoglobulin (IgG), 24-hour intrathecal IgG synthesis rate, peripheral blood leukocytes, neutrophil percentage, NLR, anti-thyroglobulin antibodies(TGAbs), and fibrinogen levels, while free triiodothyronine (FT3) and lymphocyte percentage were lower. Multivariate regression analysis indicated that an increased neutrophil percentage is an independent risk factor for BBB damage in MOGAD patients (OR=1.068, 95% CI: 1.018-1.122, P=0.008). Conclusion Neutrophil percentage is a readily available and widely used indicator reflecting the immune system's state and the body's inflammation level. The change in neutrophil percentage is independently associated with BBB damage in MOGAD patients. This finding helps provide more reference information for personalized treatment decisions and further research into the pathogenesis of MOGAD.
Collapse
Affiliation(s)
- Xuan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yidi Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yaoyao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Wenchao Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Haifeng Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yan Jiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
19
|
Zhou S, Li B, Wu D, Chen Y, Zeng W, Huang J, Tan L, Mao G, Liu F. Mechanisms of fibrinogen trans-activation of the EGFR/Ca2+ signaling axis to regulate mitochondrial transport and energy transfer and inhibit axonal regeneration following cerebral ischemia. J Neuropathol Exp Neurol 2025; 84:210-222. [PMID: 39495964 DOI: 10.1093/jnen/nlae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024] Open
Abstract
Ischemic stroke results in inhibition of axonal regeneration but the roles of fibrinogen (Fg) in neuronal signaling and energy crises in experimental stroke are under-investigated. We explored the mechanism of Fg modulation of axonal regeneration and neuronal energy crisis after cerebral ischemia using a permanent middle cerebral artery occlusion (MCAO) rat model and primary cortical neurons under low glucose-low oxygen. Behavioral tests assessed neurological deficits; immunofluorescence, immunohistochemistry, and Western-blot analyzed Fg and protein levels. Fluo-3/AM fluorescence measured free Ca2+ and ATP levels were gauged via specific assays and F560nm/F510nm ratio calculations. Mito-Tracker Green labeled mitochondria and immunoprecipitation studied protein interactions. Our comprehensive study revealed that Fg inhibited axonal regeneration post-MCAO as indicated by reduced GAP43 expression along with elevated free Ca2+, both suggesting an energy crisis. Fg impeded mitochondrial function and mediated impairment through the EGFR/Ca2+ axis by trans-activating EGFR via integrin αvβ3 interaction. These results indicate that the binding of Fg with integrin αvβ3 leads to the trans-activation of the EGFR/Ca2+ signaling axis thereby disrupting mitochondrial energy transport and axonal regeneration and exacerbating the detrimental effects of ischemic neuronal injury.
Collapse
Affiliation(s)
- Shengqiang Zhou
- National TCM Master Liu Zuyi Inheritance Studio, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Bo Li
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Dahua Wu
- Department of Neurology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Yanjun Chen
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Wen Zeng
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Jia Huang
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Lingjuan Tan
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Guo Mao
- Key Project Office, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Fang Liu
- National TCM Master Liu Zuyi Inheritance Studio, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| |
Collapse
|
20
|
Wen T, Meng L, Liu H, Zhang Q, Dai L, Huang L, Dan L, Zhu K, Luo J, Zhang Z. Fibrinogen-tau Aggregates Exacerbate Tau Pathology and Memory Deficits in Alzheimer's Disease Model Mice. Neurosci Bull 2025:10.1007/s12264-025-01366-8. [PMID: 39971888 DOI: 10.1007/s12264-025-01366-8] [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: 07/08/2024] [Accepted: 10/30/2024] [Indexed: 02/21/2025] Open
Abstract
Vascular damage plays a significant role in the onset and progression of Alzheimer's disease (AD). However, the precise molecular mechanisms underlying the induction of neuronal injury by vascular damage remain unclear. The present study aimed to examine the impact of fibrinogen (Fg) on tau pathology. The results showed that Fg deposits in the brains of tau P301S transgenic mice interact with tau, enhancing the cytotoxicity of pathological tau aggregates and promoting tau phosphorylation and aggregation. Notably, Fg-modified tau fibrils caused enhanced neuronal apoptosis and synaptic damage compared to unmodified fibrils. Furthermore, intrahippocampal injection of Fg-modified tau fibrils worsened the tau pathology, neuroinflammation, synaptic damage, neuronal apoptosis, and cognitive dysfunction in tau P301S mice compared to controls. The present study provides compelling evidence linking Fg and tau, thereby connecting cerebrovascular damage to tau pathology in AD. Consequently, inhibiting Fg-mediated tau pathology could potentially impede the progression of AD.
Collapse
Affiliation(s)
- Tingting Wen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Han Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liqin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liang Dan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kedong Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiaying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
21
|
Sun E, Torices S, Osborne OM, Toborek M. Microvascular Dysfunction, Mitochondrial Reprogramming, and Inflammasome Activation as Critical Regulators of Ischemic Stroke Severity Induced by Chronic Exposure to Prescription Opioids. J Neurosci 2025; 45:e0614242024. [PMID: 39753298 PMCID: PMC11841762 DOI: 10.1523/jneurosci.0614-24.2024] [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: 04/01/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 02/21/2025] Open
Abstract
The opioid epidemic endangers not only public health but also social and economic welfare. Growing clinical evidence indicates that chronic use of prescription opioids may contribute to an elevated risk of ischemic stroke and negatively impact poststroke recovery. In addition, NLRP3 inflammasome activation has been related to several cerebrovascular diseases, including ischemic stroke. Interestingly, an increase in NLRP3 inflammasome activation has also been reported in chronic opioid exposure. Given the pivotal roles of the blood-brain barrier (BBB) and oxidative stress in ischemic stroke pathophysiology, this study focuses on the impact of chronic exposure to prescription opioids on the integrity of cerebrovascular microvasculature, endothelial mitochondrial homeostasis, and the outcomes of ischemic stroke in male wild-type and NLRP3-deficient mice. Our results demonstrate that chronic opioid exposure can compromise the integrity of the BBB and elevate the generation of reactive oxygen species (ROS), resulting in endothelial mitochondrial dysfunction and apoptosis activation. We also provide evidence that opioid exposure enhances inflammasome activation and inflammatory responses and increases the severity of an ischemic stroke. The antioxidant N-acetylcysteine ameliorated these opioid-induced alterations and accelerated the poststroke tissue restoration and functional recovery processes in opioid-exposed mice. Importantly, there was also a significant decrease in ischemic stroke damage in the NLRP3-deficient mice with chronic opioid exposure as compared with wild-type controls. These findings indicate that chronic exposure to prescription opioids impacts the outcome of ischemic stroke by damaging microvascular cerebral integrity through inflammasome activation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Enze Sun
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| |
Collapse
|
22
|
Mahakalakar N, Mohariya G, Taksande B, Kotagale N, Umekar M, Vinchurney M. "Nattokinase as a potential therapeutic agent for preventing blood-brain barrier dysfunction in neurodegenerative disorders". Brain Res 2025; 1849:149352. [PMID: 39592088 DOI: 10.1016/j.brainres.2024.149352] [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: 05/25/2024] [Revised: 10/11/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024]
Abstract
Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis are characterized by progressive destruction of neurons and cognitive impairment, and thorough studies have provided evidence that these pathologies have a close relationship to the failure of the blood-brain barrier (BBB). Nattokinase (NK), a protease found in fermented soybeans, has been extensively studied because it displays powerful neuroprotective abilities, which is why current research was reviewed in the present article. It was concluded that there is enough evidence in preclinical studies using experimental animals that NK supplementation can alleviate the condition related to BBB dysfunction, reduce brain inflammation, and improve cognitive ability. Furthermore, the study of NK on the cardiovascular system leads to certain assumptions, which include the impact on vasculature function and the ability to manage blood flow, which is the key feature of BBB integrity. Such assumed mechanisms are fibrinolytic action, anti-inflammatory and antioxidant action, and endothelium function modulation. There are many positive research findings, and it seems that NK may serve as an effective opponent for BBB breakdown; however, a new research level should be taken to disclose the application and therapeutic use of NK in brain neurodegenerative disease.
Collapse
Affiliation(s)
- Nivedita Mahakalakar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Gunjan Mohariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy (GCOP), Kathora Naka, V.M.V. Road, Amravati (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India
| | - Madhura Vinchurney
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441 002, India.
| |
Collapse
|
23
|
Orian JM. A New Perspective on Mechanisms of Neurodegeneration in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis: the Early and Critical Role of Platelets in Neuro/Axonal Loss. J Neuroimmune Pharmacol 2025; 20:14. [PMID: 39904925 PMCID: PMC11794395 DOI: 10.1007/s11481-025-10182-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: 05/22/2024] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) autoimmune disorder, with limited treatment options. This disease is characterized by differential pathophysiology between grey matter (GM) and white matter (WM). The predominant WM hallmark is the perivascular plaque, associated with blood brain barrier (BBB) loss of function, lymphocytic infiltration, microglial reactivity, demyelination and axonal injury and is adequately addressed with immunomodulatory drugs. By contrast, mechanisms underlying GM damage remain obscure, with consequences for neuroprotective strategies. Cortical GM pathology is already significant in early MS and characterized by reduced BBB disruption and lymphocytic infiltration relative to WM, but a highly inflammatory environment, microglial reactivity, demyelination and neuro/axonal loss. There is no satisfactory explanation for the occurrence of neurodegeneration without large-scale inflammatory cell influx in cortical GM. A candidate mechanism suggests that it results from soluble factors originating from meningeal inflammatory cell aggregates, which diffuse into the underlying cortical tissue and trigger microglial activation. However, the recent literature highlights the central role of platelets in inflammation, together with the relationship between coagulation factors, particularly fibrinogen, and tissue damage in MS. Using the experimental autoimmune encephalomyelitis (EAE) model, we identified platelets as drivers of neuroinflammation and platelet-neuron associations from the pre-symptomatic stage. We propose that fibrinogen leakage across the BBB is a signal for platelet infiltration and that platelets represent a major and early participant in neurodegeneration. This concept is compatible with the new appreciation of platelets as immune cells and of neuronal damage driven by inflammatory cells sequestered in the meninges.
Collapse
Affiliation(s)
- Jacqueline Monique Orian
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Vic. 3086, Australia.
| |
Collapse
|
24
|
Ghelfi L, Mongan D, Susai SR, Föcking M, Cotter DR, Cannon M. Plasma levels of matrix metalloproteinases in early psychosis, anxiety and depression: Evidence from the ALSPAC cohort. Brain Behav Immun 2025; 124:137-143. [PMID: 39615606 DOI: 10.1016/j.bbi.2024.11.035] [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: 08/19/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Converging evidence supports the role of Matrix Metalloproteinases (MMPs) in psychiatric disorders. Originally identified as regulators of the extracellular matrix (ECM), MMPs' functions span multiple processes, including inflammation, synaptic plasticity, neuronal migration, and blood-brain barrier maintenance. Tissue Inhibitors of Metalloproteinases (TIMPs) are major regulators of MMPs. In the present study we examined the associations of plasma MMPs and TIMPs with mental disorders in young adults aged 24 years in the Avon Longitudinal Study of Parents and Children (ALSPAC). METHODS The present study was a nested case control study within the Avon Longitudinal Study of Parents and Children and comprised 374 participants who met criteria for psychiatric disorders (35 met the criteria for psychotic disorder, 201 for mild/moderate depressive disorder, and 266 for generalised anxiety disorder) and 401 controls. All cases and controls had were selected from the group of 4019 participants who had attended at age 24 years, completed psychiatric assessments and provided plasma samples. Plasma concentrations of MMP2, MMP3, MMP9 and TIMP-4 were quantified using proximity extension assays available on Olink® Cardiovascular Panel III. Logistic regression analysis compared standardised MMPs and TIMPs levels in cases and controls. Models were adjusted for sex, body mass index, and cigarette smoking. RESULTS There was evidence for an association between MMP3 and depressive disorder (Odds ratio [OR] 1.35, 95 % confidence interval [CI] 1.06-1.73). There was evidence for an association between TIMP4 and depressive disorder (OR 1.51, 95 % CI 1.22-1.88) and generalised anxiety disorder (OR 1.43, 95 % CI 1.19-1.72). There was no evidence for an association between MMPs and psychotic disorders. CONCLUSIONS The study revealed that 24-year-olds with depressive and anxiety disorders exhibited elevated plasma concentrations of TIMP-4 compared to controls. There was evidence for an association between MMP3 and depressive disorder. These findings provide further support for the involvement of metalloproteinases as biomarkers in the pathophysiology of mental disorders during early adulthood.
Collapse
Affiliation(s)
- Lorenzo Ghelfi
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - David Mongan
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Centre for Public Health, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Subash Raj Susai
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Melanie Föcking
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - David R Cotter
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Mary Cannon
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
25
|
Luo S, Wang Y, Hisatsune T. P2Y1 receptor in Alzheimer's disease. Neural Regen Res 2025; 20:440-453. [PMID: 38819047 PMCID: PMC11317937 DOI: 10.4103/nrr.nrr-d-23-02103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is the most frequent form of dementia characterized by the deposition of amyloid-beta plaques and neurofibrillary tangles consisting of hyperphosphorylated tau. Targeting amyloid-beta plaques has been a primary direction for developing Alzheimer's disease treatments in the last decades. However, existing drugs targeting amyloid-beta plaques have not fully yielded the expected results in the clinic, necessitating the exploration of alternative therapeutic strategies. Increasing evidence unravels that astrocyte morphology and function alter in the brain of Alzheimer's disease patients, with dysregulated astrocytic purinergic receptors, particularly the P2Y1 receptor, all of which constitute the pathophysiology of Alzheimer's disease. These receptors are not only crucial for maintaining normal astrocyte function but are also highly implicated in neuroinflammation in Alzheimer's disease. This review delves into recent insights into the association between P2Y1 receptor and Alzheimer's disease to underscore the potential neuroprotective role of P2Y1 receptor in Alzheimer's disease by mitigating neuroinflammation, thus offering promising avenues for developing drugs for Alzheimer's disease and potentially contributing to the development of more effective treatments.
Collapse
Affiliation(s)
- Shan Luo
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Yifei Wang
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
26
|
Fu M, Liu Y, Hou Z, Wang Z. Interpretable prediction of acute ischemic stroke after hip fracture in patients 65 years and older based on machine learning and SHAP. Arch Gerontol Geriatr 2025; 129:105641. [PMID: 39571498 DOI: 10.1016/j.archger.2024.105641] [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: 05/12/2024] [Revised: 09/01/2024] [Accepted: 09/16/2024] [Indexed: 02/18/2025]
Abstract
BACKGROUND Hip fracture and acute ischemic stroke (AIS) are prevalent conditions among the older population. The prognosis for older patients who experience AIS subsequent to hip fracture is frequently unfavorable. METHODS Patients were categorized into the AIS group and the non-AIS group. A predictive model was developed using six different machine learning algorithms. The SHapley Additive exPlanations (SHAP) method was then utilized to provide both local and global explanations. We performed adjusted mediation analyses. Furthermore, a nomogram was created to present the outcomes obtained from the LASSO regression examination. The main objective was to ascertain influential elements that can predict the occurrence of AIS. To alleviate the influence of confounding variables, propensity score matching was utilized to compare the occurrence of additional complications. Survival was compared by Kaplan-Meier methods. RESULTS The AUC of 6 ML models ranged from 0.73 to 0.87. The SVM model exhibited the greatest efficacy in forecasting AIS among older individuals with hip fractures. The leading 6 variables in the support vector machines (SVM) model were identified as systemic inflammatory response index (SIRI), carotid atherosclerosis, prior stroke, C-reactive protein (CRP), fibrinogen (FIB), and hypertension. The leading 2 variables in SHAP were identified as FIB at admission and SIRI index. There wasn't potential mediating effect of admission FIB between the SIRI index and AIS. There were statistically significant differences between the two groups in survival (P=0.003). CONCLUSIONS The model displayed good performance for prediction of AIS after hip fracture in patients 65 years and older, which might facilitate to establishment of a better clinical assessment plan.
Collapse
Affiliation(s)
- Mingming Fu
- Hebei Medical University Third Hospital, Shijiazhuang, Hebei, PR China
| | - Yan Liu
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, 050051, PR China
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, 050051, PR China; NHC Key Laboratory of Intelligent Orthopedic Equipment (Hebei Medical University Third Hospital), PR China.
| | - Zhiqian Wang
- Department of Geriatric Orthopedics, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
27
|
Noel RL, Kugelman T, Karakatsani ME, Shahriar S, Willner MJ, Choi CS, Nimi Y, Ji R, Agalliu D, Konofagou EE. Safe focused ultrasound-mediated blood-brain barrier opening is driven primarily by transient reorganization of tight junctions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635258. [PMID: 39975117 PMCID: PMC11838333 DOI: 10.1101/2025.01.28.635258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Focused ultrasound (FUS) with microbubbles opens the blood-brain barrier (BBB) to allow targeted drug delivery into the brain. The mechanisms by which endothelial cells (ECs) respond to either low acoustic pressures known to open the BBB transiently, or high acoustic pressures that cause brain damage, remain incompletely characterized. Here, we use a mouse strain where tight junctions between ECs are labelled with eGFP and apply FUS at low (450 kPa) and high (750 kPa) acoustic pressures, after which mice are sacrificed at 1 or 72 hours. We find that the EC response leading to FUS-mediated BBB opening at low pressures is localized primarily in arterioles and capillaries, and characterized by a transient loss and reorganization of tight junctions. BBB opening still occurs at low safe pressures in mice lacking caveolae, suggesting that it is driven primarily by transient dismantlement and reorganization of tight junctions. In contrast, BBB opening at high pressures is associated with obliteration of EC tight junctions that remain unrepaired even after 72 hours, allowing continuous fibrinogen passage and persistent microglial activation. Single-cell RNA-sequencing of arteriole, capillary and venule ECs from FUS mice reveals that the transcriptomic responses of ECs exposed to high pressure are dominated by genes belonging to the stress response and cell junction disassembly at both 1 and 72 hours, while lower pressures induce primarily genes responsible for intracellular repair responses in ECs. Our findings suggest that at low pressures transient reorganization of tight junctions and repair responses mediate safe BBB opening for therapeutic delivery. Significance Statement Focused ultrasound with microbubbles is used as a noninvasive method to safely open the BBB at low acoustic pressures for therapeutic delivery into the CNS, but the mechanisms mediating this process remain unclear. Kugelman et al., demonstrate that FUS-mediated BBB opening at low pressures occurs primarily in arterioles and capillaries due to transient reorganization of tight junctions. BBB opening still occurs at low safe pressures in mice lacking caveolae, suggesting a transcellular route-independent mechanism. At high unsafe pressures, cell junctions are obliterated and remain unrepaired even after 72 hours, allowing fibrinogen passage and persistent microglial activation. Single-cell RNA-sequencing supports cell biological findings that safe, FUS-mediated BBB opening may be driven by transient reorganization and repair of EC tight junctions.
Collapse
|
28
|
Nehra G, Maloney BJ, Smith RR, Chumboatong W, Abner EL, Nelson PT, Bauer B, Hartz AMS. Plasma S100β is a predictor for pathology and cognitive decline in Alzheimer's disease. Fluids Barriers CNS 2025; 22:4. [PMID: 39789614 PMCID: PMC11720585 DOI: 10.1186/s12987-024-00615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Blood-brain barrier dysfunction is one characteristic of Alzheimer's disease (AD) and is recognized as both a cause and consequence of the pathological cascade leading to cognitive decline. The goal of this study was to assess markers for barrier dysfunction in postmortem tissue samples from research participants who were either cognitively normal individuals (CNI) or diagnosed with AD at the time of autopsy and determine to what extent these markers are associated with AD neuropathologic changes (ADNC) and cognitive impairment. METHODS We used postmortem brain tissue and plasma samples from 19 participants: 9 CNI and 10 AD dementia patients who had come to autopsy from the University of Kentucky AD Research Center (UK-ADRC) community-based cohort; all cases with dementia had confirmed severe ADNC. Plasma samples were obtained within 2 years of autopsy. Aβ40, Aβ42, and tau levels in brain tissue samples were quantified by ELISA. Cortical brain sections were cleared using the X-CLARITY™ system and immunostained for neurovascular unit-related proteins. Brain slices were then imaged using confocal microscopy and analyzed for microvascular diameters and immunoreactivity coverage using Fiji/ImageJ. Isolated human brain microvessels were assayed for tight-junction protein expression using the JESS™ automated Western blot system. S100 calcium-binding protein B (S100β), matrix metalloproteinase (MMP)-2, MMP-9, and neuron-specific enolase (NSE) levels in plasma were quantified by ELISA. All outcomes were assessed for linear associations with global cognitive function (MMSE, CDR) and cerebral atrophy scores by Pearson, polyserial, or polychoric correlation, as appropriate, along with generalized linear modeling or generalized linear mixed-level modeling. RESULTS As expected, we detected elevated Aβ and tau pathology in brain tissue sections from AD patients compared to CNI. However, we found no differences in microvascular diameters in cleared AD and CNI brain tissue sections. We also observed no differences in claudin-5 protein levels in capillaries isolated from AD and CNI tissue samples. Plasma biomarker analysis showed that AD patients had 12.4-fold higher S100β plasma levels, twofold lower NSE plasma levels, 2.4-fold higher MMP-9 plasma levels, and 1.2-fold lower MMP-2 plasma levels than CNI. Data analysis revealed that elevated S100β plasma levels were predictive of AD pathology and cognitive impairment. CONCLUSION Our data suggest that among different markers relevant to barrier dysfunction, plasma S100β is the most promising diagnostic biomarker for ADNC. Further investigation is necessary to assess how plasma S100β levels relate to these changes and whether they may predict clinical outcomes, particularly in the prodromal and early stages of AD.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Bryan J Maloney
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Rebecca R Smith
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Wijitra Chumboatong
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, USA
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Pathology, College of Medicine, University of Kentucky, Lexington, USA
| | - Björn Bauer
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, 760 Press Ave, 124 HKRB, Lexington, KY, 40536-0679, USA.
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, USA.
| |
Collapse
|
29
|
Wei C, Zhai W, Zhao P, Sun L. Plasma fibrinogen as a potential biomarker of cognitive impairment after acute ischemic stroke. Sci Rep 2024; 14:32120. [PMID: 39739003 PMCID: PMC11685652 DOI: 10.1038/s41598-024-83907-1] [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: 09/17/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
Fibrinogen (FBG) has been discovered to be associated with cognitive impairment (CI) and dementia. However, the exact correlation between FBG levels and CI after acute ischemic stroke (AIS) remains uncertain. Plasma FBG levels were measured in 398 patients with AIS who underwent comprehensive neuropsychological evaluation. To assess the correlation of FBG with global cognitive function, physical status, anxiety, depression, and psychiatric symptoms. Multifactorial logistic regression was used to analyze risk factors for CI. Constructed and plotted a nomogram graph to visualize the CI prediction model. The model was further evaluated for discrimination, calibration, and clinical utility. The results indicate that plasma FBG levels are significantly elevated in patients with CI compared to those with non-cognitive impairment (NCI). Analysis of the overall population reveals that elevated FBG levels are correlated with both reduced cognitive function and decreased activity status. After adjusting for other influencing factors, high FBG levels were identified as a risk factor for the incidence of CI. We developed an intuitive and valid predictive model for CI, demonstrating its suitability for clinical application. In conclusion, our study demonstrates that plasma FBG serves as a potential biomarker of CI following AIS, offering a novel perspective for the identification of CI.
Collapse
Affiliation(s)
- Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Weijie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China.
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
30
|
Patel S, Govindarajan V, Chakravarty S, Dubey N. From blood to brain: Exploring the role of fibrinogen in the pathophysiology of depression and other neurological disorders. Int Immunopharmacol 2024; 143:113326. [PMID: 39388892 DOI: 10.1016/j.intimp.2024.113326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Recent findings indicate that fibrinogen, a protein involved in blood clotting, plays a significant role in neuroinflammation and mood disorders. Elevated fibrinogen levels are consistently observed in individuals with depression, potentially contributing to microglial activation. This could impair fibrinolysis and contribute to a pro-inflammatory environment in the brain. This neuroinflammatory response can impair neuroplasticity, a key process for learning, memory, and mood regulation. Fibrinogen may also indirectly influence neurotransmitters like serotonin, which play a vital role in mood regulation. Furthermore, fibrinogen's interaction with astrocytes may trigger a cascade of events leading to demyelination, a process where the protective sheath around nerve fibers deteriorates. This can disrupt communication within the nervous system and contribute to depression symptoms. Intriguingly, targeting fibrinogen or related pathways holds promise for therapeutic interventions. For instance, modulating PAI-1 (Plasminogen activator inhibitor-1) activity or inhibiting fibrinogen's interaction with brain cells could be potential strategies. This review explores the multifaceted relationship between fibrinogen and neurological disorders with a focus on depression highlighting its potential as a therapeutic target. Further research is necessary to fully elucidate the mechanisms underlying this association and develop effective therapeutic strategies targeting the fibrinolytic system for mood disorders.
Collapse
Affiliation(s)
- Shashikant Patel
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, India
| | - Venkatesh Govindarajan
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, Telangana, India
| | - Sumana Chakravarty
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, India.
| | - Neelima Dubey
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad 500046, Telangana, India.
| |
Collapse
|
31
|
Hartmannsberger B, Ben-Kraiem A, Kramer S, Guidolin C, Kazerani I, Doppler K, Thomas D, Gurke R, Sisignano M, Kalelkar PP, García AJ, Monje PV, Sammeth M, Nusrat A, Brack A, Krug SM, Sommer C, Rittner HL. TAM receptors mediate the Fpr2-driven pain resolution and fibrinolysis after nerve injury. Acta Neuropathol 2024; 149:1. [PMID: 39680199 DOI: 10.1007/s00401-024-02840-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Nerve injury causes neuropathic pain and multilevel nerve barrier disruption. Nerve barriers consist of perineurial, endothelial and myelin barriers. So far, it is unclear whether resealing nerve barriers fosters pain resolution and recovery. To this end, we analysed the nerve barrier property portfolio, pain behaviour battery and lipidomics for precursors of specialized pro-resolving meditators (SPMs) and their receptors in chronic constriction injury of the rat sciatic nerve to identify targets for pain resolution by resealing the selected nerve barriers. Of the three nerve barriers-perineurium, capillaries and myelin-only capillary tightness specifically against larger molecules, such as fibrinogen, recuperated with pain resolution. Fibrinogen immunoreactivity was elevated in rats not only at the time of neuropathic pain but also in nerve biopsies from patients with (but not without) painful polyneuropathy, indicating that sealing of the vascular barrier might be a novel approach in pain treatment. Hydroxyeicosatetraenoic acid (15R-HETE), a precursor of aspirin-triggered lipoxin A4, was specifically upregulated at the beginning of pain resolution. Repeated local application of resolvin D1-laden nanoparticles or Fpr2 agonists sex-independently resulted in accelerated pain resolution and fibrinogen removal. Clearing macrophages (Cd206) were boosted and fibrinolytic pathways (Plat) were induced, while inflammation (Tnfα) and inflammasomes (Nlrp3) were unaffected by this treatment. Blocking TAM receptors (Tyro3, Axl and Mer) and tyrosine kinase receptors linking haemostasis and inflammation completely inhibited all the effects. In summary, nanoparticles can be used as transporters for fleeting lipids, such as SPMs, and therefore expand the array of possible therapeutic agents. Thus, the Fpr2-Cd206-TAM receptor axis may be a suitable target for strengthening the capillary barrier, removing endoneurial fibrinogen and boosting pain resolution in patients with chronic neuropathic pain.
Collapse
Affiliation(s)
- Beate Hartmannsberger
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Adel Ben-Kraiem
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research, Diet-Induced Metabolic Alterations Group, Leipzig, Germany
| | - Sofia Kramer
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Carolina Guidolin
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ida Kazerani
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Dominique Thomas
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Robert Gurke
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Marco Sisignano
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Pranav P Kalelkar
- George W. Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, USA
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, USA
| | - Paula V Monje
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Michael Sammeth
- Department of Applied Sciences and Health, Coburg University of Applied Sciences and Art, Coburg, Germany
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Brack
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Susanne M Krug
- Charité-Universitätsmedizin Berlin, Clinical Physiology/Nutritional Medicine, Berlin, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Heike L Rittner
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
32
|
Gabdulkhaev R, Shimizu H, Kanazawa M, Kuroha Y, Hasegawa A, Idezuka J, Tainaka K, Onodera O, Kakita A. Blood-brain barrier dysfunction in multiple system atrophy: A human postmortem study. Neuropathology 2024. [PMID: 39665496 DOI: 10.1111/neup.13021] [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: 03/18/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by an accumulation of phosphorylated α-synuclein (p-αsyn) in oligodendrocytes in the form of glial cytoplasmic inclusions (GCIs). In MSA, not only mature oligodendrocytes but also oligodendrocyte precursor cells (OPCs) are affected. The latter play an important role in remyelination by differentiating into mature oligodendrocytes, as well as maintaining the blood-brain barrier (BBB) by promoting the expression of tight junction proteins. We have hypothesized that in MSA, the BBB is impaired as a result of aberrant interactions between affected OPCs and the cerebral vasculature. To verify this hypothesis, we conducted a neuropathological examination of postmortem brains from MSA patients and control subjects, focusing on the primary motor area, one of the main regions affected in MSA. Using double immunofluorescence, we quantified the expression of tight junction protein claudin-5 in capillary endothelial cells and found that it was significantly lower in MSA than in controls in both the gray matter and white matter. Furthermore, a significantly higher amount of fibrinogen was extravasated into the brain parenchyma in MSA patients than in controls. In addition, leakage of IgG was detected almost specifically in MSA brain parenchyma, as visualized in three dimensions by combining techniques of chemical tissue clearing and light sheet microscopy. Finally, we confirmed accumulation of p-αsyn-positive GCIs along the cerebral vasculature within OPCs. These results suggest that BBB dysfunction and associated fibrinogen extravasation are constant findings in MSA, presumably triggered by the deposition of p-αsyn in perivascular OPCs.
Collapse
Affiliation(s)
- Ramil Gabdulkhaev
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroshi Shimizu
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasuko Kuroha
- Department of Neurology, NHO Nishiniigata Chuo Hospital, Niigata, Japan
| | - Arika Hasegawa
- Department of Neurology, NHO Nishiniigata Chuo Hospital, Niigata, Japan
| | - Jiro Idezuka
- Department of Neurology, Ojiya Sakura Hospital, Niigata, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
33
|
Payne T, Taylor J, Kunkel D, Konieczka K, Ingram F, Blennow K, Zetterberg H, Pearce RA, Meyer-Franke A, Terrando N, Akassoglou K, Sanders RD, Lennertz RC. Association of preoperative to postoperative change in cerebrospinal fluid fibrinogen with postoperative delirium. BJA OPEN 2024; 12:100349. [PMID: 39429436 PMCID: PMC11490679 DOI: 10.1016/j.bjao.2024.100349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/20/2024] [Indexed: 10/22/2024]
Abstract
Background We aimed to assess perioperative changes in fibrinogen in the cerebrospinal fluid (CSF), their association with markers of blood-brain barrier breakdown and neuroinflammation, and their association with postoperative delirium severity. Methods We conducted a secondary analysis of the Interventions for Postoperative Delirium-Biomarker 2 (IPOD-B2, NCT02926417) study, a prospective observational cohort study. We included 24 patients aged >21 yr undergoing aortic aneurysm repair. CSF samples were obtained before (n=24) and after surgery (n=13), with some participants having multiple postoperative samples. Our primary outcome was the perioperative change in CSF fibrinogen. Delirium was assessed using the Delirium Rating Scale-Revised-98. Results CSF fibrinogen increased after surgery (P<0.001), and this was associated with an increase in CSF/plasma albumin ratio (β=1.09, 95% CI 0.47-1.71, P=0.004). The peak change in CSF fibrinogen was associated with the change in CSF interleukin (IL)-10 and IL-12p70. The peak change in CSF fibrinogen was associated with the change in CSF total tau (β=0.47, 95% CI 0.24-0.71, P=0.002); however, we did not observe an association with postoperative delirium severity (incidence rate ratio = 1.20, 95% CI 0.66-2.17, P=0.540). Conclusions Our preliminary findings support the hypothesis that fibrinogen enters the brain via blood-brain barrier disruption, promoting neuroinflammation and neuronal injury. However, we did not observe an association between cerebrospinal fluid fibrinogen and peak delirium severity in this limited cohort.
Collapse
Affiliation(s)
- Thomas Payne
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Anaesthetics, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Jennifer Taylor
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Anaesthetics, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, New South Wales, Australia
| | - David Kunkel
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Katherine Konieczka
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Frankie Ingram
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Robert A. Pearce
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Anke Meyer-Franke
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Cell Biology, and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Katerina Akassoglou
- Gladstone UCSF Center for Neurovascular Brain Immunology, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology and Weill Institute of Neuroscience, University of California San Francisco, San Francisco, CA, USA
| | - Robert D. Sanders
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Anaesthetics, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, New South Wales, Australia
- Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, New South Wales, Australia
- NHMRC Clinical Trials Centre, The University of Sydney, New South Wales, Australia
| | - Richard C. Lennertz
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
34
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
35
|
Sandeep, Subba R, Mondal AC. Does COVID-19 Trigger the Risk for the Development of Parkinson's Disease? Therapeutic Potential of Vitamin C. Mol Neurobiol 2024; 61:9945-9960. [PMID: 37957424 DOI: 10.1007/s12035-023-03756-3] [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: 02/20/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which was proclaimed a pandemic by the World Health Organization (WHO) in March 2020. There is mounting evidence that older patients with multimorbidity are more susceptible to COVID-19 complications than are younger, healthy people. Having neuroinvasive potential, SARS-CoV-2 infection may increase susceptibility toward the development of Parkinson's disease (PD), a progressive neurodegenerative disorder with extensive motor deficits. PD is characterized by the aggregation of α-synuclein in the form of Lewy bodies and the loss of dopaminergic neurons in the dorsal striatum and substantia nigra pars compacta (SNpc) of the nigrostriatal pathway in the brain. Increasing reports suggest that SARS-CoV-2 infection is linked with the worsening of motor and non-motor symptoms with high rates of hospitalization and mortality in PD patients. Common pathological changes in both diseases involve oxidative stress, mitochondrial dysfunction, neuroinflammation, and neurodegeneration. COVID-19 exacerbates the damage ensuing from the dysregulation of those processes, furthering neurological complications, and increasing the severity of PD symptomatology. Phytochemicals have antioxidant, anti-inflammatory, and anti-apoptotic properties. Vitamin C supplementation is found to ameliorate the common pathological changes in both diseases to some extent. This review aims to present the available evidence on the association between COVID-19 and PD, and discusses the therapeutic potential of vitamin C for its better management.
Collapse
Affiliation(s)
- Sandeep
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rhea Subba
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
36
|
Torres-Espin A, Radabaugh HL, Treiman S, Fitzsimons SS, Harvey D, Chou A, Lindbergh CA, Casaletto KB, Goldberger L, Staffaroni AM, Maillard P, Miller BL, DeCarli C, Hinman JD, Ferguson AR, Kramer JH, Elahi FM. Sexually dimorphic differences in angiogenesis markers are associated with brain aging trajectories in humans. Sci Transl Med 2024; 16:eadk3118. [PMID: 39602511 DOI: 10.1126/scitranslmed.adk3118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Aberrant angiogenesis could contribute to the development of cognitive impairment and represent a therapeutic target for preventing dementia. However, most studies addressing angiogenesis and cognitive impairment focus on model organisms. To test the relevance of angiogenesis to human cognitive aging, we evaluated associations of circulating blood markers of angiogenesis with brain aging trajectories in a pooled two-center sample from deeply phenotyped longitudinal human cohorts (n = 435; female = 207, age = 74 ± 9) using cognitive assessments, biospecimens, structural brain imaging, and clinical data. Blood markers included ligands involved in angiogenesis and vascular function such as basic fibroblast growth factor (bFGF), members of the vascular endothelial growth factor family (VEGFA, VEGFB, and VEGFC), and placental growth factor (PlGF), in addition to their receptors VEGF receptor 1 (VEGFR1) and tyrosine kinase with immunoglobulin and EGF homology domain 2 (Tie2). Machine learning and traditional statistics revealed sexually dimorphic associations of plasma angiogenic growth factors with brain aging outcomes, including executive function and gray matter atrophy. Specifically, markers of angiogenesis were associated with higher executive function and less brain atrophy in younger women (not men), a directionality of association that reversed around age 75. Higher concentrations of bFGF, known for pleiotropic effects on multiple cell types, predicted favorable cognitive trajectories in both women and men. An independent sample from a multicenter dataset (MarkVCID; n = 80; female = 30, age = 73 ± 9) was used to externally validate these findings. In conclusion, this analysis demonstrates the association of angiogenesis to human brain aging, with potential therapeutic implications for vascular cognitive impairment and dementia.
Collapse
Affiliation(s)
- Abel Torres-Espin
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hannah L Radabaugh
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Scott Treiman
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephen S Fitzsimons
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Danielle Harvey
- Department of Public Health Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Austin Chou
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cutter A Lindbergh
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lauren Goldberger
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam M Staffaroni
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA 95817, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA 95817, USA
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Fanny M Elahi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- James J. Peters Veterans Affairs Health Care System, Bronx, NY 10468, USA
| |
Collapse
|
37
|
Miller SR, Luxem K, Lauderdale K, Nambiar P, Honma PS, Ly KK, Bangera S, Bullock M, Shin J, Kaliss N, Qiu Y, Cai C, Shen K, Mallen KD, Yan Z, Mendiola AS, Saito T, Saido TC, Pico AR, Thomas R, Roberson ED, Akassoglou K, Bauer P, Remy S, Palop JJ. Machine learning reveals prominent spontaneous behavioral changes and treatment efficacy in humanized and transgenic Alzheimer's disease models. Cell Rep 2024; 43:114870. [PMID: 39427315 PMCID: PMC12010505 DOI: 10.1016/j.celrep.2024.114870] [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: 10/09/2023] [Revised: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Computer-vision and machine-learning (ML) approaches are being developed to provide scalable, unbiased, and sensitive methods to assess mouse behavior. Here, we used the ML-based variational animal motion embedding (VAME) segmentation platform to assess spontaneous behavior in humanized App knockin and transgenic APP models of Alzheimer's disease (AD) and to test the role of AD-related neuroinflammation in these behavioral manifestations. We found marked alterations in spontaneous behavior in AppNL-G-F and 5xFAD mice, including age-dependent changes in motif utilization, disorganized behavioral sequences, increased transitions, and randomness. Notably, blocking fibrinogen-microglia interactions in 5xFAD-Fggγ390-396A mice largely prevented spontaneous behavioral alterations, indicating a key role for neuroinflammation. Thus, AD-related spontaneous behavioral alterations are prominent in knockin and transgenic models and sensitive to therapeutic interventions. VAME outcomes had higher specificity and sensitivity than conventional behavioral outcomes. We conclude that spontaneous behavior effectively captures age- and sex-dependent disease manifestations and treatment efficacy in AD models.
Collapse
Affiliation(s)
- Stephanie R Miller
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Kevin Luxem
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Kelli Lauderdale
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pranav Nambiar
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Patrick S Honma
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Katie K Ly
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shreya Bangera
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mary Bullock
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jia Shin
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nick Kaliss
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Yuechen Qiu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Catherine Cai
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Kevin Shen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - K Dakota Mallen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Zhaoqi Yan
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Andrew S Mendiola
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi 351-0198, Japan
| | - Alexander R Pico
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA 94158, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA 94158, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA
| | - Pavol Bauer
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Stefan Remy
- German Center for Neurodegenerative Diseases (DZNE), 39118 Bonn and Magdeburg, Germany; Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany; German Center for Mental Health (DZPG), 39118 Magdeburg, Germany
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
38
|
Albashir D, Lu H, Gouda M, Acharya DR, Danhassan UA, Bakur A, Shi Y, Chen Q. A novel polydiacetylene-functionalized fibrinogen paper-based biosensor for on-spot and rapid detection of Staphylococcus aureus. Food Chem 2024; 458:140291. [PMID: 38959795 DOI: 10.1016/j.foodchem.2024.140291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/04/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
Staphylococcus aureus contamination continues to be a harmful foodborne pathogen threatening of human health, and there is a growing need for rapid detection technologies. This study proposed a novel paper biosensor based on a polydiacetylene (PDA) polymer functionalized fibrinogen (Fg) for the detection of S. aureus in food sources. The fluorophore was developed based on the high binding ability of fibrinogen-binding proteins on the surface of S. aureus. This binding caused twisting in the PDA backbone, leading to changes in chromatic and fluorescent. The detection limit of this method was 50.1 CFU/mL for S. aureus-contaminated foodstuffs and 65.0 CFU/mL for the pure S. aureus culture, and the novelty came from its rapidity and selectivity for S. aureus compared to other foodborne bacteria. In summary, the present work provides a rapid detection method for S. aureus detection, which will help in addressing food safety-related issues.
Collapse
Affiliation(s)
- Dafaallah Albashir
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Hongyun Lu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Mostafa Gouda
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Department of Nutrition and Food Science, National Research Centre, Dokki, Giza 12622, Egypt
| | - Dev Raj Acharya
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | | | - Abdelmoneim Bakur
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Department of Food Sciences and Technology, University of Kordofan, El Obeid, Sudan
| | - Ying Shi
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Qihe Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
39
|
Yekani M, Dizaj SM, Sharifi S, Sedaghat H, Saffari M, Memar MY. Nano-scaffold-based delivery systems of antimicrobial agents in the treatment of osteomyelitis ; a narrative review. Heliyon 2024; 10:e38392. [PMID: 39559197 PMCID: PMC11570522 DOI: 10.1016/j.heliyon.2024.e38392] [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: 02/02/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Osteomyelitis caused by drug-resistant pathogens is one of the most important medical challenges due to high rates of mortality and morbidity, and limited therapeutical options. The application of novel nano-scaffolds loaded with antibiotics has widely been studied and extensively evaluated for in vitro and in vivo inhibition of pathogens, regenerating damaged bone tissue, and increasing bone cell proliferation. The treatment of bone infections using the local osteogenic scaffolds loaded with antimicrobial agents may efficiently overcome the problems of the systemic use of antimicrobial agents and provide a controlled release and sufficient local levels of antibiotics in the infected sites. The present study reviewed various nano-scaffolds delivery systems of antimicrobial drugs evaluated to treat osteomyelitis. Nano-scaffolds offer promising approaches because they simulate natural tissue regeneration in terms of their mechanical, structural, and sometimes chemical properties. The potential of several nano-scaffolds prepared by natural polymers such as silk, collagen, gelatin, fibrinogen, chitosan, cellulose, hyaluronic, alginate, and synthetic compounds such as polylactic acid, polyglycolic acid, poly (lactic acid-co-glycolic acid), poly-ɛ-caprolactone have been studied for usage as drug delivery systems of antimicrobial agents to treat osteomyelitis. In addition to incorporated antimicrobial agents and the content of scaffolds, the physical and chemical characteristics of the prepared delivery systems are a determining factor in their effectiveness in treating osteomyelitis.
Collapse
Affiliation(s)
- Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Sedaghat
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmood Saffari
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Lu D, Qu C, Fang M, Zhang J. Exercise rescues cognitive impairment through inhibiting the fibrinogen neuroinflammative pathway in diabetes. Metab Brain Dis 2024; 40:2. [PMID: 39535634 DOI: 10.1007/s11011-024-01455-z] [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: 04/24/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Fibrinogen is a pivotal factor in the activation of neuroinflammation and cognitive impairment. While exercise, especifically swimming, has demonstrated cognitive benefits, the molecular protective mechanisms orchestrated by exercise in response to blood-brain barrier (BBB) leakage in diabetes remain elusive. This study systematically investigates the impact of fibrinogen on neuroinflammation and the role of exercise in diabetic rats. Diabetic rats underwent an 8-week swimming exercise regimen, and subsequent assessments included changes in interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α), astroglia activation, BBB permeability, and key epithelial tight junction proteins such as zona occludins (ZO)-1, Claudin-5, and matrix metalloproteinase-9 (MMP-9). Spatial learning and memory were evaluated using the Morris water maze test and the novel object recognition test. The study revealed that exercise significantly improved cognitive function, potentially by suppressing fibrinogen levels and astroglia activation. Intriguingly, heightened fibrinogen expression markedly attenuated the protective effects of exercise on BBB integrity. Fibrinogen emerged as a potential compromise to exercise protective effect by increasing expression levels of inflammatory factors IL-1β and TNF-α. In summary, our findings elucidate that fibrinogen may contribute to the deterioration of cognition and diminish the protective effects of exercise by amplifying the neuroinflammatory process through damaged BBB in diabetes.
Collapse
Affiliation(s)
- Dongwei Lu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chujie Qu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
- West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mei Fang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
41
|
Huan S, Zhang S, Wang L, Liu C, Xu J, Ge Y, Huang Z, Cai W, Cao Y, You S. Association between fibrinogen and white matter lesions and cerebral atrophy in patients with acute ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:108008. [PMID: 39265859 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Inflammation is a potential mechanism underlying the development of white matter lesions (WMLs) and cerebral atrophy. We aimed to investigate the relationship of fibrinogen levels with WMLs and cerebral atrophy in patients with acute ischemic stroke (AIS). METHODS A total of 701 AIS patients were enrolled. Participants were divided into four groups according to the quartiles of fibrinogen levels: Q1 < 2.58 g/L, Q2: 2.58-3.12 g/L, Q3: 3.12-3.67 g/L, Q4: ≥ 3.67 g/L. White matter hyperintensity (WMH), periventricular hyperintensity (PVH) and deep white matter hyperintensity (DWMH) were defined according to the Fazekas scale. Cerebral atrophy was defined according to global cortical atrophy scores. Univariate and multivariate logistic regression were used to explore the relationship of fibrinogen levels and WMHs, PVH, DWMH and cerebral atrophy. RESULTS Among 701 AIS patients, 498 (71.0 %), 425 (60.6 %), 442 (63.1 %), and 560 (79.9 %) had WMHs, PVH, DWMH and cerebral atrophy, respectively. After adjustment for potential covariates, the highest fibrinogen quartiles were significantly associated with increased risk of WMHs (odds ratio [OR] 1.97, 95 % confidence intervals [CI] 1.10-3.50), PVH (OR 1.85, 95 % CI 1.08-3.16) and cerebral atrophy (OR 2.53, 95 % CI 1.19-5.40) but not DWMH (OR 1.37 95 % CI 0.81-2.31) compared with the lowest fibrinogen quartile. Moreover, the association between elevated fibrinogen levels and the risk of WMLs and cerebral atrophy remained significant as continuous variables. CONCLUSIONS Increased baseline fibrinogen levels were independently associated with WMHs, PVH and cerebral atrophy in patients with ischemic stroke. Fibrinogen could be the potential blood biomarker of WMLs and cerebral atrophy.
Collapse
Affiliation(s)
- Suqiu Huan
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Shiya Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lixuan Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Department of Neurology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Chenchen Liu
- Department of Medical Iconography, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jiaping Xu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yilun Ge
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Zhichao Huang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wu Cai
- Department of Medical Iconography, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yongjun Cao
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Shoujiang You
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
42
|
Lan D, Wang M, Zhang X, Huang X, Liu N, Ren X, Fang K, Zhou D, Meng R. A retrospective cohort study on a novel marker to predict the severity and prognosis of acute cerebral venous thrombosis: D-dimer to fibrinogen ratio. Thromb J 2024; 22:95. [PMID: 39478537 PMCID: PMC11523772 DOI: 10.1186/s12959-024-00664-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND AND AIM The D-dimer to fibrinogen ratio (DFR) represents an emerging and significant clinical biomarker. However, its correlation with cerebral venous thrombosis (CVT) remains underexplored. This retrospective cohort study aims to elucidate the association between DFR values and the severity and prognosis of CVT. METHODS Severe CVT was defined as the presence of at least 1 of the following risk factors: mental status disorder, coma state, intracranial cerebral hemorrhage, or thrombosis of the deep cerebral venous system. The modified Rankin Scale was utilized to assess functional outcomes. DFR measurements were obtained within 24 h of hospital admission. Logistic regression analysis was employed to determine the prognostic significance of DFR. After Bonferroni correction, a two-tailed P value < 0.017 (0.05/3) was considered statistically significant. RESULT A total of 196 patients were included in the study, among whom 85 patients were diagnosed with severe CVT, and 35 and 14 patients experienced short-term and long-term adverse outcomes, respectively. Receiver operating characteristic curve analysis demonstrated that DFR has predictive value for severe CVT, poor short-term and long-term outcomes, with area under the curve values of 0.690 [95% CI: 0.617-0.764, P < .001], 0.773 [95% CI: 0.701-0.845, P < .001], and 0.754 [95% CI: 0.619-0.886, P = .002], respectively. DFR ≥ 0.253 was identified as a significant predictor of severe CVT [adjusted odds ratio (aOR) (95% CI): 2.03 (1.10-3.75), P = .024]. Additionally, DFR ≥ 0.322 and DFR ≥ 0.754 were significantly associated with poor short-term outcomes at discharge [aOR (95% CI): 2.63 (1.43-4.76), P = .002] and poor long-term outcomes at 12 months [aOR (95% CI): 2.86 (1.32-6.25), P = .008], respectively. CONCLUSION Elevated DFR is associated with increased severity of CVT. Additionally, higher DFR levels can predict poorer clinical outcomes in CVT.
Collapse
Affiliation(s)
- Duo Lan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Mengqi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaoming Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiangqian Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Naiqi Liu
- Capital Medical University, Beijing, 100069, China
| | - Xiangyu Ren
- Capital Medical University, Beijing, 100069, China
| | - Kun Fang
- Capital Medical University, Beijing, 100069, China
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
43
|
Xin L, Madarasz A, Ivan DC, Weber F, Aleandri S, Luciani P, Locatelli G, Proulx ST. Impairment of spinal CSF flow precedes immune cell infiltration in an active EAE model. J Neuroinflammation 2024; 21:272. [PMID: 39444001 PMCID: PMC11520187 DOI: 10.1186/s12974-024-03247-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024] Open
Abstract
Accumulation of immune cells and proteins in the subarachnoid space (SAS) is found during multiple sclerosis and in the animal model experimental autoimmune encephalomyelitis (EAE). Whether the flow of cerebrospinal fluid (CSF) along the SAS of the spinal cord is impacted is yet unknown. Combining intravital near-infrared (NIR) imaging with histopathological analyses, we observed a significantly impaired bulk flow of CSF tracers within the SAS of the spinal cord prior to EAE onset, which persisted until peak stage and was only partially recovered during chronic disease. The impairment of spinal CSF flow coincided with the appearance of fibrin aggregates in the SAS, however, it preceded immune cell infiltration and breakdown of the glia limitans superficialis. Conversely, cranial CSF efflux to cervical lymph nodes was not altered during the disease course. Our study highlights an early and persistent impairment of spinal CSF flow and suggests it as a sensitive imaging biomarker for pathological changes within the leptomeninges.
Collapse
Affiliation(s)
- Li Xin
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern, CH-3012, Switzerland
| | - Adrian Madarasz
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern, CH-3012, Switzerland
| | - Daniela C Ivan
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern, CH-3012, Switzerland
| | - Florian Weber
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Giuseppe Locatelli
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern, CH-3012, Switzerland
| | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern, CH-3012, Switzerland.
| |
Collapse
|
44
|
Osborne OM, Daftari M, Naranjo O, Johar AN, Brooks S, Colbert BM, Torices S, Lewis E, Sendaydiego J, Drexler G, Bashti M, Margetts AV, Tuesta LM, Mason C, Bilbao D, Vontell R, Griswold AJ, Dykxhoorn DM, Toborek M. Post-stroke hippocampal neurogenesis is impaired by microvascular dysfunction and PI3K signaling in cerebral amyloid angiopathy. Cell Rep 2024; 43:114848. [PMID: 39392753 PMCID: PMC11562893 DOI: 10.1016/j.celrep.2024.114848] [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: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
Ischemic stroke and cerebral amyloid angiopathy (CAA) pose significant challenges in an aging population, particularly in post-stroke recovery. Using the 5xFAD mouse model, we explore the relationship between CAA, ischemic stroke, and tissue recovery. We hypothesize that amyloid-beta accumulation worsens stroke outcomes by inducing blood-brain barrier (BBB) dysfunction, leading to impaired neurogenesis. Our findings show that CAA exacerbates stroke outcomes, with mice exhibiting constricted BBB microvessels, reduced cerebral blood flow, and impaired tissue recovery. Transcriptional analysis shows that endothelial cells and neural progenitor cells (NPCs) in the hippocampus exhibit differential gene expression in response to CAA and stroke, specifically targeting the phosphatidylinositol 3-kinase (PI3K) pathway. In vitro experiments with human NPCs validate these findings, showing that disruption of the CXCL12-PIK3C2A-CREB3L2 axis impairs neurogenesis. Notably, PI3K pathway activation restores neurogenesis, highlighting a potential therapeutic approach. These results suggest that CAA combined with stroke induces microvascular dysfunction and aberrant neurogenesis through this specific pathway.
Collapse
Affiliation(s)
- Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Manav Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adarsh N Johar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Samantha Brooks
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brett M Colbert
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elizabeth Lewis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jet Sendaydiego
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gillian Drexler
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Malek Bashti
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander V Margetts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Luis M Tuesta
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurology and Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Regina Vontell
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anthony J Griswold
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek M Dykxhoorn
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
45
|
Zvenigorodsky V, Gruenbaum BF, Shelef I, Horev A, Azab AN, Oleshko A, Abu-Rabia M, Negev S, Zlotnik A, Melamed I, Boyko M. Evaluation of Blood-Brain Barrier Disruption Using Low- and High-Molecular-Weight Complexes in a Single Brain Sample in a Rat Traumatic Brain Injury Model: Comparison to an Established Magnetic Resonance Imaging Technique. Int J Mol Sci 2024; 25:11241. [PMID: 39457023 PMCID: PMC11508800 DOI: 10.3390/ijms252011241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Traumatic brain injury (TBI), a major cause of death and disability among young people, leads to significant public health and economic challenges. Despite its frequency, treatment options remain largely unsuitable. However, examination of the blood-brain barrier (BBB) can assist with understanding the mechanisms and dynamics of brain dysfunction, which affects TBI sufferers secondarily to the injury. Here, we present a rat model of TBI focused on two standard BBB assessment markers, high- and low-molecular-weight complexes, in order to understand BBB disruption. In addition, we tested a new technique to evaluate BBB disruption on a single brain set, comparing the new technique with neuroimaging. A total of 100 Sprague-Dawley rats were separated into the following five groups: naive rats (n = 20 rats), control rats with administration (n = 20 rats), and TBI rats (n = 60 rats). Rats were assessed at different time points after the injury to measure BBB disruption using low- and high-molecular-weight complexes. Neurological severity score was evaluated at baseline and at 24 h following TBI. During the neurological exam after TBI, the rats were scanned with magnetic resonance imaging and euthanized for assessment of the BBB permeability. We found that the two markers displayed different examples of BBB disruption in the same set of brain tissues over the period of a week. Our innovative protocol for assessing BBB permeability using high- and low-molecular-weight complexes markers in a single brain set showed appropriate results. Additionally, we determined the lower limit of sensitivity, therefore demonstrating the accuracy of this method.
Collapse
Affiliation(s)
- Vladislav Zvenigorodsky
- Department of Radiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (V.Z.); (I.S.)
| | - Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (V.Z.); (I.S.)
| | - Anat Horev
- Department of Neurology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel;
| | - Abed N. Azab
- Department of Nursing, Recanati School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel;
| | - Anna Oleshko
- Department of Biology and Methods of Teaching Biology, A. S. Makarenko Sumy State Pedagogical University, 40002 Sumy, Ukraine;
| | - Mammduch Abu-Rabia
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.A.-R.); (S.N.); (A.Z.)
| | - Shahar Negev
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.A.-R.); (S.N.); (A.Z.)
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.A.-R.); (S.N.); (A.Z.)
| | - Israel Melamed
- Department of Neurosurgery, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel;
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.A.-R.); (S.N.); (A.Z.)
| |
Collapse
|
46
|
Akassoglou K, Davalos D, Mendiola AS, Petersen MA, Ryu JK, Schachtrup C, Yan Z. Pioneering discovery and therapeutics at the brain-vascular-immune interface. Cell 2024; 187:5871-5876. [PMID: 39423805 DOI: 10.1016/j.cell.2024.09.018] [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: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/21/2024]
Abstract
The brain-vascular-immune interface has emerged as a dynamic player in brain physiology and disease. We propose integrating vascular risk factors with genetic susceptibility as the nexus for the discovery of mechanisms and therapies for neuroinflammation, neurodegeneration, and neurorepair across polygenic neurologic diseases.
Collapse
Affiliation(s)
- Katerina Akassoglou
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA; Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Andrew S Mendiola
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Mark A Petersen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA; Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jae Kyu Ryu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA; Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zhaoqi Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA 94158, USA; Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| |
Collapse
|
47
|
Lehikoinen J, Strandin T, Parantainen J, Nurmi K, Eklund KK, Rivera FJ, Vaheri A, Tienari PJ. Fibrinolysis associated proteins and lipopolysaccharide bioactivity in plasma and cerebrospinal fluid in multiple sclerosis. J Neuroimmunol 2024; 395:578432. [PMID: 39151321 DOI: 10.1016/j.jneuroim.2024.578432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/24/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
The coagulation cascade and fibrinolysis have links with neuroinflammation and increased activation of the coagulation system has been reported in MS patients. We quantified levels of D-dimer, tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1) and the bioactivity of bacterial lipopolysaccharide (LPS) in cerebrospinal fluid (CSF) and plasma from newly diagnosed untreated MS patients and controls. These molecules showed multiple correlations with each other as well as with age, HLA-DRB1*15:01, body-mass-index and CSF IgG. Our results confirm previous findings of increased plasma PAI-1 and LPS in MS patients compared to controls indicating changes in platelet function and gut permeability in MS.
Collapse
Affiliation(s)
- Joonas Lehikoinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Neurocenter, Helsinki University Hospital, Helsinki, Finland.
| | - Tomas Strandin
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Jukka Parantainen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Katariina Nurmi
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Kari K Eklund
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Rheumatology, Helsinki University Hospital, Helsinki, Finland
| | - Francisco J Rivera
- Translational Regenerative Neurobiology Group (TReN), Molecular and Integrative Biosciences Research Programme (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Neurocenter, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
48
|
Su Y, Liu N, Wang P, Shang C, Sun R, Ma J, Li Z, Ma H, Sun Y, Zhang Z, Song J, Xie Z, Xu J, Zhang Z. Proteomic analysis and experimental validation reveal the blood-brain barrier protective of Huanshaodan in the treatment of SAMP8 mouse model of Alzheimer's disease. Chin Med 2024; 19:137. [PMID: 39369234 PMCID: PMC11456246 DOI: 10.1186/s13020-024-01016-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/26/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Huanshaodan (HSD) is a Chinese Herbal Compound which has a definite clinical effect on Alzheimer's disease (AD), however, the underlying mechanism remains unclear. The aim of this study is to preliminarily reveal the mechanism of HSD in the treatment of AD model of SAMP8 mice. METHODS Chemical composition of HSD and its drug-containing serum were identified by Q-Orbitrap high resolution liquid mass spectrometry. Six-month-old SAMP8 mice were treated with HSD and Donepezil hydrochloride by gavage for 2 months, and Wogonin for 28 days. Behavioral test was performed to test the learning and memory ability of mice. Immunofluorescence (IF) or Western-blot methods were used to detect the levels of pSer404-tau and β-amyloid (Aβ) in the brain of mice. Hematoxylin-eosin (H&E) staining and Transmission electron microscopy (TEM) assay was applied to observe the pathological changes of neurons. Proteomic technology was carried out to analyze and identify the protein network of HSD interventions in AD. Then the pathological process of the revealed AD-related differential proteins was investigated by IF, Q-PCR, Western-blot, Fluorescence in situ hybridization (FISH) and 16S rRNA sequencing methods. RESULTS The results showed that HSD and Wogonin, one of the components in its drug-containing serum, can effectively improve the cognitive impairments of SAMP8 mice, protect hippocampal neurons and synapses, and reduce the expression of pSer404-tau and Aβ. HSD and Wogonin reduced the levels of fibrinogen β chain (FGB) and γ chain (FGG), the potential therapeutic targets revealed by proteomics analysis, reduced the colocalization of FGB and FGG with Aβ, ionized calcium binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), increased level of and myelin basic protein (MBP). Meanwhile, HSD and Wogonin increased ZO-1 and Occludin levels, improved brain microvascular injury, and reduced levels of bacteria/bacterial DNA and lipopolysaccharide (LPS) in the brain of mice. In addition, 16S rRNA sequencing indicated that HSD regulated the structure of intestinal microbiota of mice. CONCLUSION The effects of HSD on AD may be achieved by inhibiting the levels of fibrinogen and the interactions on glia cells in the brain, and by modulating the structure of intestinal microbiota and improving the blood-brain barrier function.
Collapse
Affiliation(s)
- Yunfang Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19, Renmin Road, Zhengzhou, 450046, China
| | - Ningning Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Pan Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Congcong Shang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Ruiqin Sun
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Jinlian Ma
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zhonghua Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Huifen Ma
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Yiran Sun
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zijuan Zhang
- School of Basic Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Junying Song
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zhishen Xie
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| | - Jiangyan Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| | - Zhenqiang Zhang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| |
Collapse
|
49
|
Chen Y, Wang Y, Zhang M, Zhou Y, Zhang H, Li P, Wu J. The clinical and neuropsychological profiles of Alzheimer's disease with white matter hyperintensity in North China. Front Neurol 2024; 15:1436030. [PMID: 39416665 PMCID: PMC11480061 DOI: 10.3389/fneur.2024.1436030] [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: 05/21/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Patients with Alzheimer's disease (AD) often exhibit characteristic clinical manifestations, particularly neuropsychiatric symptoms. Previous studies have shown that white matter hyperintensity (WMH) is strongly associated with AD progression, as well as neuropsychiatric symptoms. The purpose of this study was to investigate the clinical and neuropsychological characteristics of AD patients with WMH. Methods This retrospective study involved 104 18-fluorodeoxyglucose-positron emission computed tomography (18FDG-PET-CT)-defined AD patients treated at Tianjin Huanhu Hospital from January 2010 to December 2022. Cranial magnetic resonance imaging (MRI) provided semi-quantitative data on brain structure and WMH. Collect and analyze patient clinical data. Neuropsychological assessments were used to evaluate cognitive function and psychobehavioral traits. Results Among the 104 patients, 66 were in the WMH group (63.5%) and 38 in the non-white matter hyperintensity (non-WMH) group (36.5%). There were no significant differences in gender, age, age of onset, education, BMI, smoking, drinking, diabetes, coronary heart disease, dementia family history, fasting blood glucose, total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) between the two groups. The WMH group showed higher rates of hypertension, homocysteine (Hcy) levels, NPI, and CDR scores as compared to the non-WMH group (p < 0.05). MMSE and MoCA scores were significantly lower in the WMH group (p < 0.05). In the MMSE subitem analysis, patients in the WMH group showed a decrease in attention, recall, and language scores. In the MOCA subitem analysis, WMH patients had lower scores in executive function, naming, attention, language, abstraction, and orientation (p < 0.05). Furthermore, subgroup analysis of NPI showed a higher incidence of delusions, depression, and apathy in the WMH group (p < 0.05). According to the hierarchical analysis of mild, moderate and severe dementia groups, the hypertension, leukoencephalopathy, Hcy level, Fazekas total score, PWMH and DWMH scores in the severe dementia group were significantly higher than those in the mild and moderate dementia groups (p < 0.05). As the disease progresses, more and more patients show increased white matter hyperintensity. Conclusion White matter lesions are closely correlated with cognitive decline and psychobehavioral symptoms in AD patients, and may be used as an indicator of disease progression. Priority should be given to early screening and prevention of WMH-related risk factors.
Collapse
Affiliation(s)
- Yuan Chen
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Miao Zhang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Huihong Zhang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Pan Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Jialing Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital Affiliated to Tianjin Medical University, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
50
|
Dobson DA, Fish RJ, de Vries PS, Morrison AC, Neerman-Arbez M, Wolberg AS. Regulation of fibrinogen synthesis. Thromb Res 2024; 242:109134. [PMID: 39216273 PMCID: PMC11381137 DOI: 10.1016/j.thromres.2024.109134] [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: 07/02/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The plasma protein fibrinogen is encoded by 3 structural genes (FGA, FGB, and FGG) that are transcribed to mRNA, spliced, and translated to 3 polypeptide chains (Aα, Bβ, and γ, respectively). These chains are targeted for secretion, decorated with post-translational modifications, and assembled into a hexameric "dimer of trimers" (AαBβγ)2. Fully assembled fibrinogen is secreted into the blood as a 340 kDa glycoprotein. Fibrinogen is one of the most prevalent coagulation proteins in blood, and its expression is induced by inflammatory cytokines, wherein circulating fibrinogen levels may increase up to 3-fold during acute inflammatory events. Abnormal levels of circulating fibrinogen are associated with bleeding and thrombotic disorders, as well as several inflammatory diseases. Notably, therapeutic strategies to modulate fibrinogen levels have shown promise in experimental models of disease. Herein, we review pathways mediating fibrinogen synthesis, from gene expression to secretion. Knowledge of these mechanisms may lead to the identification of biomarkers and new therapeutic targets to modulate fibrinogen in health and disease.
Collapse
Affiliation(s)
- Dre'Von A Dobson
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, NC, USA
| | - Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|