1
|
Conejero I, Chea M, Courtet P, Bouvier S, Pereira F. Alterations in the Blood-Brain Barrier in Mood Disorders and Neurodegenerative Diseases. Semin Thromb Hemost 2025; 51:457-464. [PMID: 40058364 DOI: 10.1055/s-0045-1804893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Depressive disorders and suicidal behaviors represent major causes of health loss. Modifications of brain microvasculature, and specifically alterations of the blood-brain barrier have been supposed to participate in the vulnerability to those disorders along with cognitive impairment, especially in the older adults. In this article, we addressed evidence linking blood-brain barrier impairments with mood disorders and suicide. Secondly, we investigated their relationship with depression in old age, and with neurodegenerative processes. Particular attention was drawn toward the potential interactions between the coagulation processes and the blood-brain barrier dysfunctions, as innovative treatment strategies may emerge from research in those fields. Overall, the studies reviewed highlight the implication of multiple dysfunctions of the blood-brain barrier in mood disorders and suicide. Impairments of the blood-brain barrier show relationships with altered expression of endothelial cell junction proteins. These modifications also implicate receptors of the extracellular matrix, the vascular endothelial growth factor, changes in perivascular astrocytes, and has links with local and systemic inflammatory processes. Dysfunctions of the blood-brain barrier underly chronic stress and participate in psychiatric diathesis in old age. In addition, we outline that coagulation processes are likely to interact with the blood-brain barrier and further contribute to neurodegenerative disorders. In conclusion, new pathophysiological models offer perspectives toward detecting new biomarkers in mood disorders and suicide. In parallel, these models open avenues for developing innovative therapeutic agents, although further considering their potential risks and eventual benefits is needed.
Collapse
Affiliation(s)
- Ismael Conejero
- Department of Psychiatry, CHU Nîmes, University of Montpellier, Nîmes, France
| | - Mathias Chea
- Department of Hematology, CHU Nîmes, Nîmes, France
- UMR 1318 INSERM-Université de Montpellier IDESP, France
- Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Philippe Courtet
- Institute of Functional Genomics (IGF), University of Montpellier, Montpellier, France
- Department of Emergency Psychiatry & Acute Care, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Sylvie Bouvier
- Department of Hematology, CHU Nîmes, Nîmes, France
- UMR 1318 INSERM-Université de Montpellier IDESP, France
- Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Fabricio Pereira
- Department of Radiology and Medical Imaging, CHU Nîmes, Nîmes, France
- MIPA, University of Nîmes, Nîmes, France
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Gust KA, Amar SK, Gut CP, Styles RM, Karna RR, James RA, Holtzapple DM, Stricker JL, McInturf SM, Phillips EA, Honnold C, Luo X, Mumy KL, Mattie DR, Chappell MA, Mayo ML. Multi-disciplinary investigation identifies increased potency of ethyl-parathion inhaled within a soil-dust matrix to cause acetylcholinesterase-dependent molecular impacts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104615. [PMID: 39710123 DOI: 10.1016/j.etap.2024.104615] [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: 08/16/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Neurotoxicity investigations of inhaled organophosphorus pesticide (OP), ethyl-parathion (EP), were conducted in Sprague Dawley rats comparing exposures to EP volatilized at 0, 1, 10, and 20 mg/m3 versus EP incorporated into soil dust (5 mg/m3) at 0, 0.0095, 0.09, and 0.185 mg/mg3. All exposures were sublethal, caused no respiratory effects, and no effects on balance and coordination behavior. Both volatilized and dust-incorporated EP exposures significantly decreased acetylcholinesterase (AChE) activity in plasma and hippocampus tissue. Correspondingly, plasma and hippocampal dopamine levels spiked in these exposures suggesting compensatory cholinergic / dopaminergic signal balancing. The EP exposures significantly increased expression of pro-inflammatory genes, including MAPK-14, IL6, IL1β, and TNF-α, while global RNA-seq results identified significant enrichment of inflammation, oxidative stress, and apoptosis pathways. Remarkably, dust-incorporated EP impacted similar molecular endpoints as volatilized EP but at concentrations two orders of magnitude lower highlighting potentially increased potency of EP incorporated into soil dust.
Collapse
Affiliation(s)
- Kurt A Gust
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States.
| | - Saroj K Amar
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States; Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Chet P Gut
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Renee M Styles
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Ranju R Karna
- Credere Associates LLC, Westbrook, ME located at US Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - R Arden James
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David M Holtzapple
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Joshua L Stricker
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Shawn M McInturf
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Elizabeth A Phillips
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Cary Honnold
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Xiao Luo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Karen L Mumy
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David R Mattie
- Air Force Research Laboratory/711 HPW, Wright-Patterson Air Force Base, OH, United States
| | - Mark A Chappell
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Michael L Mayo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Ghebosu RE, Hui L, Wolfram J. Increasing the biomolecular relevance of cell culture practice. J Biomed Sci 2025; 32:3. [PMID: 39748368 PMCID: PMC11697962 DOI: 10.1186/s12929-024-01095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/09/2024] [Indexed: 01/04/2025] Open
Abstract
The biomolecular relevance of medium supplements is a key challenge affecting cell culture practice. The biomolecular composition of commonly used supplements differs from that of a physiological environment, affecting the validity of conclusions drawn from in vitro studies. This article discusses the advantages and disadvantages of common supplements, including context-dependent considerations for supplement selection to improve biomolecular relevance, especially in nanomedicine and extracellular vesicle research.
Collapse
Affiliation(s)
- Raluca E Ghebosu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia
| | - Lawrence Hui
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia.
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
9
|
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
|
10
|
Schreiber S, Arndt P, Morton L, Garza AP, Müller P, Neumann K, Mattern H, Dörner M, Bernal J, Vielhaber S, Meuth SG, Dunay IR, Dityatev A, Henneicke S. Immune system activation and cognitive impairment in arterial hypertension. Am J Physiol Cell Physiol 2024; 327:C1577-C1590. [PMID: 39495252 PMCID: PMC11684865 DOI: 10.1152/ajpcell.00219.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Chronic arterial hypertension disrupts the integrity of the cerebral microvasculature, doubling the risk of age-related dementia. Despite sufficient antihypertensive therapy in still a significant proportion of individuals blood pressure lowering alone does not preserve cognitive health. Accumulating evidence highlights the role of inflammatory mechanisms in the pathogenesis of hypertension. In this review, we introduce a temporal framework to explore how early immune system activation and interactions at neurovascular-immune interfaces pave the way to cognitive impairment. The overall paradigm suggests that prohypertensive stimuli induce mechanical stress and systemic inflammatory responses that shift peripheral and meningeal immune effector mechanisms toward a proinflammatory state. Neurovascular-immune interfaces in the brain include a dysfunctional blood-brain barrier, crossed by peripheral immune cells; the perivascular space, in which macrophages respond to cerebrospinal fluid- and blood-derived immune regulators; and the meningeal immune reservoir, particularly T cells. Immune responses at these interfaces bridge peripheral and neurovascular unit inflammation, directly contributing to impaired brain perfusion, clearance of toxic metabolites, and synaptic function. We propose that deep immunophenotyping in biofluids together with advanced neuroimaging could aid in the translational determination of sequential immune and brain endotypes specific to arterial hypertension. This could close knowledge gaps on how and when immune system activation transits into neurovascular dysfunction and cognitive impairment. In the future, targeting specific immune mechanisms could prevent and halt hypertension disease progression before clinical symptoms arise, addressing the need for new interventions against one of the leading threats to cognitive health.
Collapse
Affiliation(s)
- Stefanie Schreiber
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Arndt
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
| | - Lorena Morton
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Patrick Müller
- Department of Cardiology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Katja Neumann
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Biomedical Magnetic Resonance, Faculty of Natural Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Marc Dörner
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Switzerland
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ildiko R Dunay
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Solveig Henneicke
- Department of Neurology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Magdeburg, Germany
| |
Collapse
|
11
|
Wen Q, Sun T, Chen J, Li Y, Liu X, Li H, Fu R, Liu W, Xue F, Ju M, Dong H, Dai X, Wang W, Chi Y, Yang R, Chen Y, Zhang L. Integrating chemokines and machine learning algorithms for diagnosis and bleeding assessment in primary immune thrombocytopenia: A prospective cohort study. Br J Haematol 2024; 205:1938-1950. [PMID: 39253817 DOI: 10.1111/bjh.19745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
Primary immune thrombocytopenia (ITP) is an autoimmune bleeding disorder, and chemokines have been shown to be dysregulated in autoimmune disorders. We conducted a prospective analysis to identify potential chemokines that could enhance the diagnostic accuracy and bleeding evaluation in ITP patients. In the discovery cohort, a Luminex-based assay was employed to quantify concentrations of plasma multiple chemokines. These levels were subjected to comparative analysis using a cohort of 60 ITP patients and 17 patients with thrombocytopenia other than ITP (non-ITP). Additionally, comparative evaluation was conducted between a subgroup of 12 ITP patients characterised by bleeding episodes (ITP-B, as defined by an ITP-2016 bleeding grade ≥2) and 33 ITP patients without bleeding episodes (ITP-NB, as defined by an ITP-2016 bleeding grade ≤1). Machine learning algorithms further identified CCL20, interleukin-2, CCL26, CCL25, and CXCL1 as promising indicators for accurate diagnosis of ITP and CCL21, CXCL8, CXCL10, CCL8, CCL3, and CCL15 as biomarkers for assessing bleeding risk in ITP patients. The results were confirmed using enzyme-linked immunosorbent assays in a validation cohort (43 ITP patients and 19 non-ITP patients). Overall, the findings suggest that specific chemokines show promise as potential biomarkers for diagnosis and bleeding evaluation in ITP patients.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ting Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Feng Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mankai Ju
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huan Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xinyue Dai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ying Chi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yunfei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Yue Q, Leng X, Xie N, Zhang Z, Yang D, Hoi MPM. Endothelial Dysfunctions in Blood-Brain Barrier Breakdown in Alzheimer's Disease: From Mechanisms to Potential Therapies. CNS Neurosci Ther 2024; 30:e70079. [PMID: 39548663 PMCID: PMC11567945 DOI: 10.1111/cns.70079] [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: 07/17/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 11/18/2024] Open
Abstract
Recent research has shown the presence of blood-brain barrier (BBB) breakdown in Alzheimer's disease (AD). BBB is a dynamic interface consisting of a continuous monolayer of brain endothelial cells (BECs) enveloped by pericytes and astrocytes. The restricted permeability of BBB strictly controls the exchange of substances between blood and brain parenchyma, which is crucial for brain homeostasis by excluding blood-derived detrimental factors and pumping out brain-derived toxic molecules. BBB breakdown in AD is featured as a series of BEC pathologies such as increased paracellular permeability, abnormal levels and functions of transporters, and inflammatory or oxidative profile, which may disturb the substance transportation across BBB, thereafter induce CNS disorders such as hypometabolism, Aβ accumulation, and neuroinflammation, eventually aggravate cognitive decline. Therefore, it seems important to protect BEC properties for BBB maintenance and neuroprotection. In this review, we thoroughly summarized the pathological alterations of BEC properties reported in AD patients and numerous AD models, including paracellular permeability, influx and efflux transporters, and inflammatory and oxidative profiles, and probably associated underlying mechanisms. Then we reviewed current therapeutic agents that are effective in ameliorating a series of BEC pathologies, and ultimately protecting BBB integrity and cognitive functions. Regarding the current drug development for AD proceeds extremely hard, this review aims to discuss the therapeutic potentials of targeting BEC pathologies and BBB maintenance for AD treatment, therefore expecting to shed a light on the future AD drug development by targeting BEC pathologies and BBB protection.
Collapse
Affiliation(s)
- Qian Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
| | - Xinyue Leng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| | - Ningqing Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Deguang Yang
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| |
Collapse
|
13
|
Chea M, Bouvier S, Gris JC. The hemostatic system in chronic brain diseases: A new challenging frontier? Thromb Res 2024; 243:109154. [PMID: 39305718 DOI: 10.1016/j.thromres.2024.109154] [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: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Chea
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France.
| | - Sylvie Bouvier
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France; I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
17
|
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
|
18
|
Tang N. Exosomes in multiple sclerosis and Alzheimer's disease - Adversary and ally. Biomed J 2024; 47:100665. [PMID: 37778696 PMCID: PMC11401191 DOI: 10.1016/j.bj.2023.100665] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023] Open
Abstract
Neuroinflammation and the resulting neurodegeneration is a big challenge for the healthcare system, especially with the aging population. Neuroinflammation can result from a variety of insults to the central nervous system leading to an interplay between immune and brain cells that sustains chronic inflammation and injures neural cells. One facilitator of this toxic interplay are exosomes. Exosomes are nano-sized, bilayer lipid vesicles secreted by cells containing proteins, nucleic acids and lipids. Because exosomes can be internalized by other cells, their contents can elicit inflammatory responses and trigger toxicities in recipient cells. On the flip side, exosomes can act as therapeutic vehicles carrying protective cargo to maintain homeostasis. This review discusses exosome biogenesis, composition, and its role in neuroinflammation and neurodegeneration in the context of multiple sclerosis and Alzheimer's disease. The emerging roles of exosomes as biomarkers of neurologic diseases and as therapeutic delivery vehicles are also discussed. With all of these varying roles, interest and excitement in exosomes continue to grow exponentially and their promise as brain therapeutics is only beginning to be explored and harnessed.
Collapse
Affiliation(s)
- Norina Tang
- Department of Periodontics, University of the Pacific, San Francisco, USA; Department of Laboratory Medicine, San Francisco Veterans Affairs Health Care System, San Francisco, USA.
| |
Collapse
|
19
|
Ryu JK, Yan Z, Montano M, Sozmen EG, Dixit K, Suryawanshi RK, Matsui Y, Helmy E, Kaushal P, Makanani SK, Deerinck TJ, Meyer-Franke A, Rios Coronado PE, Trevino TN, Shin MG, Tognatta R, Liu Y, Schuck R, Le L, Miyajima H, Mendiola AS, Arun N, Guo B, Taha TY, Agrawal A, MacDonald E, Aries O, Yan A, Weaver O, Petersen MA, Meza Acevedo R, Alzamora MDPS, Thomas R, Traglia M, Kouznetsova VL, Tsigelny IF, Pico AR, Red-Horse K, Ellisman MH, Krogan NJ, Bouhaddou M, Ott M, Greene WC, Akassoglou K. Fibrin drives thromboinflammation and neuropathology in COVID-19. Nature 2024; 633:905-913. [PMID: 39198643 PMCID: PMC11424477 DOI: 10.1038/s41586-024-07873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/24/2024] [Indexed: 09/01/2024]
Abstract
Life-threatening thrombotic events and neurological symptoms are prevalent in COVID-19 and are persistent in patients with long COVID experiencing post-acute sequelae of SARS-CoV-2 infection1-4. Despite the clinical evidence1,5-7, the underlying mechanisms of coagulopathy in COVID-19 and its consequences in inflammation and neuropathology remain poorly understood and treatment options are insufficient. Fibrinogen, the central structural component of blood clots, is abundantly deposited in the lungs and brains of patients with COVID-19, correlates with disease severity and is a predictive biomarker for post-COVID-19 cognitive deficits1,5,8-10. Here we show that fibrin binds to the SARS-CoV-2 spike protein, forming proinflammatory blood clots that drive systemic thromboinflammation and neuropathology in COVID-19. Fibrin, acting through its inflammatory domain, is required for oxidative stress and macrophage activation in the lungs, whereas it suppresses natural killer cells, after SARS-CoV-2 infection. Fibrin promotes neuroinflammation and neuronal loss after infection, as well as innate immune activation in the brain and lungs independently of active infection. A monoclonal antibody targeting the inflammatory fibrin domain provides protection from microglial activation and neuronal injury, as well as from thromboinflammation in the lung after infection. Thus, fibrin drives inflammation and neuropathology in SARS-CoV-2 infection, and fibrin-targeting immunotherapy may represent a therapeutic intervention for patients with acute COVID-19 and long COVID.
Collapse
Affiliation(s)
- Jae Kyu Ryu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Zhaoqi Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Elif G Sozmen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Karuna Dixit
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ekram Helmy
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Prashant Kaushal
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Sara K Makanani
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas J Deerinck
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
| | | | | | - Troy N Trevino
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Min-Gyoung Shin
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Reshmi Tognatta
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yixin Liu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Renaud Schuck
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Lucas Le
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Hisao Miyajima
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Andrew S Mendiola
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Nikhita Arun
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Brandon Guo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Eilidh MacDonald
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Oliver Aries
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Aaron Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Olivia Weaver
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Mark A Petersen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Rosa Meza Acevedo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
| | - Igor F Tsigelny
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander R Pico
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Nevan J Krogan
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Katerina Akassoglou
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
20
|
Sun W, Yang Z, Wang Y, Miao J, Pan C, Li G, Liang W, Zhao X, Lan Y, Qiu X, Wang H, Chen M, Yang Y. Peripheral inflammation and trajectories of depressive symptomology after ischemic stroke: A prospective cohort study. J Affect Disord 2024; 359:14-21. [PMID: 38729221 DOI: 10.1016/j.jad.2024.05.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 03/27/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Understanding the association of peripheral inflammation and post-stroke depressive symptomology (PSDS) might provide further insights into the complex etiological mechanism of organic depression. However, studies focusing on the longitudinal patterns of PSDS were limited and it remained unclear whether peripheral inflammation influences the occurrence and development of PSDS. METHODS A total of 427 prospectively enrolled and followed ischemic stroke patients were included in the analytical sample. Depressive symptomology was assessed on four occasions during 1 year after ischemic stroke. Peripheral inflammatory proteins on admission and repeated measures of peripheral immune markers in three stages were collected. Latent class growth analysis (LCGA) was employed to delineate group-based trajectories of peripheral immune markers and PSDS. Multinomial regression was performed to investigate the association of peripheral inflammation with PSDS trajectories. RESULTS Four distinct trajectories of PSDS were identified: stable-low (n = 237, 55.5 %), high-remitting (n = 120, 28.1 %), late-onset (n = 44, 10.3 %), and high-persistent (n = 26, 6.1 %) PSDS trajectories. The elevation of peripheral fibrinogen on admission increased the risk of high-persistent PSDS in patients with early high PSDS. Additionally, chronic elevation of innate immune levels might not only increase the risk of high-persistent PSDS in patients with early high PSDS but also increase the risk of late-onset PSDS in patients without early high PSDS. The elevation of adaptive immune levels in the convalescence of ischemic stroke may contribute to the remission of early high PSDS. CONCLUSIONS Peripheral immunity could influence the development of PSDS, and this influence might have temporal heterogeneity. These results might provide vital clues for the inflammation hypothesis of PSD.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Zhaoxia Yang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Yanyan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China.
| | - Chensheng Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Guo Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Wenwen Liang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Xin Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China.
| | - Yan Lan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Xiuli Qiu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Hao Wang
- Department of Neurology, General Hospital of the Yangtze River Shipping, No.5 Huiji Road, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China
| | - Yuan Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
21
|
Mueller TT, Pilartz M, Thakur M, LangHeinrich T, Luo J, Block R, Hoeflinger JKL, Meister S, Karaj F, Perez LG, Öllinger R, Engleitner T, Thoss J, Voelkl M, Tersteeg C, Koedel U, Kohlmaier AZ, Teupser D, Wygrecka M, Ye H, Preissner KT, Radbruch H, Elezkurtaj S, Mack M, Von Hundelshausen P, Weber C, Massberg S, Schulz C, Rad R, Huber S, Ishikawa-Ankerhold H, Engelmann B. Mutual regulation of CD4 + T cells and intravascular fibrin in infections. Haematologica 2024; 109:2487-2499. [PMID: 38572559 PMCID: PMC11290509 DOI: 10.3324/haematol.2023.284619] [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: 11/05/2023] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
Innate myeloid cells especially neutrophils and their extracellular traps are known to promote intravascular coagulation and thrombosis formation in infections and various other conditions. Innate myeloid cell-dependent fibrin formation can support systemic immunity while its dysregulation enhances the severity of infectious diseases. Less is known about the immune mechanisms preventing dysregulation of fibrin homeostasis in infection. During experimental systemic infections local fibrin deposits in the liver microcirculation cause rapid arrest of CD4+ T cells. Arrested T-helper cells mostly represent Th17 cells that partially originate from the small intestine. Intravascular fibrin deposits activate mouse and human CD4+ T cells which can be mediated by direct fibrin-CD4+ T-cell interactions. Activated CD4+ T cells suppress fibrin deposition and microvascular thrombosis by directly counteracting coagulation activation by neutrophils and classical monocytes. T-cell activation, which is initially triggered by IL-12p40- and MHC-II-dependent mechanisms, enhances intravascular fibrinolysis via LFA-1. Moreover, CD4+ T cells disfavor the association of the thrombin-activatable fibrinolysis inhibitor (TAFI) with fibrin whereby fibrin deposition is increased by TAFI in the absence but not in the presence of T cells. In human infections thrombosis development is inversely related to microvascular levels of CD4+ T cells. Thus, fibrin promotes LFA-1-dependent T-helper cell activation in infections which drives a negative feedback cycle that rapidly restricts intravascular fibrin and thrombosis development.
Collapse
Affiliation(s)
- Tonina T Mueller
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich, Germany; Medizinische Klinik I, Klinikum der Universität München, LMU, Munich.
| | - Mona Pilartz
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Manovriti Thakur
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Torben LangHeinrich
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Junfu Luo
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Rebecca Block
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Jonathan K L Hoeflinger
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Sarah Meister
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Flavio Karaj
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Laura Garcia Perez
- 1. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Rupert Öllinger
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Thomas Engleitner
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Jakob Thoss
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Michael Voelkl
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk
| | - Uwe Koedel
- Neurologische Klinik, Klinikum der Universität München,LMU, Munich
| | - Alexander Zigman Kohlmaier
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Daniel Teupser
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung (CIGL), Justus-Liebig-Universität, Giessen
| | - Haifeng Ye
- Institute of Regenerative Biology and Medicine, Helmholtz-Zentrum München, Munich
| | | | - Helena Radbruch
- Institut für Neuropathologie, Charité - Universitätsmedizin, Berlin
| | | | - Matthias Mack
- Medizinische Klinik II, University of Regensburg, Regensburg
| | - Philipp Von Hundelshausen
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Ludwig-Maximilians-Universität, Munich
| | - Christian Weber
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Ludwig-Maximilians-Universität, Munich
| | - Steffen Massberg
- Medizinische Klinik I, Klinikum der Universität München, LMU, Munich
| | - Christian Schulz
- Medizinische Klinik I, Klinikum der Universität München, LMU, Munich
| | - Roland Rad
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Samuel Huber
- 1. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | | | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich.
| |
Collapse
|
22
|
Weaver O, Gano D, Zhou Y, Kim H, Tognatta R, Yan Z, Ryu JK, Brandt C, Basu T, Grana M, Cabriga B, Alzamora MDPS, Barkovich AJ, Akassoglou K, Petersen MA. Fibrinogen inhibits sonic hedgehog signaling and impairs neonatal cerebellar development after blood-brain barrier disruption. Proc Natl Acad Sci U S A 2024; 121:e2323050121. [PMID: 39042684 PMCID: PMC11295022 DOI: 10.1073/pnas.2323050121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Cerebellar injury in preterm infants with central nervous system (CNS) hemorrhage results in lasting neurological deficits and an increased risk of autism. The impact of blood-induced pathways on cerebellar development remains largely unknown, so no specific treatments have been developed to counteract the harmful effects of blood after neurovascular damage in preterm infants. Here, we show that fibrinogen, a blood-clotting protein, plays a central role in impairing neonatal cerebellar development. Longitudinal MRI of preterm infants revealed that cerebellar bleeds were the most critical factor associated with poor cerebellar growth. Using inflammatory and hemorrhagic mouse models of neonatal cerebellar injury, we found that fibrinogen increased innate immune activation and impeded neurogenesis in the developing cerebellum. Fibrinogen inhibited sonic hedgehog (SHH) signaling, the main mitogenic pathway in cerebellar granule neuron progenitors (CGNPs), and was sufficient to disrupt cerebellar growth. Genetic fibrinogen depletion attenuated neuroinflammation, promoted CGNP proliferation, and preserved normal cerebellar development after neurovascular damage. Our findings suggest that fibrinogen alters the balance of SHH signaling in the neurovascular niche and may serve as a therapeutic target to mitigate developmental brain injury after CNS hemorrhage.
Collapse
Affiliation(s)
- Olivia Weaver
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Dawn Gano
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Hosung Kim
- Department of Neurology, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Reshmi Tognatta
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Zhaoqi Yan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Jae Kyu Ryu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
| | - Caroline Brandt
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Trisha Basu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Martin Grana
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
| | - Belinda Cabriga
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - Maria del Pilar S. Alzamora
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| | - A. James Barkovich
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA94143
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
| | - Mark A. Petersen
- Department of Pediatrics, University of California San Francisco, San Francisco, CA94158
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Center for Neurovascular Brain Immunology at Gladstone Institutes and University of California San Francisco, San Francisco, CA94158
| |
Collapse
|
23
|
Natarajan C, Le LHD, Gunasekaran M, Tracey KJ, Chernoff D, Levine YA. Electrical stimulation of the vagus nerve ameliorates inflammation and disease activity in a rat EAE model of multiple sclerosis. Proc Natl Acad Sci U S A 2024; 121:e2322577121. [PMID: 38968104 PMCID: PMC11252997 DOI: 10.1073/pnas.2322577121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Multiple sclerosis (MS) is a demyelinating central nervous system (CNS) disorder that is associated with functional impairment and accruing disability. There are multiple U.S. Food and Drug Administration (FDA)-approved drugs that effectively dampen inflammation and slow disability progression. However, these agents do not work well for all patients and are associated with side effects that may limit their use. The vagus nerve (VN) provides a direct communication conduit between the CNS and the periphery, and modulation of the inflammatory reflex via electrical stimulation of the VN (VNS) shows efficacy in ameliorating pathology in several CNS and autoimmune disorders. We therefore investigated the impact of VNS in a rat experimental autoimmune encephalomyelitis (EAE) model of MS. In this study, VNS-mediated neuroimmune modulation is demonstrated to effectively decrease EAE disease severity and duration, infiltration of neutrophils and pathogenic lymphocytes, myelin damage, blood-brain barrier disruption, fibrinogen deposition, and proinflammatory microglial activation. VNS modulates expression of genes that are implicated in MS pathogenesis, as well as those encoding myelin proteins and transcription factors regulating new myelin synthesis. Together, these data indicate that neuroimmune modulation via VNS may be a promising approach to treat MS, that not only ameliorates symptoms but potentially also promotes myelin repair (remyelination).
Collapse
Affiliation(s)
| | | | | | - Kevin J. Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY11030
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
| | | | - Yaakov A. Levine
- SetPoint Medical, Valencia, CA91355
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm171 76, Sweden
| |
Collapse
|
24
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Corrigendum to "Platelets and Hemostatic Proteins are Co-Localized with Chronic Neuroinflammation Surrounding Implanted Intracortical Microelectrodes" [Acta Biomaterialia. Volume 166, August 2023, Pages 278-290]. Acta Biomater 2024; 182:303-308. [PMID: 38845260 PMCID: PMC11295673 DOI: 10.1016/j.actbio.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
25
|
Wu X, Yu X, Chen C, Chen C, Wang Y, Su D, Zhu L. Fibrinogen and tumors. Front Oncol 2024; 14:1393599. [PMID: 38779081 PMCID: PMC11109443 DOI: 10.3389/fonc.2024.1393599] [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: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Elevated plasma fibrinogen (Fg) levels consistently correlate with an unfavorable prognosis in various tumor patient cohorts. Within the tumor microenvironment, aberrant deposition and expression of Fg have been consistently observed, interacting with multiple cellular receptors and thereby accentuating its role as a regulator of inflammatory processes. Specifically, Fg serves to stimulate and recruit immune cells and pro-inflammatory cytokines, thereby contributing to the promotion of tumor progression. Additionally, Fg and its fragments exhibit dichotomous effects on tumor angiogenesis. Notably, Fg also facilitates tumor migration through both platelet-dependent and platelet-independent mechanisms. Recent studies have illuminated several tumor-related signaling pathways influenced by Fg. This review provides a comprehensive summary of the intricate involvement of Fg in tumor biology, elucidating its multifaceted role and the underlying mechanisms.
Collapse
Affiliation(s)
- Xinyuan Wu
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaomin Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Chen
- Department of Hematology, Wenzhou Key Laboratory of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenlu Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dongyan Su
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liqing Zhu
- Department of Clinical Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
26
|
Li M, Gan J, Yang X, Liu S, Ji Y. Cerebrospinal fluid/serum albumin ratio in patients with Lewy body disease: a systematic review and meta-analysis. Front Aging Neurosci 2024; 16:1390036. [PMID: 38756533 PMCID: PMC11096505 DOI: 10.3389/fnagi.2024.1390036] [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: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Background Abnormal cerebrospinal fluid (CSF)/serum albumin ratio (Qalb) levels have been observed in patients with cognitive impairment. Few studies have specifically focused on Lewy Body Disease (LBD), and the results were controversial. Thus, we conducted this systematic review and meta-analysis to investigate Qalb levels in patients with LBD by including data from different studies. Method We systematically searched PubMed, Embase, Cochrane Library, and Web of Science databases for a collection of studies containing studies comparing Qalb levels in patients with LBD and healthy controls (including healthy controls and other dementia subtypes). In the initial search, 86 relevant papers were retrieved. Standardized mean differences (SMD) in Qalb levels were calculated using a random effects model. Results A total of 13 eligible studies were included. Mean Qalb levels were significantly higher in patients with LBD compared to healthy older adults [standardized mean difference (SMD): 2.95, 95% confidence interval (CI): 0.89-5.00, Z = 2.81, p = 0.005]; and were significantly higher in patients with LBD than in patients with Alzheimer's disease (AD) (SMD: 1.13, 95% CI: 0.42-1.83, Z = 3.15, p = 0.002);whereas mean Qalb levels were significantly higher in patients with frontotemporal lobar degeneration (FTLD) compared to those with AD (SMD: 1.13, 95% CI,0.14-2.13, Z = 2.24, p = 0.03). Conclusion Qalb levels were significantly elevated in LBD patients compared with normal older adults and were higher than those in AD patients and FTLD patients, which helped in the differential diagnosis of LBD from other neurodegenerative diseases. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024496616.
Collapse
Affiliation(s)
- Moyu Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinghuan Gan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
27
|
Han L, Song Y, Xiang W, Wang Z, Wang Y, Zhou X, Zhu DS, Guan Y. Fibrinogen deposition promotes neuroinflammation and fibrin-derived γ 377-395 peptide ameliorates neurological deficits after ischemic stroke. Int Immunopharmacol 2024; 131:111831. [PMID: 38489969 DOI: 10.1016/j.intimp.2024.111831] [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: 09/22/2023] [Revised: 02/09/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Fibrin(ogen) deposition in the central nervous system (CNS) contributes to neuropathological injury; however, its role in ischemic stroke is unknown. In this study, we identified fibrinogen as a novel proinflammatory regulator of post-stroke neuroinflammation and revealed the neuro-protection effect of fibrin-derived γ377-395peptide in stroke. METHODS Fibrinogen depletion and fibrinogen-derived γ377-395peptide treatment were performed 2 h after establishing a permanent middle cerebral artery occlusion (pMCAO) model. The infarction volume, neurological score, fibrin(ogen) deposition, and inflammatory response were evaluated 24 h after occlusion. Both in vivo and in vitro studies were conducted to assess the therapeutic potential of the γ377-395peptide in blocking the interactions between fibrin(ogen) and neutrophils. RESULTS Fibrin(ogen) deposited in the infarct core promoted post-stroke inflammation and exacerbated neurological deficits in the acute phase after stroke onset. Reducing fibrinogen deposition resulted in a decrease in infarction volume, improved neurological scores, and reduced inflammation in the brain. Additionally, the presence of neutrophil accumulation near fibrin(ogen) deposits was observed in ischemic lesions, and the engagement of fibrin(ogen) by integrin receptor αMβ2 promoted neutrophil activation and post-stroke inflammation. Finally, inhibiting fibrin(ogen)-mediated neutrophil activation using a fibrinogen-derived γ377-395peptide significantly attenuated neurological deficits. CONCLUSIONS Fibrin(ogen) is a crucial regulator of post-stroke inflammation and contributes to secondary brain injury. The inflammation induced by fibrin(ogen) is primarily driven by neutrophils during acute ischemic stroke and can be ameliorated using the fibrin-derived γ377-395peptide. Targeting the fibrin(ogen)-mediated neuropathological process represents a promising approach for neuroprotective therapy after stroke while preserving its beneficial coagulation function.
Collapse
Affiliation(s)
- Lu Han
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yaying Song
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Weiwei Xiang
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ze Wang
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yishu Wang
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiajun Zhou
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - De-Sheng Zhu
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of Neurology, Baoshan Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200444, China.
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
28
|
Tao C, Li Y, An N, Liu H, Liu Z, Sun Y, Qian Y, Li N, Xing Y, Gao Y. Pathological mechanisms and future therapeutic directions of thrombin in intracerebral hemorrhage: a systematic review. Front Pharmacol 2024; 15:1293428. [PMID: 38698822 PMCID: PMC11063263 DOI: 10.3389/fphar.2024.1293428] [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: 09/13/2023] [Accepted: 03/05/2024] [Indexed: 05/05/2024] Open
Abstract
Intracerebral hemorrhage (ICH), a common subtype of hemorrhagic stroke, often causes severe disability or death. ICH induces adverse events that might lead to secondary brain injury (SBI), and there is currently a lack of specific effective treatment strategies. To provide a new direction for SBI treatment post-ICH, the systematic review discussed how thrombin impacts secondary injury after ICH through several potentially deleterious or protective mechanisms. We included 39 studies and evaluated them using SYRCLE's ROB tool. Subsequently, we explored the potential molecular mechanisms of thrombin-mediated effects on SBI post-ICH in terms of inflammation, iron deposition, autophagy, and angiogenesis. Furthermore, we described the effects of thrombin in endothelial cells, astrocytes, pericytes, microglia, and neurons, as well as the harmful and beneficial effects of high and low thrombin concentrations on ICH. Finally, we concluded the current research status of thrombin therapy for ICH, which will provide a basis for the future clinical application of thrombin in the treatment of ICH.
Collapse
Affiliation(s)
- Chenxi Tao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haoqi Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Qian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Zhang X, Ma S, Huebner JL, Naz SI, Alnemer N, Soderblom EJ, Aliferis C, Kraus VB. Immune system-related plasma extracellular vesicles in healthy aging. Front Immunol 2024; 15:1355380. [PMID: 38633262 PMCID: PMC11021711 DOI: 10.3389/fimmu.2024.1355380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Objectives To identify age-related plasma extracellular vehicle (EVs) phenotypes in healthy adults. Methods EV proteomics by high-resolution mass spectrometry to evaluate EV protein stability and discover age-associated EV proteins (n=4 with 4 serial freeze-thaws each); validation by high-resolution flow cytometry and EV cytokine quantification by multiplex ELISA (n=28 healthy donors, aged 18-83 years); quantification of WI-38 fibroblast cell proliferation response to co-culture with PKH67-labeled young and old plasma EVs. The EV samples from these plasma specimens were previously characterized for bilayer structure, intra-vesicle mitochondria and cytokines, and hematopoietic cell-related surface markers. Results Compared with matched exo-EVs (EV-depleted supernatants), endo-EVs (EV-associated) had higher mean TNF-α and IL-27, lower mean IL-6, IL-11, IFN-γ, and IL-17A/F, and similar mean IL-1β, IL-21, and IL-22 concentrations. Some endo-EV and exo-EV cytokine concentrations were correlated, including TNF-α, IL-27, IL-6, IL-1β, and IFN-γ, but not IL-11, IL-17A/F, IL-21 or IL-22. Endo-EV IFN-γ and exo-EV IL-17A/F and IL-21 declined with age. By proteomics and confirmed by flow cytometry, we identified age-associated decline of fibrinogen (FGA, FGB and FGG) in EVs. Age-related EV proteins indicated predominant origins in the liver and innate immune system. WI-38 cells (>95%) internalized similar amounts of young and old plasma EVs, but cells that internalized PKH67-EVs, particularly young EVs, underwent significantly greater cell proliferation. Conclusion Endo-EV and exo-EV cytokines function as different biomarkers. The observed healthy aging EV phenotype reflected a downregulation of EV fibrinogen subpopulations consistent with the absence of a pro-coagulant and pro-inflammatory condition common with age-related disease.
Collapse
Affiliation(s)
- Xin Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Sisi Ma
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Janet L. Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Syeda Iffat Naz
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Noor Alnemer
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Erik J. Soderblom
- Duke Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Constantin Aliferis
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
30
|
Sulimai N, Brown J, Lominadze D. The Effect of Reduced Fibrinogen on Cerebrovascular Permeability during Traumatic Brain Injury in Fibrinogen Gene Heterozygous Knockout Mice. Biomolecules 2024; 14:385. [PMID: 38672403 PMCID: PMC11048347 DOI: 10.3390/biom14040385] [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: 02/09/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Vascular contribution to cognitive impairment and dementia (VCID) is a term referring to all types of cerebrovascular and cardiovascular disease-related cognitive decline, spanning many neuroinflammatory diseases including traumatic brain injury (TBI). This becomes particularly important during mild-to-moderate TBI (m-mTBI), which is characterized by short-term memory (STM) decline. Enhanced cerebrovascular permeability for proteins is typically observed during m-mTBI. We have previously shown that an increase in the blood content of fibrinogen (Fg) during m-mTBI results in enhanced cerebrovascular permeability. Primarily extravasated via a transcellular pathway, Fg can deposit into the parenchyma and exacerbate inflammatory reactions that can lead to neurodegeneration, resulting in cognitive impairment. In the current study, we investigated the effect of a chronic reduction in Fg concentration in blood on cerebrovascular permeability and the interactions of extravasated Fg with astrocytes and neurons. Cortical contusion injury (CCI) was used to generate m-mTBI in transgenic mice with a deleted Fg γ chain (Fg γ+/-), resulting in a low blood content of Fg, and in control C57BL/6J wild-type (WT) mice. Cerebrovascular permeability was tested in vivo. Interactions of Fg with astrocytes and neurons and the expression of neuronal nuclear factor-кB (NF-кB) were assessed via immunohistochemistry. The results showed that 14 days after CCI, there was less cerebrovascular permeability, lower extravascular deposition of Fg, less activation of astrocytes, less colocalization of Fg with neurons, and lower expression of neuronal pro-inflammatory NF-кB in Fg γ+/- mice compared to that found in WT mice. Combined, our data provide strong evidence that increased Fg extravasation, and its resultant extravascular deposition, triggers astrocyte activation and leads to potential interactions of Fg with neurons, resulting in the overexpression of neuronal NF-кB. These effects suggest that reduced blood levels of Fg can be beneficial in mitigating the STM reduction seen in m-mTBI.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
31
|
Wang HR, Ma J, Guo YZ, Liu KF, Han B, Wang MH, Zou FH, Wang J, Tian Z, Qu HQ, Huang XL, Liu F. Combination of Albumin/Fibrinogen Ratio and Admission Hunt-Hess Scale Score as an Independent Predictor of Clinical Outcome in Aneurysmal Subarachnoid Hemorrhage. World Neurosurg 2024; 181:e322-e329. [PMID: 37839575 DOI: 10.1016/j.wneu.2023.10.047] [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: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND The albumin/fibrinogen ratio (AFR) is an independent predictor of clinical outcomes of some diseases; however, the prognostic value of AFR and the admission Hunt-Hess (HH) score is still unclear for patients with an aneurysmal subarachnoid hemorrhage (aSAH). This study aimed to assess the relationship between the AFR-HH score and 6-month outcomes of aSAH patients. METHODS The clinical characteristics of aSAH patients admitted to our department between December 2017 and December 2021 were retrospectively analyzed. The candidate risk factors were screened using univariate regression analysis, and the independence of the resultant risk factors was evaluated by binary logistic regression analysis. The predictive value of the combined AFR and HH score for unfavorable outcomes was assessed using receiver operating characteristic curve analysis. RESULTS A total of 112 aSAH patients were included. Binary logistic regression analysis showed the perioperative period AFR, Glasgow coma scale score, and admission HH score were independent risk factors for unfavorable outcomes for aSAH patients. The receiver operating characteristic curve analysis showed the predictive capacity of AFR plus the admission HH score outperformed the AFR, Glasgow coma scale score, and admission HH scale alone and the combination of the AFR and Glasgow coma scale score. CONCLUSIONS A low AFR during the perioperative period is associated with unfavorable outcomes for aSAH patients at 6 months. The combination of the AFR and admission HH scale score provides superior predictive capacity to either the AFR or HH scale score alone.
Collapse
Affiliation(s)
- Hao Ran Wang
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jie Ma
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Yi Zhuo Guo
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ke Feng Liu
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Bin Han
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ming Hai Wang
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Fei Hui Zou
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jian Wang
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Zhen Tian
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - He Qi Qu
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Xian Long Huang
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Fang Liu
- Department of Neurosurgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China.
| |
Collapse
|
32
|
Zhao J, Zhang X, Li Y, Yu J, Chen Z, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Xia C, Xia J, Wu J. Interorgan communication with the liver: novel mechanisms and therapeutic targets. Front Immunol 2023; 14:1314123. [PMID: 38155961 PMCID: PMC10754533 DOI: 10.3389/fimmu.2023.1314123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
The liver is a multifunctional organ that plays crucial roles in numerous physiological processes, such as production of bile and proteins for blood plasma, regulation of blood levels of amino acids, processing of hemoglobin, clearance of metabolic waste, maintenance of glucose, etc. Therefore, the liver is essential for the homeostasis of organisms. With the development of research on the liver, there is growing concern about its effect on immune cells of innate and adaptive immunity. For example, the liver regulates the proliferation, differentiation, and effector functions of immune cells through various secreted proteins (also known as "hepatokines"). As a result, the liver is identified as an important regulator of the immune system. Furthermore, many diseases resulting from immune disorders are thought to be related to the dysfunction of the liver, including systemic lupus erythematosus, multiple sclerosis, and heart failure. Thus, the liver plays a role in remote immune regulation and is intricately linked with systemic immunity. This review provides a comprehensive overview of the liver remote regulation of the body's innate and adaptive immunity regarding to main areas: immune-related molecules secreted by the liver and the liver-resident cells. Additionally, we assessed the influence of the liver on various facets of systemic immune-related diseases, offering insights into the clinical application of target therapies for liver immune regulation, as well as future developmental trends.
Collapse
Affiliation(s)
- Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
33
|
O'Donnell JS, Fleming H, Noone D, Preston RJS. Unraveling coagulation factor-mediated cellular signaling. J Thromb Haemost 2023; 21:3342-3353. [PMID: 37391097 DOI: 10.1016/j.jtha.2023.06.019] [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: 03/02/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
Blood coagulation is initiated in response to blood vessel injury or proinflammatory stimuli, which activate coagulation factors to coordinate complex biochemical and cellular responses necessary for clot formation. In addition to these critical physiologic functions, plasma protein factors activated during coagulation mediate a spectrum of signaling responses via receptor-binding interactions on different cell types. In this review, we describe examples and mechanisms of coagulation factor signaling. We detail the molecular basis for cell signaling mediated by coagulation factor proteases via the protease-activated receptor family, considering new insights into the role of protease-specific cleavage sites, cofactor and coreceptor interactions, and distinct signaling intermediate interactions in shaping protease-activated receptor signaling diversity. Moreover, we discuss examples of how injury-dependent conformational activation of other coagulation proteins, such as fibrin(ogen) and von Willebrand factor, decrypts their signaling potential, unlocking their capacity to contribute to aberrant proinflammatory signaling. Finally, we consider the role of coagulation factor signaling in disease development and the status of pharmacologic approaches to either attenuate or enhance coagulation factor signaling for therapeutic benefit, emphasizing new approaches to inhibit deleterious coagulation factor signaling without impacting hemostatic activity.
Collapse
Affiliation(s)
- James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland, Crumlin, Dublin, Ireland. https://twitter.com/profJSOdonnell
| | - Harry Fleming
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland. https://www.twitter.com/PrestonLab_RCSI
| | - David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland. https://www.twitter.com/PrestonLab_RCSI
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland, Crumlin, Dublin, Ireland.
| |
Collapse
|
34
|
Wu PS, Liu HY, Wong TH, Lin JT, Hu FR, Lin MH. Comparative Proteomics Reveals Prolonged Corneal Preservation Impaired Ocular Surface Immunity Accompanied by Fibrosis in Human Stroma. J Proteome Res 2023; 22:3730-3741. [PMID: 37976471 DOI: 10.1021/acs.jproteome.3c00383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Cornea transplantation is one of the most commonly performed allotransplantations worldwide. Prolonged storage of donor corneas leads to decreased endothelial cell viability, severe stromal edema, and opacification, significantly compromising the success rate of corneal transplantation. Corneal stroma, which constitutes the majority of the cornea, plays a crucial role in maintaining its shape and transparency. In this study, we conducted proteomic analysis of corneal stroma preserved in Optisol-GS medium at 4 °C for 7 or 14 days to investigate molecular changes during storage. Among 1923 identified proteins, 1634 were quantifiable and 387 were significantly regulated with longer preservation. Compared to stroma preserved for 7 days, proteins involved in ocular surface immunomodulation were largely downregulated while proteins associated with extracellular matrix reorganization and fibrosis were upregulated in those preserved for 14 days. The increase in extracellular matrix structural proteins together with upregulation of growth factor signaling implies the occurrence of stromal fibrosis, which may compromise tissue clarity and cause vision impairments. This study is the first to provide insights into how storage duration affects corneal stroma from a proteomic perspective. Our findings may contribute to future research efforts aimed at developing long-term preservation techniques and improving the quality of preserved corneas, thus maximizing their clinical application.
Collapse
Affiliation(s)
- Pei-Shan Wu
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Hsin-Yu Liu
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Tzu-Hsuan Wong
- Department of Microbiology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Jui-Ti Lin
- Department of Microbiology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Fung-Rong Hu
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Miao-Hsia Lin
- Department of Microbiology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| |
Collapse
|
35
|
Hadjiagapiou MS, Krashias G, Christodoulou C, Pantzaris M, Lambrianides A. Serum Reactive Antibodies against the N-Methyl-D-Aspartate Receptor NR2 Subunit-Could They Act as Potential Biomarkers? Int J Mol Sci 2023; 24:16170. [PMID: 38003360 PMCID: PMC10671476 DOI: 10.3390/ijms242216170] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Synaptic dysfunction and disrupted communication between neuronal and glial cells play an essential role in the underlying mechanisms of multiple sclerosis (MS). Earlier studies have revealed the importance of glutamate receptors, particularly the N-methyl-D-aspartate (NMDA) receptor, in excitotoxicity, leading to abnormal synaptic transmission and damage of neurons. Our study aimed to determine whether antibodies to the NR2 subunit of NMDAR are detected in MS patients and evaluate the correlation between antibody presence and clinical outcome. Furthermore, our focus extended to examine a possible link between NR2 reactivity and anti-coagulant antibody levels as pro-inflammatory molecules associated with MS. A cross-sectional study was carried out, including 95 patients with MS and 61 age- and gender-matched healthy controls (HCs). The enzyme-linked immunosorbent assay was used to detect anti-NR2 antibodies in serum samples of participants along with IgG antibodies against factor (F)VIIa, thrombin, prothrombin, FXa, and plasmin. According to our results, significantly elevated levels of anti-NR2 antibodies were detected in MS patients compared to HCs (p < 0.05), and this holds true when we compared the Relapsing-Remitting MS course with HCs (p < 0.05). A monotonically increasing correlation was found between NR2 seropositivity and advanced disability (rs = 0.30; p < 0.01), anti-NR2 antibodies and disease worsening (rs = 0.24; p < 0.05), as well as between antibody activity against NR2 and thrombin (rs = 0.33; p < 0.01). The presence of anti-NR2 antibodies in MS patients was less associated with anti-plasmin IgG antibodies [OR:0.96 (95%CI: 0.92-0.99); p < 0.05]; however, such an association was not demonstrated when analyzing only RRMS patients. In view of our findings, NR2-reactive antibodies may play, paving the way for further research into their potential as biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Maria S. Hadjiagapiou
- Department of Neuroimmunology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (M.S.H.); (M.P.)
| | - George Krashias
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (G.K.); (C.C.)
| | - Christina Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (G.K.); (C.C.)
| | - Marios Pantzaris
- Department of Neuroimmunology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (M.S.H.); (M.P.)
| | - Anastasia Lambrianides
- Department of Neuroimmunology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (M.S.H.); (M.P.)
| |
Collapse
|
36
|
Liu Y, Xue M, Han Y, Li Y, Xiao B, Wang W, Yu J, Ye X. Exosomes from M2c macrophages alleviate intervertebral disc degeneration by promoting synthesis of the extracellular matrix via MiR-124/CILP/TGF-β. Bioeng Transl Med 2023; 8:e10500. [PMID: 38023721 PMCID: PMC10658595 DOI: 10.1002/btm2.10500] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Immuno-inflammation is highly associated with anabolic and catabolic dysregulation of the extracellular matrix (ECM) in the nucleus pulposus (NP), which dramatically propels intervertebral disc degeneration (IVDD). With the characteristics of tissue remodeling and regeneration, M2c macrophages have attracted great attention in research on immune modulation that rebuilds degenerated tissues. Therefore, we first demonstrated the facilitating effects of M2c macrophages on ECM anabolism of the NP in vitro. We subsequently found that exosomes from M2c macrophages (M2c-Exoss) mediated their metabolic rebalancing effects on the ECM. To determine whether M2c-Exoss served as positive agents protecting the ECM in IVDD, we constructed an M2c-Exos-loaded hyaluronic acid hydrogel (M2c-Exos@HA hydrogel) and implanted it into the degenerated caudal disc of rats. The results of MRI and histological staining indicated that the M2c-Exos@HA hydrogel alleviated IVDD in vivo in the long term. To elucidate the underlying molecular mechanism, we performed 4D label-free proteomics to screen dysregulated proteins in NPs treated with M2c-Exoss. Cartilage intermediate layer protein (CILP) was the key protein responsible for the rebalancing effects of M2c-Exoss on ECM metabolism in the NP. With prediction and verification using luciferase assays and rescue experiments, miR-124-3p was identified as the upstream regulator in M2c-Exoss that regulated CILP and consequently enhanced the activity of the TGF-β/smad3 pathway. In conclusion, we demonstrated ameliorating effects of M2c-Exoss on the imbalance of ECM metabolism in IVDD via the miR-124/CILP/TGF-β regulatory axis, which provides a promising theoretical basis for the application of M2c macrophages and their exosomes in the treatment of IVDD.
Collapse
Affiliation(s)
- Yi Liu
- Department of OrthopaedicsSecond Affiliated Hospital of Naval Medical UniversityShanghaiPeople's Republic of China
- Department of OrthopedicsTongren Hospital, Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Mintao Xue
- Department of OrthopaedicsSecond Affiliated Hospital of Naval Medical UniversityShanghaiPeople's Republic of China
| | - Yaguang Han
- Department of OrthopaedicsSecond Affiliated Hospital of Naval Medical UniversityShanghaiPeople's Republic of China
| | - Yucai Li
- Department of OrthopedicsTongren Hospital, Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Bing Xiao
- Department of OrthopaedicsSecond Affiliated Hospital of Naval Medical UniversityShanghaiPeople's Republic of China
| | - Weiheng Wang
- Department of OrthopaedicsSecond Affiliated Hospital of Naval Medical UniversityShanghaiPeople's Republic of China
| | - Jiangming Yu
- Department of OrthopedicsTongren Hospital, Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Xiaojian Ye
- Department of OrthopedicsTongren Hospital, Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| |
Collapse
|
37
|
Alruwaili M, Al-kuraishy HM, Alexiou A, Papadakis M, ALRashdi BM, Elhussieny O, Saad HM, Batiha GES. Pathogenic Role of Fibrinogen in the Neuropathology of Multiple Sclerosis: A Tale of Sorrows and Fears. Neurochem Res 2023; 48:3255-3269. [PMID: 37442896 PMCID: PMC10514123 DOI: 10.1007/s11064-023-03981-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease of the central nervous system (CNS) due to injury of the myelin sheath by immune cells. The clotting factor fibrinogen is involved in the pathogenesis of MS by triggering microglia and the progress of neuroinflammation. Fibrinogen level is correlated with MS severity; consequently, inhibition of the fibrinogen cascade may reduce MS neuropathology. Thus, this review aimed to clarify the potential role of fibrinogen in the pathogenesis of MS and how targeting of fibrinogen affects MS neuropathology. Accumulation of fibrinogen in the CNS may occur independently or due to disruption of blood-brain barrier (BBB) integrity in MS. Fibrinogen acts as transduction and increases microglia activation which induces the progression of inflammation, oxidative stress, and neuronal injury. Besides, brain fibrinogen impairs the remyelination process by inhibiting the differentiation of oligodendrocyte precursor cells. These findings proposed that fibrinogen is associated with MS neuropathology through interruption of BBB integrity, induction of neuroinflammation, and demyelination with inhibition of the remyelination process by suppressing oligodendrocytes. Therefore, targeting of fibrinogen and/or CD11b/CD18 receptors by metformin and statins might decrease MS neuropathology. In conclusion, inhibiting the expression of CD11b/CD18 receptors by metformin and statins may decrease the pro-inflammatory effect of fibrinogen on microglia which is involved in the progression of MS.
Collapse
Affiliation(s)
- Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany
| | - Barakat M. ALRashdi
- Biology Department, College of Science, Jouf University, Sakaka, 41412 Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Egypt
| |
Collapse
|
38
|
Li H, Ghorbani S, Ling CC, Yong VW, Xue M. The extracellular matrix as modifier of neuroinflammation and recovery in ischemic stroke and intracerebral hemorrhage. Neurobiol Dis 2023; 186:106282. [PMID: 37683956 DOI: 10.1016/j.nbd.2023.106282] [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: 05/28/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stroke is the second leading cause of death worldwide and has two major subtypes: ischemic stroke and hemorrhagic stroke. Neuroinflammation is a pathological hallmark of ischemic stroke and intracerebral hemorrhage (ICH), contributing to the extent of brain injury but also in its repair. Neuroinflammation is intricately linked to the extracellular matrix (ECM), which is profoundly altered after brain injury and in aging. In the early stages after ischemic stroke and ICH, immune cells are involved in the deposition and remodeling of the ECM thereby affecting processes such as blood-brain barrier and cellular integrity. ECM components regulate leukocyte infiltration into the central nervous system, activate a variety of immune cells, and induce the elevation of matrix metalloproteinases (MMPs) after stroke. In turn, excessive MMPs may degrade ECM into components that are pro-inflammatory and injurious. Conversely, in the later stages after stroke, several ECM molecules may contribute to tissue recovery. For example, thrombospondin-1 and biglycan may promote activity of regulatory T cells, inhibit the synthesis of proinflammatory cytokines, and aid regenerative processes. We highlight these roles of the ECM in ischemic stroke and ICH and discuss their potential cellular and molecular mechanisms. Finally, we discuss therapeutics that could be considered to normalize the ECM in stroke. Our goal is to spur research on the ECM in order to improve the prognosis of ischemic stroke and ICH.
Collapse
Affiliation(s)
- Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China; Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
39
|
Kohs TCL, Fallon ME, Oseas EC, Healy LD, Tucker EI, Gailani D, McCarty OJT, Vandenbark AA, Offner H, Verbout NG. Pharmacological targeting of coagulation factor XI attenuates experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 2023; 38:2383-2391. [PMID: 37341855 PMCID: PMC10530106 DOI: 10.1007/s11011-023-01251-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.
Collapse
Affiliation(s)
- Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA.
| | - Meghan E Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Ethan C Oseas
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Laura D Healy
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| | - David Gailani
- Department of Pathology and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
- Veterans Affairs Portland Health Care System, Portland, OR, USA
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Norah G Verbout
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| |
Collapse
|
40
|
Peng S, Lv K. The role of fibrinogen in traumatic brain injury: from molecular pathological mechanisms to clinical management. Eur J Trauma Emerg Surg 2023; 49:1665-1672. [PMID: 35972516 DOI: 10.1007/s00068-022-02084-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/08/2022] [Indexed: 11/28/2022]
Abstract
Fibrinogen is the substrate of plasma coagulation. It plays an important role in the formation of reticular network, which is crucial to the strength and stability of blood clots. In addition to directly participating in coagulation, fibrinogen also participates in the destruction of blood-brain barrier and neuroinflammation. This article reviews the pathophysiological changes of fibrinogen after traumatic brain injury. Considerable efforts have been made to understand the mechanisms by which fibrinogen damages the central nervous system. Combined with the latest research hotspots, potentially promising treatment strategies at the molecular level were discussed. We believe that understanding the role of fibrinogen-mediated damage in nerve and blood-brain barrier function will enable timely intervention in patients with nerve damage, and guide the development of novel targeted therapeutics.
Collapse
Affiliation(s)
- Shixin Peng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Ke Lv
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
41
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Platelets and hemostatic proteins are co-localized with chronic neuroinflammation surrounding implanted intracortical microelectrodes. Acta Biomater 2023; 166:278-290. [PMID: 37211307 PMCID: PMC10330779 DOI: 10.1016/j.actbio.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/23/2023]
Abstract
Intracortical microelectrodes induce vascular injury upon insertion into the cortex. As blood vessels rupture, blood proteins and blood-derived cells (including platelets) are introduced into the 'immune privileged' brain tissues at higher-than-normal levels, passing through the damaged blood-brain barrier. Blood proteins adhere to implant surfaces, increasing the likelihood of cellular recognition leading to activation of immune and inflammatory cells. Persistent neuroinflammation is a major contributing factor to declining microelectrode recording performance. We investigated the spatial and temporal relationship of blood proteins fibrinogen and von Willebrand Factor (vWF), platelets, and type IV collagen, in relation to glial scarring markers for microglia and astrocytes following implantation of non-functional multi-shank silicon microelectrode probes into rats. Together with type IV collagen, fibrinogen and vWF augment platelet recruitment, activation, and aggregation. Our main results indicate blood proteins participating in hemostasis (fibrinogen and vWF) persisted at the microelectrode interface for up to 8-weeks after implantation. Further, type IV collagen and platelets surrounded the probe interface with similar spatial and temporal trends as vWF and fibrinogen. In addition to prolonged blood-brain barrier instability, specific blood and extracellular matrix proteins may play a role in promoting the inflammatory activation of platelets and recruitment to the microelectrode interface. STATEMENT OF SIGNIFICANCE: Implanted microelectrodes have substantial potential for restoring function to people with paralysis and amputation by providing signals that feed into natural control algorithms that drive prosthetic devices. Unfortunately, these microelectrodes do not display robust performance over time. Persistent neuroinflammation is widely thought to be a primary contributor to the devices' progressive decline in performance. Our manuscript reports on the highly local and persistent accumulation of platelets and hemostatic blood proteins around the microelectrode interface of brain implants. To our knowledge neuroinflammation driven by cellular and non-cellular responses associated with hemostasis and coagulation has not been rigorously quantified elsewhere. Our findings identify potential targets for therapeutic intervention and a better understanding of the driving mechanisms to neuroinflammation in the brain.
Collapse
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
42
|
Wen T, Zhang Z. Cellular mechanisms of fibrin (ogen): insight from neurodegenerative diseases. Front Neurosci 2023; 17:1197094. [PMID: 37529232 PMCID: PMC10390316 DOI: 10.3389/fnins.2023.1197094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Neurodegenerative diseases are prevalent and currently incurable conditions that progressively impair cognitive, behavioral, and psychiatric functions of the central or peripheral nervous system. Fibrinogen, a macromolecular glycoprotein, plays a crucial role in the inflammatory response and tissue repair in the human body and interacts with various nervous system cells due to its unique molecular structure. Accumulating evidence suggests that fibrinogen deposits in the brains of patients with neurodegenerative diseases. By regulating pathophysiological mechanisms and signaling pathways, fibrinogen can exacerbate the neuro-pathological features of neurodegenerative diseases, while depletion of fibrinogen contributes to the amelioration of cognitive function impairment in patients. This review comprehensively summarizes the molecular mechanisms and biological functions of fibrinogen in central nervous system cells and neurodegenerative diseases, including Alzheimer's disease, Multiple Sclerosis, Parkinson's disease, Vascular dementia, Huntington's disease, and Amyotrophic Lateral Sclerosis. Additionally, we discuss the potential of fibrinogen-related treatments in the management of neurodegenerative disorders.
Collapse
|
43
|
Mendiola AS, Yan Z, Dixit K, Johnson JR, Bouhaddou M, Meyer-Franke A, Shin MG, Yong Y, Agrawal A, MacDonald E, Muthukumar G, Pearce C, Arun N, Cabriga B, Meza-Acevedo R, Alzamora MDPS, Zamvil SS, Pico AR, Ryu JK, Krogan NJ, Akassoglou K. Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nat Immunol 2023; 24:1173-1187. [PMID: 37291385 PMCID: PMC10307624 DOI: 10.1038/s41590-023-01522-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/24/2023] [Indexed: 06/10/2023]
Abstract
Blood protein extravasation through a disrupted blood-brain barrier and innate immune activation are hallmarks of neurological diseases and emerging therapeutic targets. However, how blood proteins polarize innate immune cells remains largely unknown. Here, we established an unbiased blood-innate immunity multiomic and genetic loss-of-function pipeline to define the transcriptome and global phosphoproteome of blood-induced innate immune polarization and its role in microglia neurotoxicity. Blood induced widespread microglial transcriptional changes, including changes involving oxidative stress and neurodegenerative genes. Comparative functional multiomics showed that blood proteins induce distinct receptor-mediated transcriptional programs in microglia and macrophages, such as redox, type I interferon and lymphocyte recruitment. Deletion of the blood coagulation factor fibrinogen largely reversed blood-induced microglia neurodegenerative signatures. Genetic elimination of the fibrinogen-binding motif to CD11b in Alzheimer's disease mice reduced microglial lipid metabolism and neurodegenerative signatures that were shared with autoimmune-driven neuroinflammation in multiple sclerosis mice. Our data provide an interactive resource for investigation of the immunology of blood proteins that could support therapeutic targeting of microglia activation by immune and vascular signals.
Collapse
Affiliation(s)
- Andrew S Mendiola
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Zhaoqi Yan
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Karuna Dixit
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | | | | | - Yu Yong
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Eilidh MacDonald
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Clairice Pearce
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Nikhita Arun
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Belinda Cabriga
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Rosa Meza-Acevedo
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | | | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA.
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
44
|
Pansieri J, Hadley G, Lockhart A, Pisa M, DeLuca GC. Regional contribution of vascular dysfunction in white matter dementia: clinical and neuropathological insights. Front Neurol 2023; 14:1199491. [PMID: 37396778 PMCID: PMC10313211 DOI: 10.3389/fneur.2023.1199491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
The maintenance of adequate blood supply and vascular integrity is fundamental to ensure cerebral function. A wide range of studies report vascular dysfunction in white matter dementias, a group of cerebral disorders characterized by substantial white matter damage in the brain leading to cognitive impairment. Despite recent advances in imaging, the contribution of vascular-specific regional alterations in white matter dementia has been not extensively reviewed. First, we present an overview of the main components of the vascular system involved in the maintenance of brain function, modulation of cerebral blood flow and integrity of the blood-brain barrier in the healthy brain and during aging. Second, we review the regional contribution of cerebral blood flow and blood-brain barrier disturbances in the pathogenesis of three distinct conditions: the archetypal white matter predominant neurocognitive dementia that is vascular dementia, a neuroinflammatory predominant disease (multiple sclerosis) and a neurodegenerative predominant disease (Alzheimer's). Finally, we then examine the shared landscape of vascular dysfunction in white matter dementia. By emphasizing the involvement of vascular dysfunction in the white matter, we put forward a hypothetical map of vascular dysfunction during disease-specific progression to guide future research aimed to improve diagnostics and facilitate the development of tailored therapies.
Collapse
|
45
|
Gu X, Chen A, You M, Guo H, Tan S, He Q, Hu B. Extracellular vesicles: a new communication paradigm of complement in neurological diseases. Brain Res Bull 2023; 199:110667. [PMID: 37192717 DOI: 10.1016/j.brainresbull.2023.110667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/25/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
The complement system is crucial to the innate immune system. It has the function of destroying pathogens by activating the classical, alternative, and lectin pathways. The complement system is important in nervous system diseases such as cerebrovascular and neurodegenerative diseases. Activation of the complement system involves a series of intercellular signaling and cascade reactions. However, research on the source and transport mechanisms of the complement system in neurological diseases is still in its infancy. Studies have increasingly found that extracellular vesicles (EVs), a classic intercellular communication paradigm, may play a role in complement signaling disorders. Here, we systematically review the EV-mediated activation of complement pathways in different neurological diseases. We also discuss the prospect of EVs as future immunotherapy targets.
Collapse
Affiliation(s)
- Xinmei Gu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Anqi Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Hongxiu Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Senwei Tan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022.
| |
Collapse
|
46
|
Li Y, Wang S, Liu P, Ma J, Liu X, Yuan J. Clinical features of patients with MOG-IgG associated disorders and analysis of the relationship between fibrinogen-to-albumin ratio and the severity at disease onset. Front Neurol 2023; 14:1140917. [PMID: 37153679 PMCID: PMC10157091 DOI: 10.3389/fneur.2023.1140917] [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: 01/09/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
OBJECTIVE The study aimed to investigate the differences in clinical features between pediatric and adult patients with first-episode MOG-IgG associated disorders (MOGAD) and evaluate the relationship between the fibrinogen-to-albumin ratio (FAR) and the severity of neurological deficits at disease onset. METHODS We retrospectively collected and analyzed biochemical test results, imaging characteristics, clinical manifestations, expanded disability status scale (EDSS) score, and FAR. The Spearman correlation analysis and logistic regression models were used to examine the association between FAR and severity. Receiver operating characteristic (ROC) curve analysis was to analyze the predictive ability of FAR for the severity of neurological deficits. RESULTS Fever (50.0%), headache (36.1%), and blurred vision (27.8%) were the most common clinical manifestations in the pediatric group (<18 years old). However, in the adult group (≥18 years old), the most common symptoms were blurred vision (45.7%), paralysis (37.0%), and paresthesia (32.6%). Fever was more common in the pediatric group, while paresthesia was more common in the adult patients, with all differences statistically significant (P < 0.05). The most frequent clinical phenotype in the pediatric group was acute disseminated encephalomyelitis (ADEM; 41.7%), whereas optic neuritis (ON; 32.6%) and transverse myelitis (TM; 26.1%) were more common in the adult group. The differences in clinical phenotype between the two groups were statistically significant (P < 0.05). In both pediatric and adult patients, cortical/subcortical and brainstem lesions were the most common lesions on cranial magnetic resonance imaging (MRI), whereas, for spinal MRI, cervical and thoracic spinal cord lesions were the most commonly observed. According to binary logistic regression analysis, FAR was an independent risk factor for the severity of neurological deficits (odds ratio = 1.717; 95% confidence interval = 1.191-2.477; P = 0.004). FAR (r = 0.359, P = 0.001) was positively correlated with the initial EDSS score. The area under the ROC curve was 0.749. CONCLUSION The current study found age-dependent phenotypes in MOGAD patients as ADEM was more commonly observed in patients < 18 years old, while ON and TM were more frequently found in patients ≥18 years old. A high FAR level was an independent indicator for more severe neurological deficits at disease onset in patients with a first episode of MOGAD.
Collapse
Affiliation(s)
| | | | | | | | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yuan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:648. [PMID: 37189395 PMCID: PMC10136045 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
48
|
Wang X, Jia Z, Zhou X, Su L, Wang M, Wang T, Zhang H. Nanoplastic-induced vascular endothelial injury and coagulation dysfunction in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 865:161271. [PMID: 36587662 DOI: 10.1016/j.scitotenv.2022.161271] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/21/2022] [Accepted: 12/25/2022] [Indexed: 06/17/2023]
Abstract
Nanoplastics are the persistent pollutants in a variety of environments, representing a potential threat to human health. Notably, plastic particles have been detected in sample of human bloodstream. It is thus significant to investigate the effects of nanoplastics on the cardiovascular system owing to its ease transfer through the bloodstream to other organs. However, few studies have been performed to evaluate the cardiovascular toxicity of nanoplastics. Herein, we pursued to investigate the adverse cardiovascular impacts of polystyrene (PS), PS-NH2 and PS-COOH nanoplastics on mice. Experimental results demonstrated that the exposure to these nanoplastics could result in structural damage of vascular endothelial cells and inflammatory response. Moreover, it was found out that the dysfunctions of coagulation and prethrombotic state were caused by nanoplastics, which could be ascribed to the activation of JAK1/STAT3/TF signaling pathway. In summary, results clearly indicated that nanoplastic exposure lead to vascular toxicity to mice, which serves as a basis for future studies about the potential physiological threat of nanoplastics to humans.
Collapse
Affiliation(s)
- Xiuxiu Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Xiuran Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Lei Su
- Department of Food Industry, Shandong Institute of Commerce and Technology, Jinan 250103, PR China
| | - Minglu Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Tian Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China.
| | - Hongyan Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan 250014, PR China.
| |
Collapse
|
49
|
Hu X, Geng P, Zhao X, Wang Q, Liu C, Guo C, Dong W, Jin X. The NG2-glia is a potential target to maintain the integrity of neurovascular unit after acute ischemic stroke. Neurobiol Dis 2023; 180:106076. [PMID: 36921779 DOI: 10.1016/j.nbd.2023.106076] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The neurovascular unit (NVU) plays a critical role in health and disease. In the current review, we discuss the critical role of a class of neural/glial antigen 2 (NG2)-expressing glial cells (NG2-glia) in regulating NVU after acute ischemic stroke (AIS). We first introduce the role of NG2-glia in the formation of NVU during development as well as aging-induced damage to NVU and accompanying NG2-glia change. We then discuss the reciprocal interactions between NG2-glia and the other component cells of NVU, emphasizing the factors that could influence NG2-glia. Damage to the NVU integrity is the pathological basis of edema and hemorrhagic transformation, the most dreaded complication after AIS. The role of NG2-glia in AIS-induced NVU damage and the effect of NG2-glia transplantation on AIS-induced NVU damage are summarized. We next discuss the role of NG2-glia and the effect of NG2-glia transplantation in oligodendrogenesis and white matter repair as well as angiogenesis which is associated with the outcome of the patients after AIS. Finally, we review the current strategies to promote NG2-glia proliferation and differentiation and propose to use the dental pulp stem cells (DPSC)-derived exosome as a promising strategy to reduce AIS-induced injury and promote repair through maintaining the integrity of NVU by regulating endogenous NG2-glia proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xiaoyun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Changqing Liu
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Wen Dong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
50
|
Berk-Rauch HE, Choudhury A, Richards AT, Singh PK, Chen ZL, Norris EH, Strickland S, Ahn HJ. Striatal fibrinogen extravasation and vascular degeneration correlate with motor dysfunction in an aging mouse model of Alzheimer’s disease. Front Aging Neurosci 2023; 15:1064178. [PMID: 36967821 PMCID: PMC10034037 DOI: 10.3389/fnagi.2023.1064178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Alzheimer’s Disease (AD) patients exhibit signs of motor dysfunction, including gait, locomotion, and balance deficits. Changes in motor function often precede other symptoms of AD as well as correlate with increased severity and mortality. Despite the frequent occurrence of motor dysfunction in AD patients, little is known about the mechanisms by which this behavior is altered.Methods and Results: In the present study, we investigated the relationship between cerebrovascular impairment and motor dysfunction in a mouse model of AD (Tg6799). We found an age-dependent increase of extravasated fibrinogen deposits in the cortex and striatum of AD mice. Interestingly, there was significantly decreased cerebrovascular density in the striatum of the 15-month-old as compared to 7-month-old AD mice. We also found significant demyelination and axonal damage in the striatum of aged AD mice. We analyzed striatum-related motor function and anxiety levels of AD mice at both ages and found that aged AD mice exhibited significant impairment of motor function but not in the younger AD mice.Discussion: Our finding suggests an enticing correlation between extravasated fibrinogen, cerebrovascular damage of the striatum, and motor dysfunction in an AD mouse model, suggesting a possible mechanism underlying motor dysfunction in AD.
Collapse
Affiliation(s)
- Hanna E. Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Allison T. Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Hyung Jin Ahn,
| |
Collapse
|