1
|
Parmaksiz D, Kim Y. Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories. Neuroscientist 2025; 31:234-261. [PMID: 39113465 PMCID: PMC12103645 DOI: 10.1177/10738584241268754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Complex mechanisms govern the transport and action of oxytocin (Oxt), a neuropeptide and hormone that mediates diverse physiologic processes. While Oxt exerts site-specific and rapid effects in the brain via axonal and somatodendritic release, volume transmission via CSF and the neurovascular interface can act as an additional mechanism to distribute Oxt signals across distant brain regions on a slower timescale. This review focuses on modes of Oxt transport and action in the CNS, with particular emphasis on the roles of perivascular spaces, the blood-brain barrier (BBB), and circumventricular organs in coordinating the triadic interaction among circulating blood, CSF, and parenchyma. Perivascular spaces, critical conduits for CSF flow, play a pivotal role in Oxt diffusion and distribution within the CNS and reciprocally undergo Oxt-mediated structural and functional reconstruction. While the BBB modulates the movement of Oxt between systemic and cerebral circulation in a majority of brain regions, circumventricular organs without a functional BBB can allow for diffusion, monitoring, and feedback regulation of bloodborne peripheral signals such as Oxt. Recognition of these additional transport mechanisms provides enhanced insight into the systemic propagation and regulation of Oxt activity.
Collapse
Affiliation(s)
- Deniz Parmaksiz
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
2
|
Rocca MA, Preziosa P, Filippi M. Advances in neuroimaging of multiple sclerosis. Curr Opin Neurol 2025; 38:205-216. [PMID: 40104925 DOI: 10.1097/wco.0000000000001360] [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: 03/20/2025]
Abstract
PURPOSE OF REVIEW To summarize recent advancements in understanding multiple sclerosis (MS) pathophysiology, predicting disease course, and monitoring treatment responses using MRI. RECENT FINDINGS Paramagnetic rim lesions (PRLs) are highly specific to MS and clinically relevant. Detected from the earliest disease phases, PRLs aid in distinguishing MS from other conditions, improving diagnostic accuracy. Moreover, PRLs are associated with more severe disability and measures of brain damage and may predict disease progression. Similarly, slowly expanding lesions (SELs) are associated with more severe disability and predict a more severe disease course. Disease-modifying therapies have limited effectiveness in reducing PRLs or SELs. Choroid plexus (CP) enlargement is associated with structural brain damage and clinical disability and predicts disease evolution. Enlarged perivascular spaces (ePVS) suggest microangiopathic changes rather than direct MS-related inflammation. Glymphatic dysfunction, evaluated using diffusion tensor image analysis along the perivascular space, emerges early in MS and correlates with disability, cognitive impairment, and structural brain damage. Aging and comorbidities exacerbate MS-related damage, complicating diagnosis and treatment. Emerging technologies, such as brain-age paradigms, aim to disentangle aging from MS-specific neurodegeneration. SUMMARY Advances in MRI have highlighted the clinical significance of chronic inflammation and glymphatic dysfunction as early contributors to MS progression as well as the interplay between aging, comorbidities and MS.
Collapse
Affiliation(s)
- Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience
- Neurology Unit, IRCCS San Raffaele Scientific Institute
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience
- Neurology Unit, IRCCS San Raffaele Scientific Institute
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience
- Neurology Unit, IRCCS San Raffaele Scientific Institute
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
3
|
Strube KA. Traumatic brain injury graphing: A case study of Charles Whitman. J Forensic Sci 2025. [PMID: 40369769 DOI: 10.1111/1556-4029.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/28/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Research has identified violent behavior (i.e., assault, murder, and suicide) as a possible sequela of moderate to severe traumatic brain injury (TBI) and repeated mild TBI (rmTBI). However, misconceptions about consciousness and its ability to control an injured brain, the diverse spectrum of potential outcomes, and the role genetics/environmental factors can play make proving TBI's influence on inciting violent behavior by a perpetrator extremely difficult. Though the cause and execution may be complex, multifaceted, and vary in each individual, violent behavior is often preceded and accompanied by other emotional, behavioral, cognitive, and/or physical consequences. This research examines the benefits of graphing, using data interpreted from in-depth, color-coded reverse chronology, as a visualization tool for analyzing/displaying the potential impacts of TBI injuries and aggravators on violent outcomes, especially in complex situations. The case of Charles J. Whitman (herein referred to as CW) was used to demonstrate this method. Results revealed that sequelae clusters appeared shortly after suspected TBI injuries, sequelae changed and progressed over time, and numerous TBI aggravators were present at the time of the tragedy, including: an extended period of excessively high ambient temperatures, sleep deprivation (SD), drug use, blood loss, and emotional stressors. TBI graphing (utilizing reverse chronology) served as a valuable tool for observing the potential TBI sequelae progression prior to the violent event.
Collapse
Affiliation(s)
- K A Strube
- Independent Researcher, Bay City, Michigan, USA
| |
Collapse
|
4
|
Song KW, Lim M, Monje M. Complex neural-immune interactions shape glioma immunotherapy. Immunity 2025; 58:1140-1160. [PMID: 40324379 DOI: 10.1016/j.immuni.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Rich neural-immune interactions in the central nervous system (CNS) shape its function and create a unique immunological microenvironment for immunotherapy in CNS malignancies. Far from the now-debunked concept of CNS "immune privilege," it is now understood that unique immunological niches and constant immune surveillance of the brain contribute in multifaceted ways to brain health and robustly influence immunotherapy approaches for CNS cancers. Challenges include immune-suppressive and neurotoxicity-promoting crosstalk between brain, immune, and tumor cells. Developing effective immunotherapies for cancers of the nervous system will require a deeper understanding of these neural-immune-malignant cell interactions. Here, we review progress and challenges in immunotherapy for gliomas of the brain and spinal cord in light of these unique neural-immune interactions and highlight future work needed to optimize promising immunotherapies for gliomas.
Collapse
Affiliation(s)
- Kun-Wei Song
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
5
|
Vara-Pérez M, Movahedi K. Border-associated macrophages as gatekeepers of brain homeostasis and immunity. Immunity 2025; 58:1085-1100. [PMID: 40324381 PMCID: PMC12094687 DOI: 10.1016/j.immuni.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 05/07/2025]
Abstract
The brain's border tissues serve as essential hubs for neuroimmune regulation and the trafficking of biomaterials to and from the brain. These complex tissues-including the meninges, perivascular spaces, choroid plexus, and circumventricular organs-balance the brain's need for immune privilege with immune surveillance and blood-brain communication. Macrophages are integral components of these tissues, taking up key strategic positions within the brain's circulatory system. These border-associated macrophages, or "BAMs," are therefore emerging as pivotal for brain homeostasis and disease. BAMs perform trophic functions that help to support border homeostasis but also act as immune sentinels essential for border defense. In this review, we integrate recent findings on BAM origins, cell states, and functions, aiming to provide global insights and perspectives on the complex relationship between these macrophages and their border niche.
Collapse
Affiliation(s)
- Mónica Vara-Pérez
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
6
|
Hong H, Tozer DJ, Chen Y, Brown RB, Low A, Markus HS. Perivascular space dysfunction in cerebral small vessel disease is related to neuroinflammation. Brain 2025; 148:1540-1550. [PMID: 39509331 PMCID: PMC12073995 DOI: 10.1093/brain/awae357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/09/2024] [Accepted: 10/13/2024] [Indexed: 11/15/2024] Open
Abstract
Enlarged perivascular spaces are a feature of cerebral small vessel disease, and it has been hypothesized that they might reflect impaired glymphatic drainage. The mechanisms underlying enlargement of perivascular spaces are not fully understood, but both increased inflammation and blood-brain barrier (BBB) permeability have been hypothesized to play a role. We investigated the relationship between perivascular spaces and both CNS and peripheral inflammation, in addition to BBB permeability, in cerebral small vessel disease. Fifty-four symptomatic sporadic cerebral small vessel disease patients were studied. Perivascular spaces were quantified both using a visual rating scale and by measurement of the volume of perivascular spaces in the white matter and the basal ganglia. PET-MRI was used to measure microglial activation using the radioligand 11C-PK11195, and simultaneously, BBB permeability was acquired using dynamic contrast-enhanced MRI. We determined 11C-PK11195 binding and BBB permeability in the local vicinity of individual perivascular spaces in concentric shells surrounding the perivascular spaces. In addition, both mean 11C-PK11195 binding and BBB permeability in both the white matter and the basal ganglia were determined. To assess systemic inflammation, a panel of 93 blood biomarkers relating to cardiovascular disease, inflammation and endothelial activation were measured. Within the white matter, tissue in closest proximity to perivascular spaces displayed greater 11C-PK11195 binding (P < 0.001) in the vicinity of perivascular spaces. Higher white matter perivascular spaces burden on the visual rating scale was associated with higher white matter 11C-PK11195 binding (ρ = 0.469, false discovery rate-corrected P = 0.009); values for the volume of perivascular spaces showed a similar trend. In contrast, there were no associations between the burden of basal ganglia perivascular spaces and 11C-PK11195 binding. No marker of perivascular spaces was correlated with blood-brain barrier permeability. There was no association between markers of perivascular spaces and blood biomarkers of systemic inflammation. Our findings demonstrate that white matter perivascular spaces are associated with increased 11C-PK11195 binding, consistent with neuroinflammation playing a role in enlargement of white matter perivascular spaces. Further longitudinal and intervention studies are required to determine whether the relationship between neuroinflammation and enlarged perivascular spaces is causal.
Collapse
Affiliation(s)
- Hui Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Radiology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Daniel J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Yutong Chen
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Robin B Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Audrey Low
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
7
|
Soulé C, Blond L, Cavalerie R, Piazza S, Baudin‐Tréhiou C. Evolution of Brain Magnetic Resonance Imaging Lesions in Dogs Treated for Meningoencephalomyelitis of Unknown Origin Between Initial Diagnosis and Relapse. J Vet Intern Med 2025; 39:e70086. [PMID: 40213932 PMCID: PMC11986682 DOI: 10.1111/jvim.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The response of meningoencephalitis of unknown origin (MUO) in dogs to immunosuppressive treatment is unpredictable, and relapses frequently occur. OBJECTIVES Our aim was to describe the evolution of brain magnetic resonance imaging (MRI) lesions in dogs treated for MUO from diagnosis to relapse and to define the diagnostic and clinical value of repeat MRI at relapse. ANIMALS Eighteen dogs treated for MUO that experienced relapse and underwent MRI both at disease onset and relapse. METHODS Retrospective, descriptive, longitudinal, case series study. Dogs were identified from medical records between 2015 and 2024. The MR images were reviewed by radiologists for lesion number, location, size, pre- and post-contrast signal aspect, meningeal enhancement, mass effect, perilesional edema, and evidence of intracranial hypertension. RESULTS Median interval between MRIs was 259 days (range, 31-876 days). In dogs with relapse delay < 157 days, lesion number tended to decrease. Residual lesions tended to enlarge and exhibit contrast enhancement and perilesional edema (suggesting an active pathologic process), but without development of new lesions. After 233 days, all dogs had developed new lesions. Half exhibited enlarged active residual lesions, whereas the others showed either remission or smaller inactive lesions. CONCLUSIONS Before a clinical relapse at approximately 6 months, remission of the initial pathologic process and development of new lesions appear unlikely. Beyond this period, new lesions may occur with or without remission of the initial pathologic process, and repeat MRI is of high diagnostic and clinical value in detecting new lesions and characterizing the underlying pathologic process.
Collapse
Affiliation(s)
- Carole Soulé
- Department of Diagnostic ImagingCentre Hospitalier Vétérinaire LanguedociaMontpellierFrance
| | - Laurent Blond
- Department of Diagnostic ImagingCentre Hospitalier Vétérinaire LanguedociaMontpellierFrance
| | - Robin Cavalerie
- Department of NeurologyCentre Hospitalier Vétérinaire LanguedociaMontpellierFrance
| | - Stéphanie Piazza
- Department of NeurologyCentre Hospitalier Vétérinaire LanguedociaMontpellierFrance
| | - Clément Baudin‐Tréhiou
- Department of Diagnostic ImagingCentre Hospitalier Vétérinaire LanguedociaMontpellierFrance
| |
Collapse
|
8
|
Lu Y, Cheng L, Xiong Y, Huang C, Liu Z, Shen C, Wang H, Qiu Y, Yang SB, Wu M, Zhang X. NLRP3 Inflammasome in Vascular Dementia: Regulatory Mechanisms, Functions, and Therapeutic Implications: A Comprehensive Review. CNS Neurosci Ther 2025; 31:e70403. [PMID: 40326096 PMCID: PMC12052953 DOI: 10.1111/cns.70403] [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: 01/20/2025] [Revised: 03/20/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Vascular dementia, the second most common type of dementia globally after Alzheimer's disease, is associated with neuroinflammation. Activation of the NLRP3 inflammasome, an important pattern recognition receptor in human innate immunity, plays a key role in the pathogenesis of vascular dementia. RESULTS The NLRP3 inflammasome pathway destroys neuronal cells primarily through the production of IL-18 and IL-1β. Moreover, it exacerbates vascular dementia by producing IL-18, IL-1β, and the N-terminal fragment of GSDMD, which also contributes to neuronal cell death. Thus, blocking the NLRP3 inflammasome pathway presents a new therapeutic strategy for treating vascular dementia, thereby delaying or curing the disease more effectively and mitigating adverse effects. CONCLUSIONS This review explores the role and mechanisms of the NLRP3 inflammasome in vascular dementia, summarizing current research and therapeutic strategies. Investigating the activation of the NLRP3 inflammasome can reveal the pathogenesis of vascular dementia from a new perspective and propose innovative preventive and treatment strategies.
Collapse
Affiliation(s)
- Yujia Lu
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Lin Cheng
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of RehabilitationClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
| | - Chunyan Huang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Ziying Liu
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Chunxiao Shen
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Huaying Wang
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Yuemin Qiu
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Seung Bum Yang
- Department of ParamedicineWonkwang Health Science UniversityIksanRepublic of Korea
| | - Moxin Wu
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Xiaorong Zhang
- Department of PathologyClinical Medical School of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
9
|
Li L, Wu Y, Wu J, Li B, Hua R, Shi F, Chen L, Wu Y. MRI-based machine learning analysis of perivascular spaces and their link to sleep disturbances, dementia, and mental distress in young adults with long-time mobile phone use. Front Neurosci 2025; 19:1555054. [PMID: 40356706 PMCID: PMC12066657 DOI: 10.3389/fnins.2025.1555054] [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: 01/03/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
Objective Long-term mobile phone use (LTMPU) has been linked to sleep disorders, mood disorders, and cognitive impairment, with MRI-detected enlarged perivascular spaces (EPVSs) as potential imaging markers. This study investigated computational MRI-visible EPVSs and their association with sleep disturbance, dementia, and mental distress in young adults with LTMPU. Methods This retrospective study included 82 LTMPU patients who underwent MRI scans and assessments using six clinical scales: Montreal Cognitive Assessment (MoCA), Pittsburgh Sleep Quality Index (PSQI), Insomnia Severity Index (ISI), Epworth Sleepiness Scale (ESS), Hamilton Anxiety (HAM-A), and Hamilton Depression (HAM-D). Deep learning algorithms segmented EPVSs lesions, extracting quantitative metrics (count, volume, mean length, and mean curvature) across 17 brain subregions. Correlation analyses explored relationships between EPVSs indicators and clinical measurements. The BrainNet Viewer tool highlighted significant brain subregions and EPVSs traits linked to dementia, sleep disturbance, and mental distress. Results Correlation analyses identified 23 significant indicator pairs (FDR-adjusted p < 0.05), including associations between nine EPVSs characteristics and MoCA scores: four with the PSQI, one with the ISI, three with the ESS, four with the HAM-A, and two with the HAM-D. Regression analyses revealed seven significant EPVSs features, with three linked to cognitive impairment: mean EPVSs length in the left basal ganglia and mean length/curvature in the left temporal lobe. Also, the mean EPVSs length in the left frontal lobe could indicate insomnia, sleepiness, and anxiety. Conclusion Computational EPVSs metrics offer insights into the EPVSs pathophysiology and its links to mood disorders, sleep disturbances, and cognitive impairment in LTMPU patients. These findings also highlight potential connections between EPVSs, excessive daytime sleepiness, and anxiety, contributing to a comprehensive understanding of these multifaceted conditions.
Collapse
Affiliation(s)
- Li Li
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yalan Wu
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jiaojiao Wu
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Bin Li
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Rui Hua
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Feng Shi
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Lizhou Chen
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yeke Wu
- Department of Stomatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Jasim MH, Saadoon Abbood R, Sanghvi G, Roopashree R, Uthirapathy S, Kashyap A, Sabarivani A, Ray S, Mustafa YF, Yasin HA. Flavonoids in the regulation of microglial-mediated neuroinflammation; focus on fisetin, rutin, and quercetin. Exp Cell Res 2025; 447:114537. [PMID: 40147710 DOI: 10.1016/j.yexcr.2025.114537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Neuroinflammation is a critical mechanism in central nervous system (CNS) inflammatory disorders, encompassing conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), traumatic brain injury (TBI), encephalitis, spinal cord injury (SCI), and cerebral stroke. Neuroinflammation is characterized by increased blood vessel permeability, leukocyte infiltration, glial cell activation, and elevated production of inflammatory mediators, such as chemokines and cytokines. Microglia act as the resident macrophages of the central nervous system, serving as the principal defense mechanism in brain tissue. After CNS injury, microglia modify their morphology and downregulate genes that promote homeostatic functions. Despite comprehensive transcriptome analyses revealing specific gene modifications in "pathological" microglia, microglia's precise protective or harmful functions in neurological disorders remain insufficiently comprehended. Accumulating data suggests that the polarization of microglia into the M1 proinflammatory phenotype or the M2 antiinflammatory phenotype may serve as a sensible therapeutic strategy for neuroinflammation. Flavonoids, including rutin, fisetin, and quercetin, function as crucial chemical reservoirs with unique structures and diverse actions and are extensively used to modulate microglial polarization in treating neuroinflammation. This paper highlights the detrimental effects of neuroinflammation seen in neurological disorders such as stroke. Furthermore, we investigate their therapeutic benefits in alleviating neuroinflammation via the modulation of macrophage polarization.
Collapse
Affiliation(s)
- Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq.
| | - Rosull Saadoon Abbood
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India.
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq.
| |
Collapse
|
11
|
Mahmud SZ, Heo HY. When CEST meets diffusion: Multi-echo diffusion-encoded CEST (dCEST) MRI to measure intracellular and extracellular CEST signal distributions. Magn Reson Med 2025. [PMID: 40228073 DOI: 10.1002/mrm.30530] [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: 11/21/2024] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE To develop a multi-echo, diffusion-encoded chemical exchange saturation transfer (dCEST) imaging technique for estimating the intracellular and extracellular/intravascular contributions to the conventional CEST signal. METHODS A dCEST pulse sequence was developed to quantify the signal fractions, transverse relaxation times (T2), and apparent diffusion coefficient (ADC) of the intracellular and extracellular/intravascular water compartments. dCEST images were acquired across a wide range of TE, b-values, RF saturation strengths, and frequency offsets. The data were analyzed using a two-compartment model with distinct diffusivities and T2 values. Intracellular and extracellular fractions of conventional water-saturation spectra (Z-spectra) and corresponding amide proton transfer (APT) signals were estimated from human brain scans of healthy volunteers at 3 T. RESULTS The multi-echo diffusion results showed that the intracellular water fractions were significantly higher than the extracellular water fractions, whereas the intracellular T2 values were shorter than those of the extracellular/intravascular compartments. The ADC for the intracellular compartment was significantly lower than that of the extracellular compartment. The dCEST analysis showed that the average intracellular and extracellular fractions of the Z-spectra were 85 ± 7% and 15 ± 4%, respectively. The overall intracellular APT-weighted values were higher than the total (i.e., intracellular + extracellular) APT-weighted values. CONCLUSIONS The dCEST imaging technique provides valuable insight into the source of signals in conventional CEST MRI, offering potential utility for clinical applications.
Collapse
Affiliation(s)
- Sultan Z Mahmud
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Hye-Young Heo
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Guo C, Li W, Liu Y, Mahaman Yacoubou AR, Wang J, Liu R, Li S, Wang X. LCN2 induces neuronal loss and facilitates sepsis-associated cognitive impairments. Cell Death Dis 2025; 16:146. [PMID: 40025014 PMCID: PMC11873032 DOI: 10.1038/s41419-025-07469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe neurological syndrome marked by widespread brain dysfunctions due to sepsis. Despite increasing data supporting the hypothesis of neuronal damage, the exact mechanism of sepsis-related cognitive disorders and therapeutic strategies remain unclear and need further investigation. In this study, a sepsis model was established in C57 mice using lipopolysaccharide (LPS). The findings demonstrated that LPS exposure induced neuronal loss, synaptic and cognitive deficits accompanied by mitochondrial damage. Bioinformatics and western blot analyses demonstrated a significant increase in Lipocalin-2 (LCN2) during sepsis as a key hub gene involved in immune and neurological inflammation. Interestingly, the recombinant LCN2 protein exhibited similar effects on synaptic dysfunction and cognitive deficits in C57 mice. Conversely, downregulating LCN2 effectively nullified the impact of LPS, leading to the amelioration of synaptic and cognitive deficits, neuronal loss, and reactive oxygen species (ROS)-associated mitochondrial damage. These findings suggest a novel etiopathogenic mechanism of SAE, which is initiated by the increased LCN2, leading to neuronal loss and cognitive deficit. Inhibition of LCN2 could be therapeutically beneficial in treating sepsis-induced synaptic and cognitive impairments.
Collapse
Affiliation(s)
- Cuiping Guo
- Department of Emergency Medicine & Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Biomedical Sciences, School of Medicine, Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wensheng Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Abdoul Razak Mahaman Yacoubou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianzhi Wang
- Institute of Biomedical Sciences, School of Medicine, Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusheng Li
- Department of Emergency Medicine & Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaochuan Wang
- Department of Emergency Medicine & Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Institute of Biomedical Sciences, School of Medicine, Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, China.
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
13
|
Song R, Glass JO, Wu S, Li Y, Robinson GW, Gajjar A, Merchant TE, Reddick WE. Perivascular space imaging during therapy for medulloblastoma. PLoS One 2025; 20:e0318278. [PMID: 39919146 PMCID: PMC11805390 DOI: 10.1371/journal.pone.0318278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
Perivascular spaces (PVS) are fluid filled compartments surrounding the small blood vessels in the brain. The impact of radiotherapy and chemotherapy on PVS remains unclear. The aim of this study is to investigate treatment effects of radiotherapy and chemotherapy at four time points (TPs) in pediatric medulloblastoma (MB) patients. We examined 778 scans from 241 MB patients at baseline (0M), after 12 weeks (about 3 months) of radiotherapy and rest (3M), after chemotherapy completion (12M), and a follow-up (FollowUp) at 18- or 21-months post-baseline. PVS was segmented by applying Frangi filter on the white matter regions on T1 weighted images acquired at 3T Siemens MRI scanner using MPRAGE. PVS volume and ratio, defined as the ratio of PVS volume to the white matter volume, were measured at the four TPs. The data was first statistically analyzed using a full model where all data were included, then a paired model, which included only patients who completed consecutive measurements under the same anesthesia and shunt conditions. Both the full model and paired model showed that PVS (including ratio and volume) increased at 3M post-radiotherapy compared to baseline. During chemotherapy, PVS decreased significantly from 3M to 12M. Subsequently, from 12M to FollowUp, PVS increased again. MRI exams under anesthesia exhibited significantly lower PVS than those without anesthesia. Patients who had undergone a shunt procedure exhibited a significantly reduced PVS compared to those who had not undergone the procedure. We concluded that craniospinal irradiation led to an elevated PVS. Conversely, chemotherapy or time post-irradiation decreased PVS. Anesthesia and shunt procedures can also influence perivascular space ratio or volume.
Collapse
Affiliation(s)
- Ruitian Song
- Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - John O. Glass
- Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Shengjie Wu
- Biostatistics, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Yimei Li
- Biostatistics, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Giles W. Robinson
- Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Amar Gajjar
- Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Thomas E. Merchant
- Radiation Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Wilburn E. Reddick
- Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
14
|
Yu X, Chen Z, Ruan F, Jiang Y, Bao W, Wu D, Chao L, Wu R, Le K. Inhibition of PAD4-mediated neutrophil extracellular traps formation attenuates hypoxic-ischemic brain injury in neonatal mice. Exp Neurol 2025; 384:115065. [PMID: 39566838 DOI: 10.1016/j.expneurol.2024.115065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is the primary cause of neonatal mortality and severe neurological sequelae. The interaction of neuroinflammation with the immune system represents a significant pathological mechanism underlying the development of HIE. Neutrophil extracellular traps (NETs) are a recently identified antimicrobial mechanism utilized by neutrophils. NETs can act as damage-associated molecular patterns, thereby amplifying the immune response and exerting proinflammatory effects. However, further research is needed to elucidate their role in the pathogenesis of HIE. In this study, we investigated the role of NETs in a hypoxic-ischemic brain injury (HIBI) model. We first reported that a pharmacological intervention to inhibit peptidylarginine deiminase type IV (PAD4) may constitute an effective strategy for reducing HI insult-induced neuroinflammation, neuronal apoptosis, and brain tissue destruction while also enhancing long-term neurobehavioral function in mice. These results support a pathological role for NETs in HIBI, and targeting PAD4 is a potential direction for the treatment of HIE.
Collapse
Affiliation(s)
- Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Lishuo Chao
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), NO.36 Mingxing Road, Guangzhou, Guangdong Province 510370, China
| | - Rui Wu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang 330006, Jiangxi Province, China; Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hong Kong, China.
| |
Collapse
|
15
|
Satyanarayanan SK, Han Z, Xiao J, Yuan Q, Yung WH, Ke Y, Chang RCC, Zhu MH, Su H, Su KP, Qin D, Lee SMY. Frontiers of Neurodegenerative Disease Treatment: Targeting Immune Cells in Brain Border Regions. Brain Behav Immun 2025; 123:483-499. [PMID: 39378973 DOI: 10.1016/j.bbi.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Neurodegenerative diseases (NDs) demonstrate a complex interaction with the immune system, challenging the traditional view of the brain as an "immune-privileged" organ. Microglia were once considered the sole guardians of the brain's immune response. However, recent research has revealed the critical role of peripheral immune cells located in key brain regions like the meninges, choroid plexus, and perivascular spaces. These previously overlooked cells are now recognized as contributors to the development and progression of NDs. This newfound understanding opens doors for pioneering therapeutic strategies. By targeting these peripheral immune cells, we may be able to modulate the brain's immune environment, offering an alternative approach to treat NDs and circumvent the challenges posed by the blood-brain barrier. This comprehensive review will scrutinize the latest findings on the complex interactions between these peripheral immune cells and NDs. It will also critically assess the prospects of targeting these cells as a ground-breaking therapeutic avenue for these debilitating disorders.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Zixu Han
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Jingwei Xiao
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Ya Ke
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Faculty of Medicine Building, Hong Kong, China
| | - Maria Huachen Zhu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Hong Kong, China
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Suki Man Yan Lee
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
16
|
Han C, Zhu M, Liu Y, Yang Y, Cheng J, Li P. Regulation of Vascular Injury and Repair by P21-Activated Kinase 1 and P21-Activated Kinase 2: Therapeutic Potential and Challenges. Biomolecules 2024; 14:1596. [PMID: 39766303 PMCID: PMC11674331 DOI: 10.3390/biom14121596] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
The PAK (p21-activated kinases) family is a class of intracellular signal transduction protein kinases that regulate various cellular functions, mainly through their interactions with small GTP enzymes. PAK1 and PAK2 in the PAK kinase family are key signal transduction molecules that play important roles in various biological processes, including morphological changes, migration, proliferation, and apoptosis, and are involved in the progression of many diseases. Abnormal expression or dysregulation of PAK1 and PAK2 may be associated with several diseases, including cancer, neurological diseases, etc. The current research mainly focuses on studying the role of PAK and PAK inhibitors in the regulation of cancer progression, but relatively few reports are available that explore their potential role in cardiovascular diseases. Vascular injury and repair are complex processes involved in many cardiovascular conditions, including atherosclerosis, restenosis, and hypertension. Emerging research suggests that PAK1 and PAK2 have pivotal roles in vascular endothelial cell functions, including migration, proliferation, and angiogenesis. These kinases also modulate vascular smooth muscle relaxation, vascular permeability, and structural alterations, which are critical in the development of atherosclerosis and vascular inflammation. By targeting these activities, PAK proteins are essential for both normal vascular physiology and the pathogenesis of vascular diseases, highlighting their potential as therapeutic targets for vascular health. This review focuses on recent studies that offer experimental insights into the mechanisms by which PAK1 and PAK2 regulate the biological processes of vascular injury and repair and the therapeutic potential of the current existing PAK inhibitors in vascular-related diseases. The limitations of treatment with some PAK inhibitors and the ways that future development can overcome these challenges are also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China; (C.H.); (M.Z.); (Y.L.); (Y.Y.); (J.C.)
| |
Collapse
|
17
|
Zou K, Deng Q, Zhang H, Huang C. Glymphatic system: a gateway for neuroinflammation. Neural Regen Res 2024; 19:2661-2672. [PMID: 38595285 PMCID: PMC11168510 DOI: 10.4103/1673-5374.391312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 04/11/2024] Open
Abstract
The glymphatic system is a relatively recently identified fluid exchange and transport system in the brain. Accumulating evidence indicates that glymphatic function is impaired not only in central nervous system disorders but also in systemic diseases. Systemic diseases can trigger the inflammatory responses in the central nervous system, occasionally leading to sustained inflammation and functional disturbance of the central nervous system. This review summarizes the current knowledge on the association between glymphatic dysfunction and central nervous system inflammation. In addition, we discuss the hypothesis that disease conditions initially associated with peripheral inflammation overwhelm the performance of the glymphatic system, thereby triggering central nervous system dysfunction, chronic neuroinflammation, and neurodegeneration. Future research investigating the role of the glymphatic system in neuroinflammation may offer innovative therapeutic approaches for central nervous system disorders.
Collapse
Affiliation(s)
- Kailu Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qingwei Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hong Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Changsheng Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
18
|
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
|
19
|
Bala N, McGurk A, Carter EM, Sidhu I, Niak S, Leddon SA, Fowell DJ. Th1 cells are critical tissue organizers of myeloid-rich perivascular activation niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.24.625073. [PMID: 39651309 PMCID: PMC11623525 DOI: 10.1101/2024.11.24.625073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Aggregating immune cells within perivascular niches (PVN) can regulate tissue immunity in infection, autoimmunity and cancer. How cells are assembled at PVNs and the activation signals imparted within remain unclear. Here, we integrate dynamic time-resolved in vivo imaging with a novel spatially-resolved platform for microanatomical interrogation of transcriptome, immune phenotype and inflammatory mediators in skin PVNs. We uncover a complex positive-feedback loop within CXCL10 + PVNs that regulates myeloid and Th1 cell positioning for exchange of critical signals for Th1 activation. Th1 cells spend ∼24h in the PVN, receiving initial peripheral activation signals, before redeploying to the inflamed dermal parenchyma. Niche-enriched, CCR2-dependent myeloid cells were critical for Th1 IFNγ-production. In turn, PVN instructional signals enabled Th1s to orchestrate PVN assembly by CXCR2-dependent intra-tissue myeloid cell aggregation. The results reveal a critical tissue organizing role for Th1s, gained rapidly on tissue entry, that could be exploited to boost regional immunity. HIGHLIGHTS Perivascular niche (PVN): myeloid hubs in inflamed mouse and healthy human skinTh1 cells enter, get activated, and leave the PVN within first 24h of tissue entryAntigen-specific signals in the PVN promote the tissue organizing functions of Th1sTh1 cells assemble the PVN via CXCR2-dependent myeloid cell aggregation.
Collapse
|
20
|
Betsholtz C, Engelhardt B, Koh GY, McDonald DM, Proulx ST, Siegenthaler J. Advances and controversies in meningeal biology. Nat Neurosci 2024; 27:2056-2072. [PMID: 39333784 PMCID: PMC11862877 DOI: 10.1038/s41593-024-01701-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/12/2024] [Indexed: 09/30/2024]
Abstract
The dura, arachnoid and pia mater, as the constituent layers of the meninges, along with cerebrospinal fluid in the subarachnoid space and ventricles, are essential protectors of the brain and spinal cord. Complemented by immune cells, blood vessels, lymphatic vessels and nerves, these connective tissue layers have held many secrets that have only recently begun to be revealed. Each meningeal layer is now known to have molecularly distinct types of fibroblasts. Cerebrospinal fluid clearance through peripheral lymphatics and lymph nodes is well documented, but its routes and flow dynamics are debated. Advances made in meningeal immune functions are also debated. This Review considers the cellular and molecular structure and function of the dura, arachnoid and pia mater in the context of conventional views, recent progress, and what is uncertain or unknown. The hallmarks of meningeal pathophysiology are identified toward developing a more complete understanding of the meninges in health and disease.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden and Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | | | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science and Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julie Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, Colorado, CO, USA.
| |
Collapse
|
21
|
Borrelli S, Leclercq S, Pasi M, Maggi P. Cerebral small vessel disease and glymphatic system dysfunction in multiple sclerosis: A narrative review. Mult Scler Relat Disord 2024; 91:105878. [PMID: 39276600 DOI: 10.1016/j.msard.2024.105878] [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: 06/24/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
As the multiple sclerosis (MS) population ages, the prevalence of vascular comorbidities increases, potentially accelerating disease progression and brain atrophy. Recent studies highlight the prevalence of cerebral small vessel disease (CSVD) in MS, suggesting a potential link between vascular comorbidities and accelerated disability. CSVD affects the brain's small vessels, often leading to identifiable markers on MRI such as enlarged perivascular spaces (EPVS). EPVS are increasingly recognized also in MS and have been associated with vascular comorbidities, lower percentage of MS-specific perivenular lesions, brain atrophy and aging. The exact sequence of event leading to MRI visible EPVS is yet to be determined, but an impaired perivascular brain fluid drainage appears a possible physiopathological explanation for EPVS in both CSVD and MS. In this context, a dysfunction of the brain fluid clearance system - also known as "glymphatic system" - appears associated in MS to aging, neuroinflammation, and vascular dysfunction. Advanced imaging techniques show an impaired glymphatic function in both MS and CSVD. Additionally, lifestyle factors such as physical exercise, diet, and sleep quality appear to influence glymphatic function, potentially revealing novel therapeutic strategies to mitigate microangiopathy and neuroinflammation in MS. This review underscores the potential role of glymphatic dysfunction in the complex and not-yet elucidated interplay between neuroinflammation and CSVD in MS.
Collapse
Affiliation(s)
- Serena Borrelli
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium.
| | - Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Marco Pasi
- Stroke Unit, Department of Neurology, CIC-IT 1415, CHRU de Tours, INSERM 1253 iBrain, Tours, France
| | - Pietro Maggi
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Av. Hippocrate 10, Brussels 1200, Belgium.
| |
Collapse
|
22
|
Menze I, Bernal J, Kaya P, Aki Ç, Pfister M, Geisendörfer J, Yakupov R, Coello RD, Valdés-Hernández MDC, Heneka MT, Brosseron F, Schmid MC, Glanz W, Incesoy EI, Butryn M, Rostamzadeh A, Meiberth D, Peters O, Preis L, Lammerding D, Gref D, Priller J, Spruth EJ, Altenstein S, Lohse A, Hetzer S, Schneider A, Fliessbach K, Kimmich O, Vogt IR, Wiltfang J, Bartels C, Schott BH, Hansen N, Dechent P, Buerger K, Janowitz D, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Sanzenbacher C, Hinderer P, Scheffler K, Spottke A, Roy-Kluth N, Lüsebrink F, Neumann K, Wardlaw J, Jessen F, Schreiber S, Düzel E, Ziegler G. Perivascular space enlargement accelerates in ageing and Alzheimer's disease pathology: evidence from a three-year longitudinal multicentre study. Alzheimers Res Ther 2024; 16:242. [PMID: 39482759 PMCID: PMC11526621 DOI: 10.1186/s13195-024-01603-8] [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: 06/20/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Perivascular space (PVS) enlargement in ageing and Alzheimer's disease (AD) and the drivers of such a structural change in humans require longitudinal investigation. Elucidating the effects of demographic factors, hypertension, cerebrovascular dysfunction, and AD pathology on PVS dynamics could inform the role of PVS in brain health function as well as the complex pathophysiology of AD. METHODS We studied PVS in centrum semiovale (CSO) and basal ganglia (BG) computationally over three to four annual visits in 503 participants (255 females; meanage = 70.78 ± 5.78) of the ongoing observational multicentre "DZNE Longitudinal Cognitive Impairment and Dementia Study" (DELCODE) cohort. We analysed data from subjects who were cognitively unimpaired (n = 401), had amnestic mild cognitive impairment (n = 71), or had AD (n = 31). We used linear mixed-effects modelling to test for changes of PVS volumes in relation to cross-sectional and longitudinal age, as well as sex, years of education, hypertension, white matter hyperintensities, AD diagnosis, and cerebrospinal-fluid-derived amyloid (A) and tau (T) status (available for 46.71%; A-T-/A + T-/A + T + n = 143/48/39). RESULTS PVS volumes increased significantly over follow-ups (CSO: B = 0.03 [0.02, 0.05], p < 0.001; BG: B = 0.05 [0.03, 0.07], p < 0.001). PVS enlargement rates varied substantially across subjects and depended on the participant's age, white matter hyperintensities volumes, and amyloid and tau status. PVS volumes were higher across elderly participants, regardless of region of interest (CSO: B = 0.12 [0.02, 0.21], p = 0.017; BG: B = 0.19 [0.09, 0.28], p < 0.001). Faster BG-PVS enlargement related to lower baseline white matter hyperintensities volumes (ρspearman = -0.17, pFDR = 0.001) and was more pronounced in individuals who presented with combined amyloid and tau positivity versus negativity (A + T + > A-T-, pFDR = 0.004) or who were amyloid positive but tau negative (A + T + > A + T-, pFDR = 0.07). CSO-PVS volumes increased at a faster rate with amyloid positivity as compared to amyloid negativity (A + T-/A + T + > A-T-, pFDR = 0.021). CONCLUSION Our longitudinal evidence supports the relevance of PVS enlargement in presumably healthy ageing as well as in AD pathology. We further discuss the region-specific involvement of white matter hyperintensities and neurotoxic waste accumulation in PVS enlargement and the possibility of additional factors contributing to PVS progression. A comprehensive understanding of PVS dynamics could facilitate the understanding of pathological cascades and might inform targeted treatment strategies. TRIAL REGISTRATION German Clinical Trials Register DRKS00007966. Registered 04.05.2015 - retrospectively registered, https://drks.de/search/en/trial/DRKS00007966 .
Collapse
Affiliation(s)
- Inga Menze
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany.
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany.
| | - Jose Bernal
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
- Centre for Clinical Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh Bioquarter, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, EH16 4SB, UK
| | - Pinar Kaya
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Çağla Aki
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Malte Pfister
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Jonas Geisendörfer
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Renat Yakupov
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Roberto Duarte Coello
- Centre for Clinical Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh Bioquarter, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, EH16 4SB, UK
| | - Maria D C Valdés-Hernández
- Centre for Clinical Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh Bioquarter, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, EH16 4SB, UK
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, 6 Avenue du Swing 4367 , Esch-Belval, Luxembourg
| | - Frederic Brosseron
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Matthias C Schmid
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Wenzel Glanz
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Enise I Incesoy
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Michaela Butryn
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, Medical Faculty, University of Cologne, Kerpener Strasse 62, Cologne, 50924, Germany
| | - Dix Meiberth
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Kerpener Strasse 62, Cologne, 50924, Germany
| | - Oliver Peters
- German Centre for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin, 10117, Germany
- Institute of Psychiatry and Psychotherapy, Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Lukas Preis
- Institute of Psychiatry and Psychotherapy, Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Dominik Lammerding
- Institute of Psychiatry and Psychotherapy, Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Daria Gref
- Institute of Psychiatry and Psychotherapy, Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Josef Priller
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh Bioquarter, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, EH16 4SB, UK
- German Centre for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin, 10117, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, 10117, Germany
- School of Medicine, Department of Psychiatry and Psychotherapy, Technical University of Munich, Ismaninger Str. 22, Munich, 81675, Germany
| | - Eike J Spruth
- German Centre for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin, 10117, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, 10117, Germany
| | - Slawek Altenstein
- German Centre for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin, 10117, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, 10117, Germany
| | - Andrea Lohse
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, 10117, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité, Charitéplatz 1, Berlin, 10117, Germany
| | - Anja Schneider
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Klaus Fliessbach
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Okka Kimmich
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Ina R Vogt
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Jens Wiltfang
- German Centre for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, Goettingen, 37075, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, Goettingen, 37075, Germany
| | - Björn H Schott
- German Centre for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, Goettingen, 37075, Germany
- Leibniz Institute for Neurobiology, Brenneckestraße 6, Magdeburg, 39118, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, Goettingen, 37075, Germany
| | - Peter Dechent
- Department of Cognitive Neurology, MR-Research in Neurosciences, Georg-August-University Goettingen, Robert-Koch-Straße 40, Göttingen, 37075, Germany
| | - Katharina Buerger
- German Centre for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich, 81377, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, Munich, 81377, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, Munich, 81377, Germany
| | - Robert Perneczky
- German Centre for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich, 81377, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstraße 7, Munich, München, 80336 , Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, Munich, 81377, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, Charing Cross Hospital, St Dunstan's Road, London, W6 8RP, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Nußbaumstraße 7, Munich, München, 80336 , Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Rd, Sheffield, Broomhall, Sheffield, S10 2HQ, UK
- Department of Neuroradiology, University Hospital LMU, Marchioninistr. 15, Munich, 81377, Germany
| | - Stefan Teipel
- German Centre for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, Rostock, 18147, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Straße 20, Rostock, 18147, Germany
| | - Ingo Kilimann
- German Centre for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, Rostock, 18147, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Straße 20, Rostock, 18147, Germany
| | - Doreen Goerss
- German Centre for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, Rostock, 18147, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Straße 20, Rostock, 18147, Germany
| | - Christoph Laske
- German Centre for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, Tübingen, 72076, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Osianderstraße 24, Tübingen, 72076, Germany
| | - Matthias H Munk
- German Centre for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, Tübingen, 72076, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Osianderstraße 24, Tübingen, 72076 , Germany
| | - Carolin Sanzenbacher
- German Centre for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, Tübingen, 72076, Germany
| | - Petra Hinderer
- German Centre for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, Tübingen, 72076, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Otfried-Müller-Straße 51, Tübingen, 72076, Germany
| | - Annika Spottke
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Department of Neurology, University of Bonn, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Nina Roy-Kluth
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Falk Lüsebrink
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Katja Neumann
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh Bioquarter, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, EH16 4SB, UK
| | - Frank Jessen
- German Centre for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn, 53127, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Kerpener Strasse 62, Cologne, 50924, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
| | - Stefanie Schreiber
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Department of Neurology, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Emrah Düzel
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| | - Gabriel Ziegler
- German Centre for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, 39120, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, 39120, Germany
| |
Collapse
|
23
|
Morozova A, Španiel F, Škoch A, Brabec M, Zolotarov G, Musil V, Zach P. Enlarged brain perivascular spaces correlate with blood plasma osmolality in the healthy population: A longitudinal study. Neuroimage 2024; 300:120871. [PMID: 39341473 DOI: 10.1016/j.neuroimage.2024.120871] [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: 03/10/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
Enlarged perivascular spaces (EPVS) are increasingly recognized as an MRI detectable feature of neuroinflammatory processes and age-related neurodegenerative changes. Understanding perivascular characteristics in healthy individuals is crucial for their applicability as a reference for pathological changes. Limited data exists on the EPVS load and interhemispheric asymmetry in distribution among young healthy subjects. Despite the known impact of hydration on brain morphometric studies, blood plasma osmolality's effect on EPVS remains unexplored. This study investigated the influence of age, total intracranial volume (TIV), and blood plasma osmolality on EPVS characteristics in 59 healthy adults, each undergoing MRI and osmolality assessment twice within 14.8 months (mean ± 4 months). EPVS analysis was conducted in the centrum semiovale using high-resolution automated segmentation, followed by an optimization algorithm to enhance EPVS segmentation accuracy. Linear Mixed Effects model was used for the statistical analysis, which unveiled significant inter-individual variability in EPVS load and inter-hemispheric asymmetry. EPVS volume increased with age, higher TIV and lower blood plasma osmolality levels. Our findings offer valuable insights into EPVS characteristics among the healthy population, establishing a foundation to further explore age-related and pathological changes.
Collapse
Affiliation(s)
- Alexandra Morozova
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czechia; National Institute of Mental Health, Klecany, Czechia.
| | - Filip Španiel
- National Institute of Mental Health, Klecany, Czechia
| | - Antonín Škoch
- National Institute of Mental Health, Klecany, Czechia
| | - Marek Brabec
- Department of Statistical Modeling, Institute of Computer Science, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Grygoriy Zolotarov
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain
| | - Vladimir Musil
- Centre of Scientific Information, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Petr Zach
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czechia; National Institute of Mental Health, Klecany, Czechia
| |
Collapse
|
24
|
Liu H, Meng L, Wang J, Qin C, Feng R, Chen Y, Chen P, Zhu Q, Ma M, Teng J, Ding X. Enlarged perivascular spaces in alcohol-related brain damage induced by dyslipidemia. J Cereb Blood Flow Metab 2024; 44:1867-1880. [PMID: 38700501 PMCID: PMC11494831 DOI: 10.1177/0271678x241251570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
Perivascular spaces (PVSs) as the anatomical basis of the glymphatic system, are increasingly recognized as potential imaging biomarkers of neurological conditions. However, it is not clear whether enlarged PVSs are associated with alcohol-related brain damage (ARBD). We aimed to investigate the effect of long-term alcohol exposure on dyslipidemia and the glymphatic system in ARBD. We found that patients with ARBD exhibited significantly enlargement of PVSs in the frontal cortex and basal ganglia, as well as a notable increased levels of total cholesterol (TC) and triglycerides (TG). The anatomical changes of the glymphatic drainage system mentioned above were positively associated with TC and TG. To further explore whether enlarged PVSs affects the function of the glymphatic system in ARBD, we constructed long alcohol exposure and high fat diet mice models. The mouse model of long alcohol exposure exhibited increased levels of TC and TG, enlarged PVSs, the loss of aquaporin-4 polarity caused by reactive astrocytes and impaired glymphatic drainage function which ultimately caused cognitive deficits, in a similar way as high fat diet leading to impairment in glymphatic drainage. Our study highlights the contribution of dyslipidemia due to long-term alcohol abuse in the impairment of the glymphatic drainage system.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Lin Meng
- Department of Neurology, Zhengzhou Central Hospital, Zhengzhou, Henan 450000, China
| | - Jiuqi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Chi Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Renyi Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Yongkang Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Pei Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Qingyong Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Mingming Ma
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, Henan 450000, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Xuebing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| |
Collapse
|
25
|
Fisher TM, Liddelow SA. Emerging roles of astrocytes as immune effectors in the central nervous system. Trends Immunol 2024; 45:824-836. [PMID: 39332912 DOI: 10.1016/j.it.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
The astrocyte, a major glial cell type in the central nervous system (CNS), is widely regarded as a functionally diverse mediator of homeostasis. During development and throughout adulthood, astrocytes have essential roles, such as providing neuron metabolic support, modulating synaptic function, and maintaining the blood-brain barrier (BBB). Recent evidence continues to underscore their functional heterogeneity and importance for CNS maintenance, as well as how these cells ensure optimal CNS and immune responses to disease, acute trauma, and infection. Advances in our understanding of neuroimmune interactions complement our knowledge of astrocyte functional heterogeneity, where astrocytes are now regarded as key effectors and propagators of immune signaling. This shift in perspective highlights the role of astrocytes not merely as support cells, but as active participants in CNS immune responses.
Collapse
Affiliation(s)
- Theodore M Fisher
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, USA; Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
26
|
Karkoska KA, Gollamudi J, Sawyer RP, Woo D, Hyacinth HI. Quantifying dilated perivascular spaces in children with sickle cell disease. Pediatr Blood Cancer 2024; 71:e31150. [PMID: 38953143 PMCID: PMC11327878 DOI: 10.1002/pbc.31150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/16/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024]
Abstract
Sickle cell disease (SCD)-related neurological effects are particularly devastating. Dilated perivascular spaces (dPVS) are a well-described component of cerebral small vessel disease in older adults without SCD. However, the burden and association of dPVS with neurological complications in children with SCD have not been described. In this study, we used the international consensus criteria to quantify dPVS in the centrum semiovale and basal ganglia in T2-weighted magnetic resonance images (MRI) of children with SCD who were randomized as part of the Silent Cerebral Infarct Transfusion (SIT) trial. We examined the relationship between global and/or regional dPVS burden and presence or area of silent cerebral infarctions, hematological measures, demographic variables, and full-scale intelligence quotient (FSIQ) scores. The study included 156 SIT trial participants who had pre-randomization and study exit MRI. Their median age was 9.6 (5-15) years, 39% were female, and 94 (60%) participants had a high dPVS burden. Participants randomized to the blood transfusion arm and who had a high dPVS burden at baseline had a moderate decline in dPVS score over 36 months compared to no change in the observation group. On multivariable logistic regression, intelligence quotient was not associated with dPVS burden. Children with SCD included in the SIT trial have a high burden of dPVS compared to children without SCD. However, dPVS do not appear to have the same pathophysiology of silent cerebral infarcts. Further study is needed to determine both their etiology and clinical relevance.
Collapse
Affiliation(s)
- Kristine A Karkoska
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jahnavi Gollamudi
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Russell P Sawyer
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hyacinth I Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
27
|
Sterling JK, Rajesh A, Droho S, Gong J, Wang AL, Voigt AP, Brookins CE, Lavine JA. Retinal perivascular macrophages regulate immune cell infiltration during neuroinflammation in mouse models of ocular disease. J Clin Invest 2024; 134:e180904. [PMID: 39207852 PMCID: PMC11473146 DOI: 10.1172/jci180904] [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: 03/07/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The blood-retina barrier (BRB), which is disrupted in diabetic retinopathy (DR) and uveitis, is an important anatomical characteristic of the retina, regulating nutrient, waste, water, protein, and immune cell flux. The BRB is composed of endothelial cell tight junctions, pericytes, astrocyte end feet, a collagen basement membrane, and perivascular macrophages. Despite the importance of the BRB, retinal perivascular macrophage function remains unknown. We found that retinal perivascular macrophages resided on postcapillary venules in the superficial vascular plexus and expressed MHC class II. Using single-cell RNA-Seq, we found that perivascular macrophages expressed a prochemotactic transcriptome and identified platelet factor 4 (Pf4, also known as CXCL4) as a perivascular macrophage marker. We used Pf4Cre mice to specifically deplete perivascular macrophages. To model retinal inflammation, we performed intraocular CCL2 injections. Ly6C+ monocytes crossed the BRB proximal to perivascular macrophages. Depletion of perivascular macrophages severely hampered Ly6C+ monocyte infiltration. These data suggest that retinal perivascular macrophages orchestrate immune cell migration across the BRB, with implications for inflammatory ocular diseases including DR and uveitis.
Collapse
Affiliation(s)
- Jacob K. Sterling
- Department of Medicine, Feinberg School of Medicine
- Physician Scientist Track Program, Internal Medicine Residency, and
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Amrita Rajesh
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Joyce Gong
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Andrew L. Wang
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Andrew P. Voigt
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C. Elysse Brookins
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jeremy A. Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
28
|
Yamamoto EA, Koike S, Wong C, Dennis LE, Luther MN, Scatena A, Khambadkone S, Iliff JJ, Lim MM, Levendovszky SR, Elliott JE, Barisano G, Müller-Oehring EM, Morales AM, Baker FC, Nagel BJ, Piantino J. Biological sex and BMI influence the longitudinal evolution of adolescent and young adult MRI-visible perivascular spaces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.17.608337. [PMID: 39229241 PMCID: PMC11370374 DOI: 10.1101/2024.08.17.608337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background and Purpose An association recently emerged between magnetic resonance imaging (MRI)-visible perivascular spaces (MV-PVS) with intracerebral solute clearance and neuroinflammation, in adults. However, it is unknown how MV-PVS change throughout adolescence and what factors influence MV-PVS volume and morphology. This study assesses the temporal evolution of MV-PVS volume in adolescents and young adults, and secondarily evaluates the relationship between MV-PVS, age, sex, and body mass index (BMI). Materials and Methods This analysis included a 783 participant cohort from the longitudinal multicenter National Consortium on Alcohol and Neurodevelopment in Adolescence study that involved up to 6 imaging visits spanning 5 years. Healthy adolescents aged 12-21 years at study entry with at least two MRI scans were included. The primary outcome was mean MV-PVS volume (mm 3 /white matter cm 3 ). Results On average, males had greater MV-PVS volume at all ages compared to females. A linear mixed-effect model for MV-PVS volume was performed. Mean BMI and increases in a person's BMI were associated with increases in MV-PVS volume over time. In females only, changes in BMI correlated with MV-PVS volume. One unit increase in BMI above a person's average BMI was associated with a 0.021 mm 3 /cm 3 increase in MV-PVS volume (p<0.001). Conclusion This longitudinal study showed sex differences in MV-PVS features during adolescence and young adulthood. Importantly, we report that increases in BMI from a person's mean BMI are associated with increases in MV-PVS volume in females only. These findings suggest a potential link between MV-PVS, sex, and BMI that warrants future study.
Collapse
|
29
|
Lee DA, Lee HJ, Kim J, Park KM. Association between patients with migraine and sarcopenia: A retrospective study. Medicine (Baltimore) 2024; 103:e38941. [PMID: 38996151 PMCID: PMC11245205 DOI: 10.1097/md.0000000000038941] [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: 02/23/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Recently, interest in sarcopenia has been increasing in patients with various neurological diseases. Thus, we investigated the presence of sarcopenia in patients with episodic migraine (EM) based on temporal muscle thickness (TMT). This was a retrospectively observational study following STROBE guidelines. We enrolled patients with EM and healthy controls. Both groups underwent brain magnetic resonance imaging, including three-dimensional T1-weighted imaging. We calculated the TMT using T1-weighted imaging, which is a marker for sarcopenia. We compared TMT between patients with EM and healthy controls, and analyzed it according to presence of migraine aura. We retrospectively enrolled 82 patients with EM and 53 healthy controls. TMT was not different between patients with EM and healthy controls (10.804 ± 2.045 mm in patients with EM vs 10.721 ± 1.547 mm in healthy controls, P = .801). Furthermore, TMT was not different according to presence of migraine aura in patients with EM (10.994 ± 2.016 mm in patients with migraine aura vs 10.716 ± 2.071 mm in those without, P = .569). There were no correlations between TMT and clinical characteristics in patients with EM, including age, age of onset, duration of migraine, headache intensity, and headache frequency. This study found no statistical difference in TMT between patients with EM and healthy controls or between patients with EM with and without aura. These findings suggest that there is no evidence of sarcopenia in patients with EM.
Collapse
Affiliation(s)
- Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jinseung Kim
- Department of Family Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
30
|
VanElzakker MB, Bues HF, Brusaferri L, Kim M, Saadi D, Ratai EM, Dougherty DD, Loggia ML. Neuroinflammation in post-acute sequelae of COVID-19 (PASC) as assessed by [ 11C]PBR28 PET correlates with vascular disease measures. Brain Behav Immun 2024; 119:713-723. [PMID: 38642615 PMCID: PMC11225883 DOI: 10.1016/j.bbi.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/28/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has triggered a consequential public health crisis of post-acute sequelae of COVID-19 (PASC), sometimes referred to as long COVID. The mechanisms of the heterogeneous persistent symptoms and signs that comprise PASC are under investigation, and several studies have pointed to the central nervous and vascular systems as being potential sites of dysfunction. In the current study, we recruited individuals with PASC with diverse symptoms, and examined the relationship between neuroinflammation and circulating markers of vascular dysfunction. We used [11C]PBR28 PET neuroimaging, a marker of neuroinflammation, to compare 12 PASC individuals versus 43 normative healthy controls. We found significantly increased neuroinflammation in PASC versus controls across a wide swath of brain regions including midcingulate and anterior cingulate cortex, corpus callosum, thalamus, basal ganglia, and at the boundaries of ventricles. We also collected and analyzed peripheral blood plasma from the PASC individuals and found significant positive correlations between neuroinflammation and several circulating analytes related to vascular dysfunction. These results suggest that an interaction between neuroinflammation and vascular health may contribute to common symptoms of PASC.
Collapse
Affiliation(s)
- Michael B VanElzakker
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; PolyBio Research Foundation, Medford, MA, USA.
| | - Hannah F Bues
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Computer Science And Informatics, School of Engineering, London South Bank University, London, UK
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena Saadi
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Borrelli S, Guisset F, Vanden Bulcke C, Stölting A, Bugli C, Lolli V, Du Pasquier R, van Pesch V, Absinta M, Pasi M, Maggi P. Enlarged perivascular spaces are associated with brain microangiopathy and aging in multiple sclerosis. Mult Scler 2024; 30:983-993. [PMID: 38850029 DOI: 10.1177/13524585241256881] [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: 06/09/2024]
Abstract
BACKGROUND Growing evidence links brain-MRI enlarged perivascular spaces (EPVS) and multiple sclerosis (MS), but their role remains unclear. OBJECTIVE This study aimed to investigate the cross-sectional associations of EPVS with several neuroinflammatory and neurodegenerative features in a large multicentric-MS cohort. METHODS In total, 207 patients underwent 3T axial-T2-weighted brain-MRI for EPVS assessment (EPVS dichotomized into high/low according to ⩾ 2/< 2 rating categories). MRI biomarkers included brain-predicted age and brain-predicted age difference (brain-PAD), central vein sign (CVS)-positive lesion percentage (CVS%), paramagnetic rim and cortical lesions, T2-lesion load, and brain volumetry. The variable relative importance for EPVS-category prediction was explored using a classification random forest approach. RESULTS High EPVS patients were older (49 vs 44 years, p = 0.003), had ⩾ 1 vascular risk factors (VRFs; p = 0.005), lower CVS% (67% vs 78%, p < 0.001), reduced brain volumes (whole brain: 0.63 vs 0.73, p = 0.01; gray matter: 0.36 vs 0.40; p = 0.002), and older brain-predicted age (58 vs 50 years, p < 0.001). No differences were found for neuroinflammatory markers. After adjusting for age and VFRs (multivariate analyses), the high EPVS category correlated with lower CVS% (odds ratio (OR) = 0.98, 95% confidence interval (CI) = 0.96-0.99; p = 0.02), lower whole brain (OR = 0.01, 95% CI = 0.0003-0.5; p = 0.02), gray matter (OR = 0.0004, 95% CI = 0.0000004-0.4; p = 0.03) volumes, and higher brain-PAD (OR = 1.05, 95% CI = 1.01-1.09; p = 0.02). Random forest identified brain-PAD as the most important predictor of high EPVS. CONCLUSION EPVS in MS likely reflect microangiopathic disease rather than neuroinflammation, potentially contributing to accelerated neurodegeneration.
Collapse
Affiliation(s)
- Serena Borrelli
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium/Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium
| | - François Guisset
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Colin Vanden Bulcke
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium/ICTEAM Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Anna Stölting
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Céline Bugli
- Plateforme technologique de Support en Méthodologie et Calcul Statistique, Université catholique de Louvain, Brussels, Belgium
| | - Valentina Lolli
- Department of Radiology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium
| | - Renaud Du Pasquier
- Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Vincent van Pesch
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Martina Absinta
- Vita-Salute San Raffaele University, Milan, Italy/Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy/Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marco Pasi
- Stroke Unit, Department of Neurology, CIC-IT 1415, CHRU de Tours, INSERM 1253 iBrain, Tours, France
| | - Pietro Maggi
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium/Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland/Department of Neurology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
32
|
Zhao Y, Lv X, Chen Y, Zhang C, Zhou D, Deng Y. Neuroinflammatory response on a newly combinatorial cell-cell interaction chip. Biomater Sci 2024; 12:2096-2107. [PMID: 38441146 DOI: 10.1039/d4bm00125g] [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: 04/17/2024]
Abstract
Neuroinflammation is a common feature in various neurological disorders. Understanding neuroinflammation and neuro-immune interactions is of significant importance. However, the intercellular interactions in the inflammatory model are intricate. Microfluidic chips, with their complex micrometer-scale structures and real-time observation capabilities, offer unique advantages in tackling these complexities compared to other techniques. In this study, microfluidic chip technology was used to construct a microarray physical barrier structure with 15 μm spacing, providing well-defined cell growth areas and clearly delineated interaction channels. Moreover, an innovative hydrophilic treatment process on the glass surface facilitated long-term co-culture of cells. The developed neuroinflammation model on the chip revealed that SH-SY5Y cytotoxicity was predominantly influenced by co-cultured THP-1 cells. The co-culture model fostered complex interactions that may exacerbate cytotoxicity, including irregular morphological changes of cells, cell viability reduction, THP-1 cell migration, and the release of inflammatory factors. The integration of the combinatorial cell-cell interaction chip not only offers a clear imaging detection platform but also provides diverse data on cell migration distance, migration direction, and migration angle. Furthermore, the designed ample space for cell culture, along with microscale channels with fluid characteristics, allow for the study of inflammatory factor distribution patterns on the chip, offering vital theoretical data on biological relevance that conventional experiments cannot achieve. The fabricated user-friendly, reusable, and durable co-culture chip serves as a valuable in vitro tool, providing an intuitive platform for gaining insights into the complex mechanisms underlying neuroinflammation and other interacting models.
Collapse
Affiliation(s)
- Yimeng Zhao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Xuefei Lv
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yu Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Chen Zhang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Di Zhou
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
33
|
Koller BH, Nguyen M, Snouwaert JN, Gabel CA, Ting JPY. Species-specific NLRP3 regulation and its role in CNS autoinflammatory diseases. Cell Rep 2024; 43:113852. [PMID: 38427558 PMCID: PMC12054400 DOI: 10.1016/j.celrep.2024.113852] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
The NLRP3 inflammasome is essential for caspase-1 activation and the release of interleukin (IL)-1β, IL-18, and gasdermin-D in myeloid cells. However, research on species-specific NLRP3's physiological impact is limited. We engineer mice with the human NLRP3 gene, driven by either the human or mouse promoter, via syntenic replacement at the mouse Nlrp3 locus. Both promoters facilitate hNLRP3 expression in myeloid cells, but the mouse promoter responds more robustly to LPS. Investigating the disease impact of differential NLRP3 regulation, we introduce the D305N gain-of-function mutation into both humanized lines. Chronic inflammation is evident with both promoters; however, CNS outcomes vary significantly. Despite poor response to LPS, the human promoter results in D305N-associated aseptic meningitis, mirroring human pathology. The mouse promoter, although leading to increased CNS expression post-LPS, does not induce meningitis in D305N mutants. Therefore, human-like NLRP3 expression may be crucial for accurate modeling of its role in disease pathogenesis.
Collapse
Affiliation(s)
- Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - MyTrang Nguyen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John N Snouwaert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Jenny P-Y Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
34
|
Karvelas N, Oh B, Wang E, Cobigo Y, Tsuei T, Fitzsimons S, Younes K, Ehrenberg A, Geschwind MD, Schwartz D, Kramer JH, Ferguson AR, Miller BL, Silbert LC, Rosen HJ, Elahi FM. Enlarged perivascular spaces are associated with white matter injury, cognition and inflammation in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Brain Commun 2024; 6:fcae071. [PMID: 38495305 PMCID: PMC10943571 DOI: 10.1093/braincomms/fcae071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/18/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
Enlarged perivascular spaces have been previously reported in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, but their significance and pathophysiology remains unclear. We investigated associations of white matter enlarged perivascular spaces with classical imaging measures, cognitive measures and plasma proteins to better understand what enlarged perivascular spaces represent in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and whether radiographic measures of enlarged perivascular spaces would be of value in future therapeutic discovery studies for cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Twenty-four individuals with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and 24 age- and sex-matched controls were included. Disease status was determined based on the presence of NOTCH3 mutation. Brain imaging measures of white matter hyperintensity, brain parenchymal fraction, white matter enlarged perivascular space volumes, clinical and cognitive measures as well as plasma proteomics were used in models. White matter enlarged perivascular space volumes were calculated via a novel, semiautomated pipeline, and levels of 7363 proteins were quantified in plasma using the SomaScan assay. The relationship of enlarged perivascular spaces with global burden of white matter hyperintensity, brain atrophy, functional status, neurocognitive measures and plasma proteins was modelled with linear regression models. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and control groups did not exhibit differences in mean enlarged perivascular space volumes. However, increased enlarged perivascular space volumes in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy were associated with increased white matter hyperintensity volume (β = 0.57, P = 0.05), Clinical Dementia Rating Sum-of-Boxes score (β = 0.49, P = 0.04) and marginally with decreased brain parenchymal fraction (β = -0.03, P = 0.10). In interaction term models, the interaction term between cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy disease status and enlarged perivascular space volume was associated with increased white matter hyperintensity volume (β = 0.57, P = 0.02), Clinical Dementia Rating Sum-of-Boxes score (β = 0.52, P = 0.02), Mini-Mental State Examination score (β = -1.49, P = 0.03) and marginally with decreased brain parenchymal fraction (β = -0.03, P = 0.07). Proteins positively associated with enlarged perivascular space volumes were found to be related to leukocyte migration and inflammation, while negatively associated proteins were related to lipid metabolism. Two central hub proteins were identified in protein networks associated with enlarged perivascular space volumes: CXC motif chemokine ligand 8/interleukin-8 and C-C motif chemokine ligand 2/monocyte chemoattractant protein 1. The levels of CXC motif chemokine ligand 8/interleukin-8 were also associated with increased white matter hyperintensity volume (β = 42.86, P = 0.03), and levels of C-C motif chemokine ligand 2/monocyte chemoattractant protein 1 were further associated with decreased brain parenchymal fraction (β = -0.0007, P < 0.01) and Mini-Mental State Examination score (β = -0.02, P < 0.01) and increased Trail Making Test B completion time (β = 0.76, P < 0.01). No proteins were associated with all three studied imaging measures of pathology (brain parenchymal fraction, enlarged perivascular spaces, white matter hyperintensity). Based on associations uncovered between enlarged perivascular space volumes and cognitive functions, imaging and plasma proteins, we conclude that white matter enlarged perivascular space volumes may capture pathologies contributing to chronic brain dysfunction and degeneration in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy.
Collapse
Affiliation(s)
- Nikolaos Karvelas
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bradley Oh
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Earnest Wang
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Yann Cobigo
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Torie Tsuei
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Stephen Fitzsimons
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyan Younes
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304, USA
| | - Alexander Ehrenberg
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael D Geschwind
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Daniel Schwartz
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Adam R Ferguson
- Department of Neurological surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94110, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Lisa C Silbert
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- NIA-Layton Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Portland Veterans Affairs Health Care System, Portland, OR 97239, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Fanny M Elahi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| |
Collapse
|
35
|
Kouba BR, de Araujo Borba L, Borges de Souza P, Gil-Mohapel J, Rodrigues ALS. Role of Inflammatory Mechanisms in Major Depressive Disorder: From Etiology to Potential Pharmacological Targets. Cells 2024; 13:423. [PMID: 38474387 PMCID: PMC10931285 DOI: 10.3390/cells13050423] [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: 01/18/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The involvement of central and peripheral inflammation in the pathogenesis and prognosis of major depressive disorder (MDD) has been demonstrated. The increase of pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-18, and TNF-α) in individuals with depression may elicit neuroinflammatory processes and peripheral inflammation, mechanisms that, in turn, can contribute to gut microbiota dysbiosis. Together, neuroinflammation and gut dysbiosis induce alterations in tryptophan metabolism, culminating in decreased serotonin synthesis, impairments in neuroplasticity-related mechanisms, and glutamate-mediated excitotoxicity. This review aims to highlight the inflammatory mechanisms (neuroinflammation, peripheral inflammation, and gut dysbiosis) involved in the pathophysiology of MDD and to explore novel anti-inflammatory therapeutic approaches for this psychiatric disturbance. Several lines of evidence have indicated that in addition to antidepressants, physical exercise, probiotics, and nutraceuticals (agmatine, ascorbic acid, and vitamin D) possess anti-inflammatory effects that may contribute to their antidepressant properties. Further studies are necessary to explore the therapeutic benefits of these alternative therapies for MDD.
Collapse
Affiliation(s)
- Bruna R. Kouba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Laura de Araujo Borba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Pedro Borges de Souza
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| |
Collapse
|
36
|
Zhao H, Sun M, Zhang Y, Kong W, Fan L, Wang K, Xu Q, Chen B, Dong J, Shi Y, Wang Z, Wang S, Zhuang X, Li Q, Lin F, Yao X, Zhang W, Kong C, Zhang R, Feng D, Zhao X. Connecting the Dots: The Cerebral Lymphatic System as a Bridge Between the Central Nervous System and Peripheral System in Health and Disease. Aging Dis 2024; 15:115-152. [PMID: 37307828 PMCID: PMC10796102 DOI: 10.14336/ad.2023.0516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/16/2023] [Indexed: 06/14/2023] Open
Abstract
As a recently discovered waste removal system in the brain, cerebral lymphatic system is thought to play an important role in regulating the homeostasis of the central nervous system. Currently, more and more attention is being focused on the cerebral lymphatic system. Further understanding of the structural and functional characteristics of cerebral lymphatic system is essential to better understand the pathogenesis of diseases and to explore therapeutic approaches. In this review, we summarize the structural components and functional characteristics of cerebral lymphatic system. More importantly, it is closely associated with peripheral system diseases in the gastrointestinal tract, liver, and kidney. However, there is still a gap in the study of the cerebral lymphatic system. However, we believe that it is a critical mediator of the interactions between the central nervous system and the peripheral system.
Collapse
Affiliation(s)
- Hongxiang Zhao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Meiyan Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Yue Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Wenwen Kong
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Lulu Fan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Kaifang Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Baiyan Chen
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Jianxin Dong
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Yanan Shi
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Zhengyan Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - ShiQi Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Xiaoli Zhuang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Qi Li
- Department of Anesthesiology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Feihong Lin
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Xinyu Yao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - WenBo Zhang
- Department of Neurosurgery, The Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Chang Kong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China.
| | - Rui Zhang
- Department of Anesthesiology, Affiliated Hospital of Weifang Medical University, Weifang, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Dayun Feng
- Department of neurosurgery, Tangdu hospital, Fourth Military Medical University, Xi'an, China.
| | - Xiaoyong Zhao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Department of Anesthesiology, Affiliated Hospital of Weifang Medical University, Weifang, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| |
Collapse
|
37
|
Okar SV, Fagiani F, Absinta M, Reich DS. Imaging of brain barrier inflammation and brain fluid drainage in human neurological diseases. Cell Mol Life Sci 2024; 81:31. [PMID: 38212566 PMCID: PMC10838199 DOI: 10.1007/s00018-023-05073-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
The intricate relationship between the central nervous system (CNS) and the immune system plays a crucial role in the pathogenesis of various neurological diseases. Understanding the interactions among the immunopathological processes at the brain borders is essential for advancing our knowledge of disease mechanisms and developing novel diagnostic and therapeutic approaches. In this review, we explore the emerging role of neuroimaging in providing valuable insights into brain barrier inflammation and brain fluid drainage in human neurological diseases. Neuroimaging techniques have enabled us not only to visualize and assess brain structures, but also to study the dynamics of the CNS in health and disease in vivo. By analyzing imaging findings, we can gain a deeper understanding of the immunopathology observed at the brain-immune interface barriers, which serve as critical gatekeepers that regulate immune cell trafficking, cytokine release, and clearance of waste products from the brain. This review explores the integration of neuroimaging data with immunopathological findings, providing valuable insights into brain barrier integrity and immune responses in neurological diseases. Such integration may lead to the development of novel diagnostic markers and targeted therapeutic approaches that can benefit patients with neurological disorders.
Collapse
Affiliation(s)
- Serhat V Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Francesca Fagiani
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
38
|
Coleman A, Langan MT, Verma G, Knights H, Sturrock A, Leavitt BR, Tabrizi SJ, Scahill RI, Hobbs NZ. Assessment of Perivascular Space Morphometry Across the White Matter in Huntington's Disease Using MRI. J Huntingtons Dis 2024; 13:91-101. [PMID: 38517798 DOI: 10.3233/jhd-231508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Background Perivascular spaces (PVS) are fluid-filled cavities surrounding small cerebral blood vessels. There are limited reports of enlarged PVS across the grey matter in manifest Huntington's disease (HD). Little is known about how PVS morphometry in the white matter may contribute to HD. Enlarged PVS have the potential to both contribute to HD pathology and affect the distribution and success of intraparenchymal and intrathecally administered huntingtin-lowering therapies. Objective To investigate PVS morphometry in the global white matter across the spectrum of HD. Relationships between PVS morphometry and disease burden and severity measures were examined. Methods White matter PVS were segmented on 3T T2 W MRI brain scans of 33 healthy controls, 30 premanifest HD (pre-HD), and 32 early manifest HD (early-HD) participants from the Vancouver site of the TRACK-HD study. PVS count and total PVS volume were measured. Results PVS total count slightly increased in pre-HD (p = 0.004), and early-HD groups (p = 0.005), compared to healthy controls. PVS volume, as a percentage of white matter volume, increased subtly in pre-HD compared to healthy controls (p = 0.044), but not in early-HD. No associations between PVS measures and HD disease burden or severity were found. Conclusions This study reveals relatively preserved PVS morphometry across the global white matter of pre-HD and early-HD. Subtle morphometric abnormalities are implied but require confirmation in a larger cohort. However, in conjunction with previous publications, further investigation of PVS in HD and its potential impact on future treatments, with a focus on subcortical grey matter, is warranted.
Collapse
Affiliation(s)
- Annabelle Coleman
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Mackenzie T Langan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Gaurav Verma
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Harry Knights
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Aaron Sturrock
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
39
|
Hartung HP, Cree BA, Barnett M, Meuth SG, Bar-Or A, Steinman L. Bioavailable central nervous system disease-modifying therapies for multiple sclerosis. Front Immunol 2023; 14:1290666. [PMID: 38162670 PMCID: PMC10755740 DOI: 10.3389/fimmu.2023.1290666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Disease-modifying therapies for relapsing multiple sclerosis reduce relapse rates by suppressing peripheral immune cells but have limited efficacy in progressive forms of the disease where cells in the central nervous system play a critical role. To our knowledge, alemtuzumab, fumarates (dimethyl, diroximel, and monomethyl), glatiramer acetates, interferons, mitoxantrone, natalizumab, ocrelizumab, ofatumumab, and teriflunomide are either limited to the periphery or insufficiently studied to confirm direct central nervous system effects in participants with multiple sclerosis. In contrast, cladribine and sphingosine 1-phosphate receptor modulators (fingolimod, ozanimod, ponesimod, and siponimod) are central nervous system-penetrant and could have beneficial direct central nervous system properties.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Palacký University Olomouc, Olomouc, Czechia
| | - Bruce A.C. Cree
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, United States
| | - Michael Barnett
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
40
|
Faigle W, Piccirelli M, Hortobágyi T, Frontzek K, Cannon AE, Zürrer WE, Granberg T, Kulcsar Z, Ludersdorfer T, Frauenknecht KBM, Reimann R, Ineichen BV. The Brainbox -a tool to facilitate correlation of brain magnetic resonance imaging features to histopathology. Brain Commun 2023; 5:fcad307. [PMID: 38025281 PMCID: PMC10664401 DOI: 10.1093/braincomms/fcad307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/20/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Magnetic resonance imaging (MRI) has limitations in identifying underlying tissue pathology, which is relevant for neurological diseases such as multiple sclerosis, stroke or brain tumours. However, there are no standardized methods for correlating MRI features with histopathology. Thus, here we aimed to develop and validate a tool that can facilitate the correlation of brain MRI features to corresponding histopathology. For this, we designed the Brainbox, a waterproof and MRI-compatible 3D printed container with an integrated 3D coordinate system. We used the Brainbox to acquire post-mortem ex vivo MRI of eight human brains, fresh and formalin-fixed, and correlated focal imaging features to histopathology using the built-in 3D coordinate system. With its built-in 3D coordinate system, the Brainbox allowed correlation of MRI features to corresponding tissue substrates. The Brainbox was used to correlate different MR image features of interest to the respective tissue substrate, including normal anatomical structures such as the hippocampus or perivascular spaces, as well as a lacunar stroke. Brain volume decreased upon fixation by 7% (P = 0.01). The Brainbox enabled degassing of specimens before scanning, reducing susceptibility artefacts and minimizing bulk motion during scanning. In conclusion, our proof-of-principle experiments demonstrate the usability of the Brainbox, which can contribute to improving the specificity of MRI and the standardization of the correlation between post-mortem ex vivo human brain MRI and histopathology. Brainboxes are available upon request from our institution.
Collapse
Affiliation(s)
- Wolfgang Faigle
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Marco Piccirelli
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Tibor Hortobágyi
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, WC1N 1PJ London, United Kingdom
| | - Amelia Elaine Cannon
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Wolfgang Emanuel Zürrer
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Thomas Ludersdorfer
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Katrin B M Frauenknecht
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
- Luxembourg Center of Neuropathology (LCNP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Regina Reimann
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
| | - Benjamin Victor Ineichen
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- Center for Reproducible Science, University of Zurich, CH-8001 Zurich, Switzerland
| |
Collapse
|
41
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [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: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
42
|
VanElzakker MB, Bues HF, Brusaferri L, Kim M, Saadi D, Ratai EM, Dougherty DD, Loggia ML. Neuroinflammation in post-acute sequelae of COVID-19 (PASC) as assessed by [ 11C]PBR28 PET correlates with vascular disease measures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563117. [PMID: 37905031 PMCID: PMC10614970 DOI: 10.1101/2023.10.19.563117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has triggered a consequential public health crisis of post-acute sequelae of COVID-19 (PASC), sometimes referred to as long COVID. The mechanisms of the heterogeneous persistent symptoms and signs that comprise PASC are under investigation, and several studies have pointed to the central nervous and vascular systems as being potential sites of dysfunction. In the current study, we recruited individuals with PASC with diverse symptoms, and examined the relationship between neuroinflammation and circulating markers of vascular dysfunction. We used [11C]PBR28 PET neuroimaging, a marker of neuroinflammation, to compare 12 PASC individuals versus 43 normative healthy controls. We found significantly increased neuroinflammation in PASC versus controls across a wide swath of brain regions including midcingulate and anterior cingulate cortex, corpus callosum, thalamus, basal ganglia, and at the boundaries of ventricles. We also collected and analyzed peripheral blood plasma from the PASC individuals and found significant positive correlations between neuroinflammation and several circulating analytes related to vascular dysfunction. These results suggest that an interaction between neuroinflammation and vascular health may contribute to common symptoms of PASC.
Collapse
Affiliation(s)
- Michael B VanElzakker
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- PolyBio Research Foundation, Medford, MA, USA
| | - Hannah F Bues
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Computer Science And Informatics, School of Engineering, London South Bank University, London, UK
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena Saadi
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Amin J, Gee C, Stowell K, Coulthard D, Boche D. T Lymphocytes and Their Potential Role in Dementia with Lewy Bodies. Cells 2023; 12:2283. [PMID: 37759503 PMCID: PMC10528562 DOI: 10.3390/cells12182283] [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: 08/03/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative cause of dementia. People with DLB have an inferior prognosis compared to Alzheimer's disease (AD), but the diseases overlap in their neuropathology and clinical syndrome. It is imperative that we enhance our understanding of the aetiology and pathogenesis of DLB. The impact of peripheral inflammation on the brain in dementia has been increasingly explored in recent years, with T lymphocyte recruitment into brain parenchyma identified in AD and Parkinson's disease. There is now a growing range of literature emerging on the potential role of innate and adaptive immune cells in DLB, including T lymphocytes. In this review, we examine the profile of T lymphocytes in DLB, focusing on studies of post-mortem brain tissue, cerebrospinal fluid, and the blood compartment. We present an integrated viewpoint on the results of these studies by proposing how changes to the T lymphocyte profile in the brain and periphery may relate to each other. Improving our understanding of T lymphocytes in DLB has the potential to guide the development of disease-modifying treatments.
Collapse
Affiliation(s)
- Jay Amin
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Memory Assessment and Research Centre, Tom Rudd Unit, Moorgreen Hospital, Southern Health NHS Foundation Trust, Southampton SO30 3JB, UK
| | - Claire Gee
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Memory Assessment and Research Centre, Tom Rudd Unit, Moorgreen Hospital, Southern Health NHS Foundation Trust, Southampton SO30 3JB, UK
| | - Kiran Stowell
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Daisy Coulthard
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
44
|
Laaker C, Baenen C, Kovács KG, Sandor M, Fabry Z. Immune cells as messengers from the CNS to the periphery: the role of the meningeal lymphatic system in immune cell migration from the CNS. Front Immunol 2023; 14:1233908. [PMID: 37662908 PMCID: PMC10471710 DOI: 10.3389/fimmu.2023.1233908] [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: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
In recent decades there has been a large focus on understanding the mechanisms of peripheral immune cell infiltration into the central nervous system (CNS) in neuroinflammatory diseases. This intense research led to several immunomodulatory therapies to attempt to regulate immune cell infiltration at the blood brain barrier (BBB), the choroid plexus (ChP) epithelium, and the glial barrier. The fate of these infiltrating immune cells depends on both the neuroinflammatory environment and their type-specific interactions with innate cells of the CNS. Although the fate of the majority of tissue infiltrating immune cells is death, a percentage of these cells could become tissue resident immune cells. Additionally, key populations of immune cells can possess the ability to "drain" out of the CNS and act as messengers reporting signals from the CNS toward peripheral lymphatics. Recent data supports that the meningeal lymphatic system is involved not just in fluid homeostatic functions in the CNS but also in facilitating immune cell migration, most notably dendritic cell migration from the CNS to the meningeal borders and to the draining cervical lymph nodes. Similar to the peripheral sites, draining immune cells from the CNS during neuroinflammation have the potential to coordinate immunity in the lymph nodes and thus influence disease. Here in this review, we will evaluate evidence of immune cell drainage from the brain via the meningeal lymphatics and establish the importance of this in animal models and humans. We will discuss how targeting immune cells at sites like the meningeal lymphatics could provide a new mechanism to better provide treatment for a variety of neurological conditions.
Collapse
Affiliation(s)
- Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Cameron Baenen
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Kristóf G. Kovács
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
45
|
Mehta RI, Carpenter JS, Mehta RI, Haut MW, Wang P, Ranjan M, Najib U, D'Haese PF, Rezai AR. Ultrasound-mediated blood-brain barrier opening uncovers an intracerebral perivenous fluid network in persons with Alzheimer's disease. Fluids Barriers CNS 2023; 20:46. [PMID: 37328855 DOI: 10.1186/s12987-023-00447-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is under investigation as a therapeutic modality for neurodegeneration, yet its effects in humans are incompletely understood. Here, we assessed physiologic responses to FUS administered in multifocal brain sites of persons with Alzheimer's disease (AD). METHODS At a tertiary neuroscience institute, eight participants with AD (mean age 65, 38% F) enrolled in a phase 2 clinical trial underwent three successive targeted BBB opening procedures at 2 week intervals using a 220 kHz FUS transducer in combination with systemically administered microbubbles. In all, 77 treatment sites were evaluated and encompassed hippocampal, frontal, and parietal brain regions. Post-FUS imaging changes, including susceptibility effects and spatiotemporal gadolinium-based contrast agent enhancement patterns, were analyzed using serial 3.0-Tesla MRI. RESULTS Post-FUS MRI revealed expected intraparenchymal contrast extravasation due to BBB opening at all targeted brain sites. Immediately upon BBB opening, hyperconcentration of intravenously-administered contrast tracer was consistently observed around intracerebral veins. Following BBB closure, within 24-48 h of FUS intervention, permeabilization of intraparenchymal veins was observed and persisted for up to one week. Notably, extraparenchymal meningeal venous permeabilization and associated CSF effusions were also elicited and persisted up to 11 days post FUS treatment, prior to complete spontaneous resolution in all participants. Mild susceptibility effects were detected, however no overt intracranial hemorrhage or other serious adverse effects occurred in any participant. CONCLUSIONS FUS-mediated BBB opening is safely and reproducibly achieved in multifocal brain regions of persons with AD. Post-FUS tracer enhancement phenomena suggest the existence of a brain-wide perivenous fluid efflux pathway in humans and demonstrate reactive physiological changes involving these conduit spaces in the delayed, subacute phase following BBB disruption. The delayed reactive venous and perivenous changes are consistent with a dynamic, zonal exudative response to upstream capillary manipulation. Further preclinical and clinical investigations of these FUS-related imaging phenomena and of intracerebral perivenous compartment changes are needed to elucidate physiology of this pathway as well as biological effects of FUS administered with and without adjuvant neurotherapeutics. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03671889, registered 9/14/2018.
Collapse
Affiliation(s)
- Rashi I Mehta
- Department of Neuroradiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV, 26506, USA.
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA.
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA.
| | - Jeffrey S Carpenter
- Department of Neuroradiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV, 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - Rupal I Mehta
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Marc W Haut
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, 26506, USA
- Department of Neurology, West Virginia University, Morgantown, WV, 26506, USA
| | - Peng Wang
- Department of Neuroradiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV, 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - Manish Ranjan
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Neurosurgery, West Virginia University, Morgantown, WV, 26506, USA
| | - Umer Najib
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Neurology, West Virginia University, Morgantown, WV, 26506, USA
| | | | - Ali R Rezai
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Neurosurgery, West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
46
|
Ineichen BV, Cananau C, Plattén M, Ouellette R, Moridi T, Frauenknecht KBM, Okar SV, Kulcsar Z, Kockum I, Piehl F, Reich DS, Granberg T. Dilated Virchow-Robin spaces are a marker for arterial disease in multiple sclerosis. EBioMedicine 2023; 92:104631. [PMID: 37253317 DOI: 10.1016/j.ebiom.2023.104631] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/28/2023] [Accepted: 05/11/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Virchow-Robin spaces (VRS) have been associated with neurodegeneration and neuroinflammation. However, it remains uncertain to what degree non-dilated or dilated VRS reflect specific features of neuroinflammatory pathology. Thus, we aimed at investigating the clinical relevance of VRS as imaging biomarker in multiple sclerosis (MS) and to correlate VRS to their histopathologic signature. METHODS In a cohort study comprising 142 MS patients and 30 control subjects, we assessed the association of non-dilated and dilated VRS to clinical and magnetic resonance imaging (MRI) outcomes. Findings were corroborated in a validation cohort comprising 63 MS patients. Brain blocks from 6 MS patients and 3 non-MS controls were histopathologically processed to correlate VRS to their tissue substrate. FINDINGS In our actively treated clinical cohort, the count of dilated centrum semiovale VRS was associated with increased T1 and T2 lesion volumes. There was no systematic spatial colocalization of dilated VRS with MS lesions. At tissue level, VRS mostly corresponded to arteries and were not associated with MS pathological hallmarks. Interestingly, in our ex vivo cohort comprising mostly progressive MS patients, dilated VRS in MS were associated with signs of small vessel disease. INTERPRETATION Contrary to prior beliefs, these observations suggest that VRS in MS do not associate with an accumulation of immune cells. But instead, these findings indicate vascular pathology as a driver and/or consequence of neuroinflammatory pathology for this imaging feature. FUNDING NIH, Swedish Society for Medical Research, Swiss National Science Foundation and University of Zurich.
Collapse
Affiliation(s)
- Benjamin V Ineichen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Center for Reproducible Science, University of Zurich, Zurich, Switzerland; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MA, USA.
| | - Carmen Cananau
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Plattén
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Russell Ouellette
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Moridi
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Center of Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Katrin B M Frauenknecht
- National Centre for Pathology (NCP), Laboratoire National de Santé, Dudelange, Luxembourg; Luxembourg Centre for Neuropathology (LCNP), Laboratoire National de Santé, Dudelange, Luxembourg
| | - Serhat V Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MA, USA
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Center of Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MA, USA
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Okar SV, Hu F, Shinohara RT, Beck ES, Reich DS, Ineichen BV. The etiology and evolution of magnetic resonance imaging-visible perivascular spaces: Systematic review and meta-analysis. Front Neurosci 2023; 17:1038011. [PMID: 37065926 PMCID: PMC10098201 DOI: 10.3389/fnins.2023.1038011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
ObjectivesPerivascular spaces have been involved in neuroinflammatory and neurodegenerative diseases. Upon a certain size, these spaces can become visible on magnetic resonance imaging (MRI), referred to as enlarged perivascular spaces (EPVS) or MRI-visible perivascular spaces (MVPVS). However, the lack of systematic evidence on etiology and temporal dynamics of MVPVS hampers their diagnostic utility as MRI biomarker. Thus, the goal of this systematic review was to summarize potential etiologies and evolution of MVPVS.MethodsIn a comprehensive literature search, out of 1,488 unique publications, 140 records assessing etiopathogenesis and dynamics of MVPVS were eligible for a qualitative summary. 6 records were included in a meta-analysis to assess the association between MVPVS and brain atrophy.ResultsFour overarching and partly overlapping etiologies of MVPVS have been proposed: (1) Impairment of interstitial fluid circulation, (2) Spiral elongation of arteries, (3) Brain atrophy and/or perivascular myelin loss, and (4) Immune cell accumulation in the perivascular space. The meta-analysis in patients with neuroinflammatory diseases did not support an association between MVPVS and brain volume measures [R: −0.15 (95%-CI −0.40–0.11)]. Based on few and mostly small studies in tumefactive MVPVS and in vascular and neuroinflammatory diseases, temporal evolution of MVPVS is slow.ConclusionCollectively, this study provides high-grade evidence for MVPVS etiopathogenesis and temporal dynamics. Although several potential etiologies for MVPVS emergence have been proposed, they are only partially supported by data. Advanced MRI methods should be employed to further dissect etiopathogenesis and evolution of MVPVS. This can benefit their implementation as an imaging biomarker.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?RecordID=346564, identifier CRD42022346564.
Collapse
Affiliation(s)
- Serhat V. Okar
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Fengling Hu
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Russell T. Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Erin S. Beck
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Daniel S. Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Benjamin V. Ineichen
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Reproducible Science, University of Zurich, Zurich, Switzerland
- *Correspondence: Benjamin V. Ineichen, , ; orcid.org/0000-0003-1362-4819
| |
Collapse
|
48
|
Ineichen BV, Cananau C, Platt N M, Ouellette R, Moridi T, Frauenknecht KBM, Okar SV, Kulcsar Z, Kockum I, Piehl F, Reich DS, Granberg T. Dilated Virchow-Robin Spaces are a Marker for Arterial Disease in Multiple Sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529871. [PMID: 36945422 PMCID: PMC10028816 DOI: 10.1101/2023.02.24.529871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Virchow-Robin spaces (VRS) have been associated with neurodegeneration and neuroinflammation. However, it remains uncertain to what degree non-dilated or dilated VRS reflect specific features of neuroinflammatory pathology. Thus, we aimed at investigating the clinical relevance of VRS as imaging biomarker in multiple sclerosis (MS) and to correlate VRS to their histopathologic signature. In a cohort study comprising 205 MS patients (including a validation cohort) and 30 control subjects, we assessed the association of non-dilated and dilated VRS to clinical and magnetic resonance imaging (MRI) out-comes. Brain blocks from 6 MS patients and 3 non-MS controls were histopathologically processed to correlate VRS to their tissue substrate. The count of dilated centrum semiovale VRS was associated with increased T1 and T2 lesion volumes. There was no systematic spatial colocalization of dilated VRS with MS lesions. At tissue level, VRS mostly corresponded to arteries and were not associated with MS pathological hallmarks. Interestingly, dilated VRS in MS were associated with signs of small vessel disease. Contrary to prior beliefs, these observations suggest that VRS in MS do not associate with accumulation of immune cells. But instead, these findings indicate vascular pathology as a driver and/or consequence of neuroinflammatory pathology for this imaging feature.
Collapse
|
49
|
Khan M, Clijsters M, Choi S, Backaert W, Claerhout M, Couvreur F, Van Breda L, Bourgeois F, Speleman K, Klein S, Van Laethem J, Verstappen G, Dereli AS, Yoo SJ, Zhou H, Dan Do TN, Jochmans D, Laenen L, Debaveye Y, De Munter P, Gunst J, Jorissen M, Lagrou K, Meersseman P, Neyts J, Thal DR, Topsakal V, Vandenbriele C, Wauters J, Mombaerts P, Van Gerven L. Anatomical barriers against SARS-CoV-2 neuroinvasion at vulnerable interfaces visualized in deceased COVID-19 patients. Neuron 2022; 110:3919-3935.e6. [PMID: 36446381 PMCID: PMC9647025 DOI: 10.1016/j.neuron.2022.11.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
Can SARS-CoV-2 hitchhike on the olfactory projection and take a direct and short route from the nose into the brain? We reasoned that the neurotropic or neuroinvasive capacity of the virus, if it exists, should be most easily detectable in individuals who died in an acute phase of the infection. Here, we applied a postmortem bedside surgical procedure for the rapid procurement of tissue, blood, and cerebrospinal fluid samples from deceased COVID-19 patients infected with the Delta, Omicron BA.1, or Omicron BA.2 variants. Confocal imaging of sections stained with fluorescence RNAscope and immunohistochemistry afforded the light-microscopic visualization of extracellular SARS-CoV-2 virions in tissues. We failed to find evidence for viral invasion of the parenchyma of the olfactory bulb and the frontal lobe of the brain. Instead, we identified anatomical barriers at vulnerable interfaces, exemplified by perineurial olfactory nerve fibroblasts enwrapping olfactory axon fascicles in the lamina propria of the olfactory mucosa.
Collapse
Affiliation(s)
- Mona Khan
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Marnick Clijsters
- Department of Neurosciences, Experimental Otorhinolaryngology, Rhinology Research, KU Leuven, Leuven, Belgium
| | - Sumin Choi
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Wout Backaert
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Michiel Claerhout
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Floor Couvreur
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Laure Van Breda
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Florence Bourgeois
- Department of Otorhinolaryngology, Head and Neck Surgery, AZ Sint-Jan Brugge-Oostende AV, Bruges, Belgium
| | - Kato Speleman
- Department of Otorhinolaryngology, Head and Neck Surgery, AZ Sint-Jan Brugge-Oostende AV, Bruges, Belgium
| | - Sam Klein
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Johan Van Laethem
- Department of Infectious Diseases, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gill Verstappen
- Department of Otorhinolaryngology - Head and Neck Surgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Seung-Jun Yoo
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany; Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Hai Zhou
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Thuc Nguyen Dan Do
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Dirk Jochmans
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Lies Laenen
- Department of Laboratory Medicine & National Reference Center for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
| | - Yves Debaveye
- Intensive Care Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium
| | - Paul De Munter
- Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Jan Gunst
- Intensive Care Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium
| | - Mark Jorissen
- Department of Neurosciences, Experimental Otorhinolaryngology, Rhinology Research, KU Leuven, Leuven, Belgium; Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Laboratory Medicine & National Reference Center for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Bacteriology and Mycology, KU Leuven, Leuven, Belgium
| | - Philippe Meersseman
- Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Neuropathology, Department of Imaging & Pathology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Vedat Topsakal
- Department of Otorhinolaryngology - Head and Neck Surgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christophe Vandenbriele
- Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Joost Wauters
- Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Laboratory for Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium
| | - Peter Mombaerts
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany.
| | - Laura Van Gerven
- Department of Neurosciences, Experimental Otorhinolaryngology, Rhinology Research, KU Leuven, Leuven, Belgium; Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium.
| |
Collapse
|