1
|
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
|
2
|
Manelis A, Hu H, Satz S. The Relationship Between Reduced Hand Dexterity and Brain Structure Abnormality in Older Adults. Geriatrics (Basel) 2024; 9:165. [PMID: 39727824 DOI: 10.3390/geriatrics9060165] [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: 11/12/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Hand dexterity is affected by normal aging and neuroinflammatory processes in the brain. Understanding the relationship between hand dexterity and brain structure in neurotypical older adults may be informative about prodromal pathological processes, thus providing an opportunity for earlier diagnosis and intervention to improve functional outcomes. METHODS this study investigates the associations between hand dexterity and brain measures in neurotypical older adults (≥65 years) using the Nine-Hole Peg Test (9HPT) and magnetic resonance imaging (MRI). RESULTS Elastic net regularized regression revealed that reduced hand dexterity in dominant and non-dominant hands was associated with an enlarged volume of the left choroid plexus, the region implicated in neuroinflammatory and altered myelination processes, and reduced myelin content in the left frontal operculum, the region implicated in motor imagery, action production, and higher-order motor functions. Distinct neural mechanisms underlying hand dexterity in dominant and non-dominant hands included the differences in caudate and thalamic volumes as well as altered cortical myelin patterns in frontal, temporal, parietal, and occipital regions supporting sensorimotor and visual processing and integration, attentional control, and eye movements. Although elastic net identified more predictive features for the dominant vs. non-dominant hand, the feature stability was higher for the latter, thus indicating higher generalizability for the non-dominant hand model. CONCLUSIONS Our findings suggest that the 9HPT for hand dexterity might be a cost-effective screening tool for early detection of neuroinflammatory and neurodegenerative processes. Longitudinal studies are needed to validate our findings in a larger sample and explore the potential of hand dexterity as an early clinical marker.
Collapse
Affiliation(s)
- Anna Manelis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hang Hu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Skye Satz
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
3
|
Sun Z, Li C, Zhang J, Wisniewski T, Ge Y. Choroid plexus aging: structural and vascular insights from the HCP-aging dataset. Fluids Barriers CNS 2024; 21:98. [PMID: 39639335 PMCID: PMC11619641 DOI: 10.1186/s12987-024-00603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The choroid plexus (ChP), a highly vascularized structure within the ventricles, is essential for cerebrospinal fluid (CSF) production and metabolic waste clearance, crucial for neurofluid homeostasis and cognitive function. ChP enlargement is seen in normal aging and neurodegenerative diseases like Alzheimer's disease (AD). Despite its key role of in the blood-CSF barrier (BCSFB), detailed studies on age-related changes in its perfusion and microstructure remain limited. METHODS We analyzed data from 641 healthy individuals aged between 36 and 90, using the Human Connectome Project Aging (HCP-A) dataset. Volumetric, perfusion, and diffusion metrics of the ChP were derived from structural MRI, arterial spin labeling (ASL), and diffusion-weighted imaging (DWI), respectively. Partial correlations were used to explore age-related ChP changes, and independent t-tests to examine sex differences across age decades. One-way ANOVA was employed to compare perfusion characteristics among ChP, gray matter (GM), and white matter (WM). Relationships between volume, perfusion, and diffusion were investigated, adjusting for age and sex. Additionally, the distribution of cyst-like structures within the ChP and their diffusion/perfusion MRI characteristics were analyzed across different age groups. RESULTS The ChP undergoes notable changes with age, including an increase in volume (r2 = 0.2, P < 0.001), a decrease in blood flow (r2 = 0.17, P < 0.001), and elevated mean diffusivity (MD) values (r2 = 0.16, P < 0.001). Perfusion characteristics showed significant differences between the ChP, GM, and WM (P < 0.001). Both the ChP and GM exhibited age-related declines in CBF, with a more pronounced decline in the ChP. A negative correlation was observed between the age-related increase in ChP volume and the decrease in CBF, suggesting compensatory dystrophic hyperplasia in response to perfusion decline. Cyst-like structures in ChP, characterized by lower MD and reduced CBF, were found to be more prevalent in older individuals. CONCLUSIONS Our findings provide a detailed quantitative assessment of age-related changes in ChP perfusion and diffusion, which may affect CSF production and circulation, potentially leading to waste solute accumulation and cognitive impairment. GRANT SUPPORT This work was supported in part by the NIH U01AG052564, P30AG066512, P01AG060882, RF1 NS110041, R01 NS108491, U24 NS135568.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Radiology, NYU Grossman School of Medicine, 660 First Ave, Room 405, New York, NY, 10016, USA
- Vilcek Institute of Graduate Medical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Chenyang Li
- Department of Radiology, NYU Grossman School of Medicine, 660 First Ave, Room 405, New York, NY, 10016, USA
- Vilcek Institute of Graduate Medical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Jiangyang Zhang
- Department of Radiology, NYU Grossman School of Medicine, 660 First Ave, Room 405, New York, NY, 10016, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yulin Ge
- Department of Radiology, NYU Grossman School of Medicine, 660 First Ave, Room 405, New York, NY, 10016, USA.
| |
Collapse
|
4
|
Hochstetler A, Lehtinen MK. Choroid Plexus as a Mediator of CNS Inflammation in Multiple Sclerosis. Mult Scler 2024; 30:19-23. [PMID: 39503321 PMCID: PMC11634642 DOI: 10.1177/13524585241292974] [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: 12/12/2024]
Abstract
The pathophysiology of multiple sclerosis (MS) remains poorly understood despite decades of tremendous research efforts. Advances in neuroradiography coupled with availability of unbiased approaches including spatial transcriptomics, proteomics, metabolomics, and lipidomics that are compatible with brain and fluid specimens from patients raise hope that discovery of novel disease drivers is on the horizon. Once thought to be little more than salty bathwater, our modern understanding of cerebrospinal fluid (CSF) suggests the CSF as a compelling, critical regulator of brain function in health and disease. Recent studies in the field have reinvigorated interest in CSF as a medium to better understand MS and to deliver disease-modifying therapies. In turn, the choroid plexus, an epithelial tissue located within each brain ventricle that regulates CSF and forms a key blood-CSF barrier, is uniquely positioned to orchestrate neuroinflammation associated with MS. In this perspective review, we will discuss what is known about the choroid plexus as a conductor of immune responses and how it may propagate neuroinflammation associated with MS via the CSF.
Collapse
Affiliation(s)
- Alexandra Hochstetler
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Li C, Zhang H, Wang J, Han X, Liu C, Li Y, Gong T, Hou T, Wang Y, Cong L, Kalpouzos G, Wardlaw J, Song L, Du Y, Qiu C. Choroid Plexus Volume in Rural Chinese Older Adults: Distribution and Association With Cardiovascular Risk Factors and Cerebral Small Vessel Disease. J Am Heart Assoc 2024; 13:e035941. [PMID: 39424375 DOI: 10.1161/jaha.124.035941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The choroid plexus (CP) is involved in neurodegenerative diseases. However, the association of CP with cardiovascular risk factors and cerebral small vessel disease in older adults remains unclear. METHODS AND RESULTS This population-based study included 1263 participants (60 years and older) from the MIND-China (Multimodal Interventions to Delay Dementia and Disability in Rural China) substudy (2018-2020), of which 111 individuals completed diffusion tensor imaging examination. CP volume was automatically segmented. White matter hyperintensities (WMHs), enlarged perivascular spaces (EPVS), cerebral microbleeds, and lacunes were assessed following the Standards for Reporting Vascular Changes on Neuroimaging 1. Peak width of skeletonized mean diffusivity and free water were derived from diffusion tensor imaging images. We used linear regression models to evaluate the association between CP volume and cardiovascular risk factors, WMH volumes, and diffusion tensor imaging metrics, and logistic regression models to examine the association between CP volume and EPVS, cerebral microbleeds, and lacunes. The CP volume increased with age (P<0.001). Men (β coefficient=0.47 [95% CI, 0.29-0.64]) and participants with diabetes (β coefficient=0.16 [95% CI, 0.01-0.31]) had larger CP volumes than women and individuals without diabetes, respectively (P<0.05). Greater CP volume was significantly associated with larger total and periventricular WMH volumes and moderate to severe EPVS in basal ganglia (P<0.05) but not with deep WMHs, EPVS in centrum semiovale, lacunes, or cerebral microbleeds. In the diffusion tensor imaging subsample, enlarged CP was significantly associated with higher peak width of skeletonized mean diffusivity and free water of periventricular and deep white matter (P<0.05). CONCLUSIONS An enlarged CP is associated with larger global and periventricular WMH volume and higher likelihoods of EPVS in basal ganglia and impaired white matter integrity, suggesting that an enlarged CP may represent a precursor of cerebral small vessel disease.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
| | - Huisi Zhang
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
| | - Jiafeng Wang
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases Beijing China
| | - Cuicui Liu
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
| | - Yuanjing Li
- Aging Research Center, Department of Neurobiology, Care Sciences and Society Karolinska Institutet-Stockholm University Stockholm Sweden
| | - Tao Gong
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
| | - Tingting Hou
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
| | - Yongxiang Wang
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong China
| | - Lin Cong
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
| | - Grégoria Kalpouzos
- Aging Research Center, Department of Neurobiology, Care Sciences and Society Karolinska Institutet-Stockholm University Stockholm Sweden
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences Edinburgh Centre in the UK Dementia Research Institute, University of Edinburgh Edinburgh UK
| | - Lin Song
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
| | - Yifeng Du
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Department of Neurology, Shandong Provincial Hospital Shandong University Jinan Shandong China
- Shandong Provincial Clinical Research Center for Neurological Diseases Jinan Shandong China
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong China
| | - Chengxuan Qiu
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society Karolinska Institutet-Stockholm University Stockholm Sweden
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong China
| |
Collapse
|
6
|
Gu L, Chen H, Geng R, Liang T, Chen Y, Wang Z, Ye L, Sun M, Shi Q, Wan G, Chang J, Wei J, Ma W, Xiao J, Bao X, Wang R. Endothelial pyroptosis-driven microglial activation in choroid plexus mediates neuronal apoptosis in hemorrhagic stroke rats. Neurobiol Dis 2024; 201:106695. [PMID: 39370051 DOI: 10.1016/j.nbd.2024.106695] [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: 07/10/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Spontaneous intracerebral hemorrhage (ICH) is associated with alarmingly high rates of disability and mortality, and current therapeutic options are suboptimal. A critical component of ICH pathology is the initiation of a robust inflammatory response, often termed "cytokine storm," which amplifies the secondary brain injury following the initial hemorrhagic insult. The precise sources and consequences of this cytokine-driven inflammation are not fully elucidated, necessitating further investigation. METHODS To address this knowledge gap, our study conducted a comprehensive cytokine profiling using Luminex® assays, assessing 23 key cytokines. We then employed single-cell RNA sequencing and spatial transcriptomics at three critical time points post-ICH: the hyperacute, acute, and subacute phases. Integrating these multimodal analyses allowed us to identify the cellular origins of cytokines and elucidate their mechanisms of action. RESULTS Luminex® cytokine assays revealed a significant upregulation of IL-6 and IL-1β levels at the 24-h post-ICH time point. Through the integration of scRNA-seq and spatial transcriptomics in the hemorrhagic hemisphere of rats, we observed a pronounced activation of cytokine-related signaling pathways within the choroid plexus. Initially, immune cell presence was sparse, but it surged 24 h post-ICH, particularly in the choroid plexus, indicating a substantial shift in the immune microenvironment. We traced the source of IL-1β and IL-6 to endothelial cells, establishing a link to pyroptosis. Endothelial pyroptosis post-ICH induced the production of IL-1β and IL-6, which activated microglial polarization characterized by elevated expression of Msr1, Lcn2, and Spp1 via the NF-κB pathway in the choroid plexus. Furthermore, we identified neuronal populations undergoing apoptosis, mediated by the Lcn2-SLC22A17 pathway in response to IL-1β and IL-6 signaling. Notably, the inhibition of pyroptosis using VX-765 significantly mitigated neurological impairments. CONCLUSIONS Our study provides evidence that endothelial pyroptosis, characterized by the release of IL-1β and IL-6, triggers microglial polarization through NF-κB pathway activation, ultimately leading to microglia-mediated neuronal apoptosis in the choroid plexus post-ICH. These findings suggest that targeted therapeutic strategies aimed at mitigating endothelial cell pyroptosis and neutralizing inflammatory cytokines may offer neuroprotection for both microglia and neurons, presenting a promising avenue for ICH treatment.
Collapse
Affiliation(s)
- Lingui Gu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Hualin Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Ruxu Geng
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Tingyu Liang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yihao Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhuo Wang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, PR China.
| | - Liguo Ye
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Mingjiang Sun
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Qinglei Shi
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China; Shenzhen Research Institute of Big Data, Shenzhen 518172, China
| | - Gui Wan
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianbo Chang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Junji Wei
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Wenbin Ma
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China.
| | - Xinjie Bao
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China.
| | - Renzhi Wang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China.
| |
Collapse
|
7
|
Lee DA, Lee HJ, Jo G, Park KM. Choroid plexus volumes in patients with transient global amnesia: A retrospective study. Medicine (Baltimore) 2024; 103:e40077. [PMID: 39465804 PMCID: PMC11479460 DOI: 10.1097/md.0000000000040077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Increased choroid plexus (ChP) volume is well known to be associated with glymphatic system dysfunction. This study aimed to investigate glymphatic system function in patients with transient global amnesia (TGA) compared to healthy controls through ChP volumes measurements. We retrospectively enrolled patients with TGA from our hospital, as well as healthy controls. This was retrospectively observational study followed STROBE guideline. All participants underwent brain magnetic resonance imaging, including three-dimensional T1-weighted imaging. We analyzed and compared ChP volumes between patients with TGA and healthy controls and investigated the relationship between ChP volumes and clinical characteristics in patients with TGA. We enrolled 44 patients with TGA and 47 healthy controls. Among the 44 patients with TGA, 38 experienced a single TGA event, while 6 had recurrent TGA events. ChP volumes did not significantly differ between patients with TGA and healthy controls (2.140% vs 2.089%, P = .568). However, ChP volumes were higher in patients with a single TGA event compared to those with recurrent events (2.204% vs 1.740%, P < .013). We observed a significant positive correlation between ChP volumes and age in patients with TGA (R = 0.282, P = .007). ChP volumes were not associated with the duration of amnesia in patients with TGA (R = 0.187, P = .274). We find no differences in the glymphatic system function, as demonstrated by ChP volume for the first time. This study also found a significant correlation between ChP volume and age in patients with TGA, indicating that aging influences glymphatic system function.
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
| | - Geunyeol Jo
- Department of Rehabilitation Medicine, Haeundae 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
|
8
|
McCarthy S, Villarreal N, Sailor M. Acute Streptococcus pneumoniae Meningitis: A Case Report. AACN Adv Crit Care 2024; 35:244-250. [PMID: 39213628 DOI: 10.4037/aacnacc2024820] [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: 09/04/2024]
Abstract
A 69-year-old woman without significant medical history presented to an emergency department for evaluation and management of altered mental status and a 10-day history of worsening symptoms of upper respiratory infection. Two days previously, she had been evaluated at an urgent care center, where she reported productive cough and neck pain. Evaluation in the emergency department aroused suspicions of sepsis and meningitis, and computed tomography of the head revealed nontraumatic pneumocephalus with evidence of bony erosion of the sinus into the brain. Culture results revealed disseminated Streptococcus pneumoniae. Cerebral vasculopathy secondary to the meningitis caused bilateral acute ischemic strokes in areas of the brain, with the potential to lead to significant disability.
Collapse
Affiliation(s)
- Sarah McCarthy
- Sarah McCarthy is Adjunct Clinical Faculty, University of Minnesota School of Nursing, Adult and Gerontological Health Cooperative, School of Nursing, 5-140 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN 55455
| | - Nicholas Villarreal
- Nicholas Villarreal is Nurse Practitioner, University of Minnesota Physicians, Department of Cardiology, Minneapolis, Minnesota
| | - Mandy Sailor
- Mandy Sailor is a nurse practitioner student, Georgetown University, Washington, DC
| |
Collapse
|
9
|
Schwerk C, Schroten H. In vitro models of the choroid plexus and the blood-cerebrospinal fluid barrier: advances, applications, and perspectives. Hum Cell 2024; 37:1235-1242. [PMID: 39103559 PMCID: PMC11341628 DOI: 10.1007/s13577-024-01115-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The choroid plexus (CP), a highly vascularized endothelial-epithelial convolute, is placed in the ventricular system of the brain and produces a large part of the cerebrospinal fluid (CSF). Additionally, the CP is the location of a blood-CSF barrier (BCSFB) that separates the CSF from the blood stream in the CP endothelium. In vitro models of the CP and the BCSFB are of high importance to investigate the biological functions of the CP and the BCSFB. Since the CP is involved in several serious diseases, these in vitro models promise help in researching the processes contributing to the diseases and during the development of treatment options. In this review, we provide an overview on the available models and the advances that have been made toward more sophisticated and "in vivo near" systems as organoids and microfluidic lab-on-a-chip approaches. We go into the applications and research objectives for which the various modeling systems can be used and discuss the possible future prospects and perspectives.
Collapse
Affiliation(s)
- Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| |
Collapse
|
10
|
Huang C, Hoque MT, Qu QR, Henderson J, Bendayan R. Antiretroviral drug dolutegravir induces inflammation at the mouse brain barriers. FASEB J 2024; 38:e23790. [PMID: 38982638 DOI: 10.1096/fj.202400558r] [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: 03/12/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Integrase strand transfer inhibitors (INSTIs) based antiretroviral therapy (ART) is currently used as first-line regimen to treat HIV infection. Despite its high efficacy and barrier to resistance, ART-associated neuropsychiatric adverse effects remain a major concern. Recent studies have identified a potential interaction between the INSTI, dolutegravir (DTG), and folate transport pathways at the placental barrier. We hypothesized that such interactions could also occur at the two major blood-brain interfaces: blood-cerebrospinal fluid barrier (BCSFB) and blood-brain barrier (BBB). To address this question, we evaluated the effect of two INSTIs, DTG and bictegravir (BTG), on folate transporters and receptor expression at the mouse BCSFB and the BBB in vitro, ex vivo and in vivo. We demonstrated that DTG but not BTG significantly downregulated the mRNA and/or protein expression of folate transporters (RFC/SLC19A1, PCFT/SLC46A1) in human and mouse BBB models in vitro, and mouse brain capillaries ex vivo. Our in vivo study further revealed a significant downregulation in Slc19a1 and Slc46a1 mRNA expression at the BCSFB and the BBB following a 14-day DTG oral treatment in C57BL/6 mice. However, despite the observed downregulatory effect of DTG in folate transporters/receptor at both brain barriers, a 14-day oral treatment of DTG-based ART did not significantly alter the brain folate level in animals. Interestingly, DTG treatment robustly elevated the mRNA and/or protein expression of pro-inflammatory cytokines and chemokines (Cxcl1, Cxcl2, Cxcl3, Il6, Il23, Il12) in primary cultures of mouse brain microvascular endothelial cells (BBB). DTG oral treatment also significantly upregulated proinflammatory cytokines and chemokine (Il6, Il1β, Tnfα, Ccl2) at the BCSFB in mice. We additionally observed a downregulated mRNA expression of drug efflux transporters (Abcc1, Abcc4, and Abcb1a) and tight junction protein (Cldn3) at the CP isolated from mice treated with DTG. Despite the structural similarities, BTG only elicited minor effects on the markers of interest at both the BBB and BCSFB. In summary, our current data demonstrates that DTG but not BTG strongly induced inflammatory responses in a rodent BBB and BCSFB model. Together, these data provide valuable insights into the mechanism of DTG-induced brain toxicity, which may contribute to the pathogenesis of DTG-associated neuropsychiatric adverse effect.
Collapse
Affiliation(s)
- Chang Huang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Md Tozammel Hoque
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Qing Rui Qu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey Henderson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Prapas P, Anagnostouli M. Macrophages and HLA-Class II Alleles in Multiple Sclerosis: Insights in Therapeutic Dynamics. Int J Mol Sci 2024; 25:7354. [PMID: 39000461 PMCID: PMC11242320 DOI: 10.3390/ijms25137354] [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: 06/15/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.
Collapse
Affiliation(s)
- Petros Prapas
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
| | - Maria Anagnostouli
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
- Multiple Sclerosis and Demyelinating Diseases Unit, Center of Expertise for Rare Demyelinating and Autoimmune Diseases of CNS, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens NKUA, Aeginition University Hospital, Vas. Sofias 72-74, 11528 Athens, Greece
| |
Collapse
|
12
|
Bergsland N, Dwyer MG, Jakimovski D, Tavazzi E, Weinstock-Guttman B, Zivadinov R. Choroid plexus enlargement is associated with future periventricular neurodegeneration in multiple sclerosis. Mult Scler Relat Disord 2024; 87:105668. [PMID: 38744032 DOI: 10.1016/j.msard.2024.105668] [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: 02/02/2024] [Revised: 04/10/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND The choroid plexus (CP), located within the ventricles of the brain and the primary producer of cerebrospinal fluid, has been shown to be enlarged in patients with multiple sclerosis (MS) and linked to periventricular remyelination failure. Atrophied T2-lesion volume (aT2-LV), a promising neurodegenerative imaging marker in progressive MS (PMS), reflects the volume of periventricular lesions subsumed into cerebrospinal fluid over the follow-up. METHODS In a cohort of 143 people with relapsing-remitting MS (RRMS) and 53 with PMS, we used 3T magnetic resonance imaging (MRI) to quantify CP volume (CPV) at baseline and aT2-LV over an average of 5.4 years of follow-up. Partial correlations, adjusting for age and sex, and linear regression analyses were used to assess the relationships between imaging measures. RESULTS In both cohorts, CPV was associated with aT2-LV in both the RRMS group (r = 0.329, p < 0.001) as well as the PMS group (r = 0.522, p < 0.001). In regression analyses predicting aT2-LV, ventricular volume (final adjusted R2 = 0.407, p < 0.001) explained additional variance beyond age, sex, and T2-lesion volume in the RRMS group while CPV (final adjusted R2 = 0.446, p = 0.009) was retained in the PMS group. CONCLUSION Findings from this study suggest that the CP enlargement is associated with future neurodegeneration, with a particularly relevant role in PMS.
Collapse
Affiliation(s)
- Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Eleonora Tavazzi
- Multiple Sclerosis Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
13
|
O’Hara BA, Lukacher AS, Garabian K, Kaiserman J, MacLure E, Ishikawa H, Schroten H, Haley SA, Atwood WJ. Highly restrictive and directional penetration of the blood cerebral spinal fluid barrier by JCPyV. PLoS Pathog 2024; 20:e1012335. [PMID: 39038049 PMCID: PMC11293668 DOI: 10.1371/journal.ppat.1012335] [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: 03/12/2024] [Revised: 08/01/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024] Open
Abstract
The human polyomavirus JCPyV is an opportunistic pathogen that infects greater than 60% of the world's population. The virus establishes a persistent and asymptomatic infection in the urogenital system but can cause a fatal demyelinating disease in immunosuppressed or immunomodulated patients following invasion of the CNS. The mechanisms responsible for JCPyV invasion into CNS tissues are not known but direct invasion from the blood to the cerebral spinal fluid via the choroid plexus has been hypothesized. To study the potential of the choroid plexus as a site of neuroinvasion, we used an adult human choroid plexus epithelial cell line to model the blood-cerebrospinal fluid (B-CSF) barrier in a transwell system. We found that these cells formed a highly restrictive barrier to virus penetration either as free virus or as virus associated with extracellular vesicles (EVJC+). The restriction was not absolute and small amounts of virus or EVJC+ penetrated and were able to establish foci of infection in primary astrocytes. Disruption of the barrier with capsaicin did not increase virus or EVJC+ penetration leading us to hypothesize that virus and EVJC+ were highly cell-associated and crossed the barrier by an active process. An inhibitor of macropinocytosis increased virus penetration from the basolateral (blood side) to the apical side (CSF side). In contrast, inhibitors of clathrin and raft dependent transcytosis reduced virus transport from the basolateral to the apical side of the barrier. None of the drugs inhibited apical to basolateral transport suggesting directionality. Pretreatment with cyclosporin A, an inhibitor of P-gp, MRP2 and BCRP multidrug resistance transporters, restored viral penetration in cells treated with raft and clathrin dependent transcytosis inhibitors. Because choroid plexus epithelial cells are known to be susceptible to JCPyV infection both in vitro and in vivo we also examined the release of infectious virus from the barrier. We found that virus was preferentially released from the cells into the apical (CSF) chamber. These data show clearly that there are two mechanisms of penetration, direct transcytosis which is capable of seeding the CSF with small amounts of virus, and infection followed by directional release of infectious virions into the CSF compartment.
Collapse
Affiliation(s)
- Bethany A. O’Hara
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Avraham S. Lukacher
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Kaitlin Garabian
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Jacob Kaiserman
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Evan MacLure
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | | | - Horst Schroten
- Department of Pediatrics, Medical Faculty Mannheim, Mannheim, Germany
| | - Sheila A. Haley
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Walter J. Atwood
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
14
|
Haley SA, O'Hara BA, Schorl C, Atwood WJ. JCPyV infection of primary choroid plexus epithelial cells reduces expression of critical junctional proteins and increases expression of barrier disrupting inflammatory cytokines. Microbiol Spectr 2024; 12:e0062824. [PMID: 38874395 PMCID: PMC11302677 DOI: 10.1128/spectrum.00628-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024] Open
Abstract
The human polyomavirus, JCPyV, establishes a lifelong persistent infection in the peripheral organs of a majority of the human population worldwide. Patients who are immunocompromised due to underlying infections, cancer, or to immunomodulatory treatments for autoimmune disease are at risk for developing progressive multifocal leukoencephalopathy (PML) when the virus invades the CNS and infects macroglial cells in the brain parenchyma. It is not yet known how the virus enters the CNS to cause disease. The blood-choroid plexus barrier is a potential site of virus invasion as the cells that make up this barrier are known to be infected with virus both in vivo and in vitro. To understand the effects of virus infection on these cells we challenged primary human choroid plexus epithelial cells with JCPyV and profiled changes in host gene expression. We found that viral infection induced the expression of proinflammatory chemokines and downregulated junctional proteins essential for maintaining blood-CSF and blood-brain barrier function. These data contribute to our understanding of how JCPyV infection of the choroid plexus can modulate the host cell response to neuroinvasive pathogens. IMPORTANCE The human polyomavirus, JCPyV, causes a rapidly progressing demyelinating disease in the CNS of patients whose immune systems are compromised. JCPyV infection has been demonstrated in the choroid plexus both in vivo and in vitro and this highly vascularized organ may be important in viral invasion of brain parenchyma. Our data show that infection of primary choroid plexus epithelial cells results in increased expression of pro-inflammatory chemokines and downregulation of critical junctional proteins that maintain the blood-CSF barrier. These data have direct implications for mechanisms used by JCPyV to invade the CNS and cause neurological disease.
Collapse
Affiliation(s)
- Sheila A. Haley
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Bethany A. O'Hara
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Christoph Schorl
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Walter J. Atwood
- Department of Cell Biology, Biochemistry, and Molecular Biology, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
15
|
Shkundin A, Halaris A. IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions. J Pers Med 2024; 14:488. [PMID: 38793070 PMCID: PMC11122344 DOI: 10.3390/jpm14050488] [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: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Interleukin-8 (IL-8/CXCL8), an essential CXC chemokine, significantly influences psychoneuroimmunological processes and affects neurological and psychiatric health. It exerts a profound effect on immune cell activation and brain function, suggesting potential roles in both neuroprotection and neuroinflammation. IL-8 production is stimulated by several factors, including reactive oxygen species (ROS) known to promote inflammation and disease progression. Additionally, CXCL8 gene polymorphisms can alter IL-8 production, leading to potential differences in disease susceptibility, progression, and severity across populations. IL-8 levels vary among neuropsychiatric conditions, demonstrating sensitivity to psychosocial stressors and disease severity. IL-8 can be detected in blood circulation, cerebrospinal fluid (CSF), and urine, making it a promising candidate for a broad-spectrum biomarker. This review highlights the need for further research on the diverse effects of IL-8 and the associated implications for personalized medicine. A thorough understanding of its complex role could lead to the development of more effective and personalized treatment strategies for neuropsychiatric conditions.
Collapse
Affiliation(s)
| | - Angelos Halaris
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA;
| |
Collapse
|
16
|
Storelli L, Pagani E, Rubin M, Margoni M, Filippi M, Rocca MA. A Fully Automatic Method to Segment Choroid Plexuses in Multiple Sclerosis Using Conventional MRI Sequences. J Magn Reson Imaging 2024; 59:1643-1652. [PMID: 37530734 DOI: 10.1002/jmri.28937] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Choroid plexus (CP) volume has been recently proposed as a proxy for brain neuroinflammation in multiple sclerosis (MS). PURPOSE To develop and validate a fast automatic method to segment CP using routinely acquired brain T1-weighted and FLAIR MRI. STUDY TYPE Retrospective. POPULATION Fifty-five MS patients (33 relapsing-remitting, 22 progressive; mean age = 46.8 ± 10.2 years; 31 women) and 60 healthy controls (HC; mean age = 36.1 ± 12.6 years, 33 women). FIELD STRENGTH/SEQUENCE 3D T2-weighted FLAIR and 3D T1-weighted gradient echo sequences at 3.0 T. ASSESSMENT Brain tissues were segmented on T1-weighted sequences and a Gaussian Mixture Model (GMM) was fitted to FLAIR image intensities obtained from the ventricle masks of the SIENAX. A second GMM was then applied on the thresholded and filtered ventricle mask. CP volumes were automatically determined and compared with those from manual segmentation by two raters (with 3 and 10 years' experience; reference standard). CP volumes from previously published automatic segmentation methods (freely available Freesurfer [FS] and FS-GMM) were also compared with reference standard. Expanded Disability Status Scale (EDSS) score was assessed within 3 days of MRI. Computational time was assessed for each automatic technique and manual segmentation. STATISTICAL TESTS Comparisons of CP volumes with reference standard were evaluated with Bland Altman analysis. Dice similarity coefficients (DSC) were computed to assess automatic CP segmentations. Volume differences between MS and HC for each method were assessed with t-tests and correlations of CP volumes with EDSS were assessed with Pearson's correlation coefficients (R). A P value <0.05 was considered statistically significant. RESULTS Compared to manual segmentation, the proposed method had the highest segmentation accuracy (mean DSC = 0.65 ± 0.06) compared to FS (mean DSC = 0.37 ± 0.08) and FS-GMM (0.58 ± 0.06). The percentage CP volume differences relative to manual segmentation were -0.1% ± 0.23, 4.6% ± 2.5, and -0.48% ± 2 for the proposed method, FS, and FS-GMM, respectively. The Pearson's correlations between automatically obtained CP volumes and the manually obtained volumes were 0.70, 0.54, and 0.56 for the proposed method, FS, and FS-GMM, respectively. A significant correlation between CP volume and EDSS was found for the proposed automatic pipeline (R = 0.2), for FS-GMM (R = 0.3) and for manual segmentation (R = 0.4). Computational time for the proposed method (32 ± 2 minutes) was similar to the manual segmentation (20 ± 5 minutes) but <25% of the FS (120 ± 15 minutes) and FS-GMM (125 ± 15 minutes) methods. DATA CONCLUSION This study developed an accurate and easily implementable method for automatic CP segmentation in MS using T1-weighted and FLAIR MRI. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Loredana Storelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Rubin
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
17
|
Manelis A, Hu H, Miceli R, Satz S, Lau R, Iyengar S, Swartz HA. The relationship between the size and asymmetry of the lateral ventricles and cortical myelin content in individuals with mood disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.30.24306621. [PMID: 38746112 PMCID: PMC11092679 DOI: 10.1101/2024.04.30.24306621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Although enlargement of the lateral ventricles was previously observed in individuals with mood disorders, the link between ventricular size and asymmetry with other indices of brain structure remains underexplored. In this study, we examined the association of lateral ventricular size and asymmetry with cortical myelin content in individuals with bipolar (BD) and depressive (DD) disorders compared to healthy controls (HC). Methods Magnetic resonance imaging (MRI) was used to obtain T1w and T2w images from 149 individuals (age=27.7 (SD=6.1) years, 78% female, BD=38, DD=57, HC=54). Cortical myelin content was calculated using the T1w/T2w ratio. Elastic net regularized regression identified brain regions whose myelin content was associated with ventricular size and asymmetry. A post-hoc linear regression examined how participants' diagnosis, illness duration, and current level of depression moderated the relationship between the size and asymmetry of the lateral ventricles and levels of cortical myelin in the selected brain regions. Results Individuals with mood disorders had larger lateral ventricles than HC. Larger ventricles and lower asymmetry were observed in individuals with BD who had longer lifetime illness duration and more severe current depressive symptoms. A greater left asymmetry was observed in participants with DD than in those with BD (p<0.01). Elastic net revealed that both ventricular enlargement and asymmetry were associated with altered myelin content in cingulate, frontal, and sensorimotor cortices. In BD, but not other groups, ventricular enlargement was related to altered myelin content in the right insular regions. Conclusions Lateral ventricular enlargement and asymmetry are linked to myelin content imbalance, thus, potentially leading to emotional and cognitive dysfunction in mood disorders.
Collapse
|
18
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
19
|
Qiao H, Deng X, Qiu L, Qu Y, Chiu Y, Chen F, Xia S, Muenzel C, Ge T, Zhang Z, Song P, Bonnin A, Zhao Z, Yuan W. SARS-CoV-2 induces blood-brain barrier and choroid plexus barrier impairments and vascular inflammation in mice. J Med Virol 2024; 96:e29671. [PMID: 38747003 PMCID: PMC11446308 DOI: 10.1002/jmv.29671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 07/18/2024]
Abstract
The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.
Collapse
Affiliation(s)
- Haowen Qiao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Xiangxue Deng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Lingxi Qiu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yafei Qu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yuanpu Chiu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Feixiang Chen
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Cheyene Muenzel
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Tenghuan Ge
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zixin Zhang
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Pengfei Song
- Department of Electrical and Computer Engineering, the Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana–Champaign, Urbana, IL, USA
| | - Alexandre Bonnin
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| |
Collapse
|
20
|
Wan Y, Fu X, Zhang T, Hua Y, Keep RF, Xi G. Choroid plexus immune cell response in murine hydrocephalus induced by intraventricular hemorrhage. Fluids Barriers CNS 2024; 21:37. [PMID: 38654318 PMCID: PMC11036653 DOI: 10.1186/s12987-024-00538-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Intraventricular hemorrhage (IVH) and associated hydrocephalus are significant complications of intracerebral and subarachnoid hemorrhage. Despite proximity to IVH, the immune cell response at the choroid plexus (ChP) has been relatively understudied. This study employs CX3CR-1GFP mice, which marks multiple immune cell populations, and immunohistochemistry to outline that response. METHODS This study had four parts all examining male adult CX3CR-1GFP mice. Part 1 examined naïve mice. In part 2, mice received an injection 30 µl of autologous blood into right ventricle and were euthanized at 24 h. In part 3, mice underwent intraventricular injection of saline, iron or peroxiredoxin 2 (Prx-2) and were euthanized at 24 h. In part 4, mice received intraventricular iron injection and were treated with either control or clodronate liposomes and were euthanized at 24 h. All mice underwent magnetic resonance imaging to quantify ventricular volume. The ChP immune cell response was examined by combining analysis of GFP(+) immune cells and immunofluorescence staining. RESULTS IVH and intraventricular iron or Prx-2 injection in CX3CR-1GFP mice all induced ventriculomegaly and activation of ChP immune cells. There were very marked increases in the numbers of ChP epiplexus macrophages, T lymphocytes and neutrophils. Co-injection of clodronate liposomes with iron reduced the ventriculomegaly which was associated with fewer epiplexus and stromal macrophages but not reduced T lymphocytes and neutrophils. CONCLUSION There is a marked immune cell response at the ChP in IVH involving epiplexus cells, T lymphocytes and neutrophils. The blood components iron and Prx-2 may play a role in eliciting that response. Reduction of ChP macrophages with clodronate liposomes reduced iron-induced ventriculomegaly suggesting that ChP macrophages may be a promising therapeutic target for managing IVH-induced hydrocephalus.
Collapse
Affiliation(s)
- Yingfeng Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
- R5018 Biomedical Science Research Building, University of Michigan, 109 Zina Pitcher Place, 48109-2200, Ann Arbor, MI, USA.
| | - Xiongjie Fu
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Tianjie Zhang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Andravizou A, Stavropoulou De Lorenzo S, Kesidou E, Michailidou I, Parissis D, Boziki MK, Stamati P, Bakirtzis C, Grigoriadis N. The Time Trajectory of Choroid Plexus Enlargement in Multiple Sclerosis. Healthcare (Basel) 2024; 12:768. [PMID: 38610190 PMCID: PMC11011748 DOI: 10.3390/healthcare12070768] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Choroid plexus (CP) can be seen as a watchtower of the central nervous system (CNS) that actively regulates CNS homeostasis. A growing body of literature suggests that CP alterations are involved in the pathogenesis of multiple sclerosis (MS) but the underlying mechanisms remain elusive. CPs are enlarged and inflamed in relapsing-remitting (RRMS) but also in clinically isolated syndrome (CIS) and radiologically isolated syndrome (RIS) stages, far beyond MS diagnosis. Increases in the choroid plexus/total intracranial volume (CP/TIV) ratio have been robustly associated with increased lesion load, higher translocator protein (TSPO) uptake in normal-appearing white matter (NAWM) and thalami, as well as with higher annual relapse rate and disability progression in highly active RRMS individuals, but not in progressive MS. The CP/TIV ratio has only slightly been correlated with magnetic resonance imaging (MRI) findings (cortical or whole brain atrophy) and clinical outcomes (EDSS score) in progressive MS. Therefore, we suggest that plexus volumetric assessments should be mainly applied to the early disease stages of MS, whereas it should be taken into consideration with caution in progressive MS. In this review, we attempt to clarify the pathological significance of the temporal CP volume (CPV) changes in MS and highlight the pitfalls and limitations of CP volumetric analysis.
Collapse
Affiliation(s)
- Athina Andravizou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Sotiria Stavropoulou De Lorenzo
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Evangelia Kesidou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Iliana Michailidou
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Dimitrios Parissis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Marina-Kleopatra Boziki
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, 41334 Larissa, Greece;
| | - Christos Bakirtzis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| | - Nikolaos Grigoriadis
- Multiple Sclerosis Center, Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece; (A.A.); (S.S.D.L.); (E.K.); (I.M.); (D.P.); (M.-K.B.); (N.G.)
| |
Collapse
|
22
|
Pu X, Bu W, Qin Y, Wang C, Xu L, Fang M, Ji Q, Wang H, Shao M. Activation and functional modification of mucosal-associated invariant T cells in patients with intracranial infection following craniotomy. Int Immunopharmacol 2024; 130:111699. [PMID: 38377855 DOI: 10.1016/j.intimp.2024.111699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Intracranial infections are among the most common complications of neurosurgery, with their incidence remaining high despite advancements in current neurosurgical techniques and aseptic technology. While the role of mucosal-associated invariant T (MAIT) cells, a subset of innate-like T lymphocytes, in bacterial defense is well-established, their involvement in intracranial infections remains unclear. In this study, we utilized flow cytometry to assess the phenotype and function of circulating and CSF MAIT cells. Our findings revealed that MAIT cells were higher in the CSF compared to blood. Notably, a higher percentage of IL-17A + MAIT cells was detected in the CSF of patients with intracranial infections. Moreover, markers indicating activation and exhaustion were significantly upregulated in CSF MAIT cells. Furthermore, elevated levels of pro-inflammatory cytokines, including IL-1β, IL-12, and IL-18, were detected in the CSF supernatants. We hypothesized that the elevated levels of IL-1β, IL-12, and IL-18 in the inflammatory milieu synergistically activate MAIT cells in the CSF. In particular, CD25 and Tim-3 expression of MAIT cells was increased by stimulation with IL-1β, IL-12, and IL-18 or CSF supernatants of intracranial infection patients. Collectively, these findings provide important information underlying the innate immune response of patients with intracranial infections.
Collapse
Affiliation(s)
- Xuexue Pu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Wei Bu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Yu Qin
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Cui Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Lunbing Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Ming Fang
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Qiang Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, Anhui, China.
| | - Min Shao
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
23
|
Bonifacio C, Savini G, Reca C, Garoli F, Levi R, Vatteroni G, Balzarini L, Allocca M, Furfaro F, Dal Buono A, Armuzzi A, Danese S, Matteoli M, Rescigno M, Fiorino G, Politi LS. The gut-brain axis: Correlation of choroid plexus volume and permeability with inflammatory biomarkers in Crohn's disease. Neurobiol Dis 2024; 192:106416. [PMID: 38272141 DOI: 10.1016/j.nbd.2024.106416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The dysregulation of the gut-brain axis in chronic inflammatory bowel diseases can cause neuro-psychological disturbances, but the underlying mechanisms are still not fully understood. The choroid plexus (CP) maintains brain homeostasis and nourishment through the secretion and clearance of cerebrospinal fluid. Recent research has demonstrated the existence of a CP vascular barrier in mice which is modulated during intestinal inflammation. This study investigates possible correlations between CP modifications and inflammatory activity in patients with Crohn's disease (CD). METHODS In this prospective study, 17 patients with CD underwent concomitant abdominal and brain 3 T MRI. The volume and permeability of CP were compared with levels of C-reactive protein (CRP), fecal calprotectin (FC), sMARIA and SES-CD scores. RESULTS The CP volume was negatively correlated with CRP levels (R = -0.643, p-value = 0.024) and FC (R = -0.571, p-value = 0.050). DCE metrics normalized by CP volume were positively correlated with CRP (K-trans: R = 0.587, p-value = 0.045; Vp: R = 0.706, p-value = 0.010; T1: R = 0.699, p-value = 0.011), and FC (Vp: R = 0.606, p-value = 0.037). CONCLUSIONS Inflammatory activity in patients with CD is associated with changes in CP volume and permeability, thus supporting the hypothesis that intestinal inflammation could affect the brain through the modulation of CP vascular barrier also in humans.
Collapse
Affiliation(s)
- Cristiana Bonifacio
- Radiology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Giovanni Savini
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Christian Reca
- Radiology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
| | - Federico Garoli
- Radiology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
| | - Riccardo Levi
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Giulia Vatteroni
- Radiology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
| | - Luca Balzarini
- Radiology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Mariangela Allocca
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Furfaro
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Arianna Dal Buono
- IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Alessandro Armuzzi
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; IBD Center, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy; Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Maria Rescigno
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Gionata Fiorino
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy; Gastroenterology and Digestive Endoscopy, San Camillo-Forlanini Hospital, Rome, Italy
| | - Letterio S Politi
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
24
|
Raghib MF, Bao F, Elkhooly M, Bernitsas E. Choroid plexus volume as a marker of retinal atrophy in relapsing remitting multiple sclerosis. J Neurol Sci 2024; 457:122884. [PMID: 38237367 DOI: 10.1016/j.jns.2024.122884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To evaluate choroid plexus (CP) volume as a biomarker for predicting clinical disability and retinal layer atrophy in relapsing remitting multiple sclerosis (RRMS). METHODS Ninety-five RRMS patients and 26 healthy controls (HCs) underwent 3 T whole brain MRI, expanded disability status scale (EDSS) and optical coherence tomography (OCT). Fully automated intra-retinal segmentation was performed to obtain the volumes of the retinal nerve fiber layer (RNFL), combined ganglion cell layer -inner plexiform layer (GCIPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), retinal pigment epithelium (RPE), total macular volume (TMV) and papillomacular bundle (PMB). Automated segmentation of the CP within the lateral ventricles was performed and the choroid plexus volume (CPV) was normalized by total intracranial volume (TIV). Linear regression analysis and generalized estimating equation (GEE) models were applied to evaluate relationships between nCPV and EDSS, T2 lesion volume, disease duration, and retinal layer volumes, followed by Bonferroni correction analysis for multiple comparisons. RESULTS RRMS patients had larger tChPV compared to HCs (p < 0.001). After Bonferroni correction, there was a significant positive correlation between tChPV and EDSS (r2 = 0.25, p = 0.0002), disease duration (r2 = 0.30, p = 0.01), and T2 lesion volume (r2 = 0.39, p = 0.0000). A robust negative correlation was found between tChPV and RNFL (p < 0.001), GCIPL (p = 0.003), TMV (p = 0.0185), PMB (p < 0.0001), G (p = 0.04), T(p = 0.0001). CONCLUSIONS Our findings support the association of tChPV with disability and altered retinal integrity in RRMS.
Collapse
Affiliation(s)
- Muhammad F Raghib
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Fen Bao
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Mahmoud Elkhooly
- Department of Neurology, Wayne State University School of Medicine, United States of America; Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, United States of America; Department of Neurology and Psychiatry, Minia University, Minia, Egypt
| | - Evanthia Bernitsas
- Department of Neurology, Wayne State University School of Medicine, United States of America; Detroit Medical Center, Detroit, MI, United States of America.
| |
Collapse
|
25
|
Rodriguez-Mogeda C, van Ansenwoude CMJ, van der Molen L, Strijbis EMM, Mebius RE, de Vries HE. The role of CD56 bright NK cells in neurodegenerative disorders. J Neuroinflammation 2024; 21:48. [PMID: 38350967 PMCID: PMC10865604 DOI: 10.1186/s12974-024-03040-8] [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: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
Emerging evidence suggests a potential role for natural killer (NK) cells in neurodegenerative diseases, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. However, the precise function of NK cells in these diseases remains ambiguous. The existence of two NK cell subsets, CD56bright and CD56dim NK cells, complicates the understanding of the contribution of NK cells in neurodegeneration as their functions within the context of neurodegenerative diseases may differ significantly. CD56bright NK cells are potent cytokine secretors and are considered more immunoregulatory and less terminally differentiated than their mostly cytotoxic CD56dim counterparts. Hence, this review focusses on NK cells, specifically on CD56bright NK cells, and their role in neurodegenerative diseases. Moreover, it explores the mechanisms underlying their ability to enter the central nervous system. By consolidating current knowledge, we aim to provide a comprehensive overview on the role of CD56bright NK cells in neurodegenerative diseases. Elucidating their impact on neurodegeneration may have implications for future therapeutic interventions, potentially ameliorating disease pathogenesis.
Collapse
Affiliation(s)
- Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Chaja M J van Ansenwoude
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Lennart van der Molen
- IQ Health Science Department, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eva M M Strijbis
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Department of Neurology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Rebejac J, Eme-Scolan E, Rua R. Role of meningeal immunity in brain function and protection against pathogens. J Inflamm (Lond) 2024; 21:3. [PMID: 38291415 PMCID: PMC10829400 DOI: 10.1186/s12950-023-00374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
The brain and spinal cord collectively referred to as the Central Nervous System (CNS) are protected by the blood-brain barrier that limits molecular, microbial and immunological trafficking. However, in the last decade, many studies have emphasized the protective role of 'border regions' at the surface of the CNS which are highly immunologically active, in contrast with the CNS parenchyma. In the steady-state, lymphoid and myeloid cells residing in the cranial meninges can affect brain function and behavior. Upon infection, they provide a first layer of protection against microbial neuroinvasion. The maturation of border sites over time enables more effective brain protection in adults as compared to neonates. Here, we provide a comprehensive update on the meningeal immune system and its role in physiological brain function and protection against infectious agents.
Collapse
Affiliation(s)
- Julie Rebejac
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Elisa Eme-Scolan
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Rejane Rua
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France.
| |
Collapse
|
27
|
Aspden JW, Murphy MA, Kashlan RD, Xiong Y, Poznansky MC, Sîrbulescu RF. Intruders or protectors - the multifaceted role of B cells in CNS disorders. Front Cell Neurosci 2024; 17:1329823. [PMID: 38269112 PMCID: PMC10806081 DOI: 10.3389/fncel.2023.1329823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
B lymphocytes are immune cells studied predominantly in the context of peripheral humoral immune responses against pathogens. Evidence has been accumulating in recent years on the diversity of immunomodulatory functions that B cells undertake, with particular relevance for pathologies of the central nervous system (CNS). This review summarizes current knowledge on B cell populations, localization, infiltration mechanisms, and function in the CNS and associated tissues. Acute and chronic neurodegenerative pathologies are examined in order to explore the complex, and sometimes conflicting, effects that B cells can have in each context, with implications for disease progression and treatment outcomes. Additional factors such as aging modulate the proportions and function of B cell subpopulations over time and are also discussed in the context of neuroinflammatory response and disease susceptibility. A better understanding of the multifactorial role of B cell populations in the CNS may ultimately lead to innovative therapeutic strategies for a variety of neurological conditions.
Collapse
Affiliation(s)
- James W. Aspden
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Matthew A. Murphy
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rommi D. Kashlan
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yueyue Xiong
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Ruxandra F. Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Traetta ME, Chaves Filho AM, Akinluyi ET, Tremblay MÈ. Neurodevelopmental and Neuropsychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:457-495. [PMID: 39207708 DOI: 10.1007/978-3-031-55529-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This chapter will focus on microglial involvement in neurodevelopmental and neuropsychiatric disorders, particularly autism spectrum disorder (ASD), schizophrenia and major depressive disorder (MDD). We will describe the neuroimmune risk factors that contribute to the etiopathology of these disorders across the lifespan, including both in early life and adulthood. Microglia, being the resident immune cells of the central nervous system, could play a key role in triggering and determining the outcome of these disorders. This chapter will review preclinical and clinical findings where microglial morphology and function were examined in the contexts of ASD, schizophrenia and MDD. Clinical evidence points out to altered microglial morphology and reactivity, as well as increased expression of pro-inflammatory cytokines, supporting the idea that microglial abnormalities are involved in these disorders. Indeed, animal models for these disorders found altered microglial morphology and homeostatic functions which resulted in behaviours related to these disorders. Additionally, as microglia have emerged as promising therapeutic targets, we will also address in this chapter therapies involving microglial mechanisms for the treatment of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada.
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
29
|
Yao S, Gao Z, Fang W, Fu Y, Xue Q, Lai T, Shangguan H, Sun W, Lin Y, Lin F, Kang D. DPA714 PET Imaging Shows That Inflammation of the Choroid Plexus Is Active in Chronic-Phase Intracerebral Hemorrhage. Clin Nucl Med 2024; 49:56-65. [PMID: 38054504 DOI: 10.1097/rlu.0000000000004948] [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: 12/07/2023]
Abstract
PURPOSE Our aims were to investigate the presence of choroid plexus (CP) inflammation in chronic-phase intracerebral hemorrhage (ICH) patients and to characterize any inflammatory cells in the CP. PATIENTS AND METHODS An in vivo 18 F-DPA714 PET study was undertaken in 22 chronic-phase ICH patients who were admitted to the First Affiliated Hospital of Fujian Medical University or Tianjin Medical University General Hospital from April 2017 to June 2020. Ten control participants with nonhemorrhagic central nervous system diseases were included. Choroid plexus 18 F-DPA714 uptake was calculated as the average SUVR. To aid the interpretation of the 18 F-DPA714 uptake results at the CP level, Cy5-DPA714 in vivo imaging and immunofluorescence staining were used to show the presence of CP inflammation in an ICH mouse model during the chronic phase (14 weeks after ICH). Then immunofluorescence staining against translocator protein and other specific biomarkers was used to characterize the cells present in the inflamed CP of ICH mice in the chronic phase. RESULTS PET imaging showed that CP DPA714 SUVRs in chronic-phase ICH patients were higher than in controls (mean CP SUVR ± SD; ICH group: 1.05 ± 0.35; control group: 0.81 ± 0.21; P = 0.006). Immunofluorescence staining of the CP in ICH model mice identified a population of CD45 + immune cells, peripheral monocyte-derived CD14 + cells, CD68 + phagocytes, and CD11b + resident microglia/macrophages expressing translocator protein, possibly contributing to the increased 18 F-DPA714 uptake. CONCLUSIONS Our study shows that CP DPA714 uptake in chronic-phase ICH patients was higher than that of participants with nonhemorrhagic central nervous system diseases, which means that CP inflammation is still active in chronic-phase ICH patients.
Collapse
Affiliation(s)
- Shaobo Yao
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Zhuyu Gao
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | | - Ying Fu
- Department of Neurology, Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian
| | - Qianqian Xue
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Tianmin Lai
- Department of Neurology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Huangcheng Shangguan
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | - Weiwei Sun
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | | - Fuxin Lin
- From the Departments of Department of Neurosurgery, Department of Nuclear Medicine, Neurosurgery Research Institute
| | | |
Collapse
|
30
|
Butic AB, Spencer SA, Shaheen SK, Lukacher AE. Polyomavirus Wakes Up and Chooses Neurovirulence. Viruses 2023; 15:2112. [PMID: 37896889 PMCID: PMC10612099 DOI: 10.3390/v15102112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
JC polyomavirus (JCPyV) is a human-specific polyomavirus that establishes a silent lifelong infection in multiple peripheral organs, predominantly those of the urinary tract, of immunocompetent individuals. In immunocompromised settings, however, JCPyV can infiltrate the central nervous system (CNS), where it causes several encephalopathies of high morbidity and mortality. JCPyV-induced progressive multifocal leukoencephalopathy (PML), a devastating demyelinating brain disease, was an AIDS-defining illness before antiretroviral therapy that has "reemerged" as a complication of immunomodulating and chemotherapeutic agents. No effective anti-polyomavirus therapeutics are currently available. How depressed immune status sets the stage for JCPyV resurgence in the urinary tract, how the virus evades pre-existing antiviral antibodies to become viremic, and where/how it enters the CNS are incompletely understood. Addressing these questions requires a tractable animal model of JCPyV CNS infection. Although no animal model can replicate all aspects of any human disease, mouse polyomavirus (MuPyV) in mice and JCPyV in humans share key features of peripheral and CNS infection and antiviral immunity. In this review, we discuss the evidence suggesting how JCPyV migrates from the periphery to the CNS, innate and adaptive immune responses to polyomavirus infection, and how the MuPyV-mouse model provides insights into the pathogenesis of JCPyV CNS disease.
Collapse
Affiliation(s)
| | | | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (A.B.B.); (S.A.S.); (S.K.S.)
| |
Collapse
|
31
|
Conde JN, Himmler GE, Mladinich MC, Setoh YX, Amarilla AA, Schutt WR, Saladino N, Gorbunova EE, Salamango DJ, Benach J, Kim HK, Mackow ER. Establishment of a CPER reverse genetics system for Powassan virus defines attenuating NS1 glycosylation sites and an infectious NS1-GFP11 reporter virus. mBio 2023; 14:e0138823. [PMID: 37489888 PMCID: PMC10470542 DOI: 10.1128/mbio.01388-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 07/26/2023] Open
Abstract
Powassan virus (POWV) is an emerging tick-borne Flavivirus that causes lethal encephalitis and long-term neurologic damage. Currently, there are no POWV therapeutics, licensed vaccines, or reverse genetics systems for producing infectious POWVs from recombinant DNA. Using a circular polymerase extension reaction (CPER), we generated recombinant LI9 (recLI9) POWVs with attenuating NS1 protein mutations and a recLI9-split-eGFP reporter virus. NS1 proteins are highly conserved glycoproteins that regulate replication, spread, and neurovirulence. POWV NS1 contains three putative N-linked glycosylation sites that we modified individually in infectious recLI9 mutants (N85Q, N208Q, and N224Q). NS1 glycosylation site mutations reduced replication kinetics and were attenuated, with 1-2 log decreases in titer. Severely attenuated recLI9-N224Q exhibited a 2- to 3-day delay in focal cell-to-cell spread and reduced NS1 secretion but was lethal when intracranially inoculated into suckling mice. However, footpad inoculation of recLI9-N224Q resulted in the survival of 80% of mice and demonstrated that NS1-N224Q mutations reduce POWV neuroinvasion in vivo. To monitor NS1 trafficking, we CPER fused a split GFP11-tag to the NS1 C-terminus and generated an infectious reporter virus, recLI9-NS1-GFP11. Cells infected with recLI9-NS1-GFP11 revealed NS1 trafficking in live cells and the novel formation of large NS1-lined intracellular vesicles. An infectious recLI9-NS1-GFP11 reporter virus permits real-time analysis of NS1 functions in POWV replication, assembly, and secretion and provides a platform for evaluating antiviral compounds. Collectively, our robust POWV reverse genetics system permits analysis of viral spread and neurovirulence determinants in vitro and in vivo and enables the rational genetic design of live attenuated POWV vaccines. IMPORTANCE Our findings newly establish a mechanism for genetically modifying Powassan viruses (POWVs), systematically defining pathogenic determinants and rationally designing live attenuated POWV vaccines. This initial study demonstrates that mutating POWV NS1 glycosylation sites attenuates POWV spread and neurovirulence in vitro and in vivo. Our findings validate a robust circular polymerase extension reaction approach as a mechanism for developing, and evaluating, attenuated genetically modified POWVs. We further designed an infectious GFP-tagged reporter POWV that permits us to monitor secretory trafficking of POWV in live cells, which can be applied to screen potential POWV replication inhibitors. This robust system for modifying POWVs provides the ability to define attenuating POWV mutations and create genetically attenuated recPOWV vaccines.
Collapse
Affiliation(s)
- Jonas N. Conde
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Queensland, Australia
| | - Grace E. Himmler
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Queensland, Australia
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
| | - Megan C. Mladinich
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Queensland, Australia
| | - Yin Xiang Setoh
- Microbiology and Molecular Epidemiology Division, Environmental Health Institute, National Environmental Agency, Singapore, Singapore
| | - Alberto A. Amarilla
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Queensland, Australia
| | - William R. Schutt
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| | - Nicholas Saladino
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Elena E. Gorbunova
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| | - Daniel J. Salamango
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Jorge Benach
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| | - Hwan Keun Kim
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| | - Erich R. Mackow
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
32
|
Jankowska A, Chwojnicki K, Grzywińska M, Trzonkowski P, Szurowska E. Choroid Plexus Volume Change-A Candidate for a New Radiological Marker of MS Progression. Diagnostics (Basel) 2023; 13:2668. [PMID: 37627928 PMCID: PMC10453931 DOI: 10.3390/diagnostics13162668] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Multiple sclerosis (MS) is an auto-immune, chronic, neuroinflammatory, demyelinating disease that affects mainly young patients. This progressive inflammatory process causes the chronic loss of brain tissue and results in a deterioration in quality of life. To monitor neuroinflammatory process activity and predict the further development of disease, it is necessary to find a suitable biomarker that could easily be used. In this research, we verify the usability of choroid plexus (CP) volume, a new MS biomarker, in the monitoring of the progression of multiple sclerosis disease. (2) Methods: A single-center, prospective study with three groups of patients was conducted based on the following groups: MS patients who received experimental cellular therapy (Treg), treatment-naïve MS patients and healthy controls. (3) Results: This study concludes that there is a correlation between the CPV/TIV (choroid plexus/total intracranial volume) ratio and the progress of multiple sclerosis disease-patients with MS (MS + Treg) had larger volumes of choroid plexuses. CPV/TIV ratios in MS groups were constantly and significantly growing. In the Treg group, patients with relapses had larger plexuses in comparison to the group with no relapses of MS. A similar correlation was observed for the GD+ group (patients with postcontrast enhancing plaques) compared against the non-GD group (patients without postcontrast enhancing plaques). (4) Conclusion: Choroid plexus volume, due to its immunological function, correlates with the inflammatory process in the central nervous system. We consider it to become a valuable radiological biomarker of MS activity.
Collapse
Affiliation(s)
- Anna Jankowska
- 2nd Department of Radiology, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| | - Kamil Chwojnicki
- Department of Anesthesiology and Intensive Care, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Małgorzata Grzywińska
- Neuroinformatics and Artificial Intelligence Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7, 80-210 Gdańsk, Poland;
| | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| |
Collapse
|
33
|
Silva JEDS, Jussiani GG, Grano FG, Pelissari MCC, de Melo GD, Negrão Watanabe TT, de Lima VF, Machado GF. Increased CCL-5 (RANTES) Gene Expression in the Choroid Plexus of Dogs with Canine Leishmaniosis. Animals (Basel) 2023; 13:2060. [PMID: 37443858 DOI: 10.3390/ani13132060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Visceral canine leishmaniasis (CanL) can cause several clinical manifestations, including neurological lesions. Few reports have characterized the lesions observed in the central nervous system (CNS) during CanL; however, its pathogenesis remains unclear. The choroid plexus (CP) is a specialized structure responsible for the production and secretion of cerebrospinal fluid (CSF) and considered an interface between the peripheral immune system and CNS. It can allow the passage of inflammatory cells or pathogens and has the potential to act as a source of inflammatory mediators in several diseases. Thus, this study aimed to evaluate the role of CP as a possible route of inflammatory cells in the development of brain lesions in dogs with CanL, as well as its association with blood-CSF barrier (BCSFB) dysfunction. Samples were collected from 19 dogs that were naturally infected with CanL. We evaluated the histopathological lesions in the brain and investigated the gene expression of the cytokines. Capture enzyme-linked immunosorbent assay (ELISA) was used to evaluate the presence of the same cytokines in the CSF. Biochemical analysis was performed to compare the presence of albumin in the serum and CSF. Indirect ELISA was performed to measure the presence of anti-Leishmania antibodies in the CSF, which would suggest the disruption of the BCSFB. Histopathological evaluation of the dogs' brains revealed mild-to-severe inflammatory infiltrates, mainly in the CP and meninges. We also detected the presence of anti-Leishmania antibodies and albumin in the CSF, as well as Leishmania DNA in the CP. The gene expression of CCL-5 was increased in the CP of infected dogs compared with that of controls, and there was a tendency for the increase in the gene expression of CXCL-10. Thus, our findings confirm the disfunction of the BCSFB during CanL and suggest that the chemokines CCL-5 and CXCL-10 can be responsible for the recruitment of inflammatory cells found in CP.
Collapse
Affiliation(s)
- José Eduardo Dos Santos Silva
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| | - Giulia Gonçalves Jussiani
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| | - Fernanda Grecco Grano
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| | - Maria Cecília Clarindo Pelissari
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| | - Guilherme Dias de Melo
- Lyssavirus Epidemiology and Neuropathology Unit, Intitut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Tatiane Terumi Negrão Watanabe
- Department Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Antech Diagnostics, 12401 West Olympic Blvd, Los Angeles, CA 90064, USA
| | - Valéria Felix de Lima
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| | - Gisele Fabrino Machado
- School of Veterinary Medicine, Faculdade de Medicina Veterinária de Araçatuba (FMVA), São Paulo State University (UNESP), R. Clóvis Pestana, 793, Dona Amélia, Araçatuba 16050-680, SP, Brazil
| |
Collapse
|
34
|
Ding Z, Fan X, Zhang Y, Yao M, Wang G, Dong Y, Liu J, Song W. The glymphatic system: a new perspective on brain diseases. Front Aging Neurosci 2023; 15:1179988. [PMID: 37396658 PMCID: PMC10308198 DOI: 10.3389/fnagi.2023.1179988] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/29/2023] [Indexed: 07/04/2023] Open
Abstract
The glymphatic system is a brain-wide perivascular pathway driven by aquaporin-4 on the endfeet of astrocytes, which can deliver nutrients and active substances to the brain parenchyma through periarterial cerebrospinal fluid (CSF) influx pathway and remove metabolic wastes through perivenous clearance routes. This paper summarizes the composition, overall fluid flow, solute transport, related diseases, affecting factors, and preclinical research methods of the glymphatic system. In doing so, we aim to provide direction and reference for more relevant researchers in the future.
Collapse
|
35
|
Verrienti G, Megliola G, Colamaria A, Condò T, Lozupone E. Hydrocephalus as Possible Neurological Complication of COVID-19: A Case Report and Systematic Literature Review. Cureus 2023; 15:e41199. [PMID: 37525817 PMCID: PMC10387289 DOI: 10.7759/cureus.41199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/02/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), typically affects the respiratory system but can also present with neurological manifestations. Although some cases of hydrocephalus related to COVID-19 infection have been reported, a clear association between these two entities is not universally recognized yet. Here, we report another interesting case of hydrocephalus in a 60-year-old man with a previous aneurysmal subarachnoid haemorrhage (aSAH) who tested positive for COVID-19. Secondly, we illustrate a systematic overview of the previously reported cases of hydrocephalus related to COVID-19 infection. Finally, in light of the literature, we discuss the supposed underlying mechanisms that could make the association between COVID-19 infection and hydrocephalus plausible.
Collapse
Affiliation(s)
- Giulio Verrienti
- Department of Neurorehabilitation, Casa di Cura Villa Verde, Lecce, ITA
| | | | | | - Tommaso Condò
- Department of Neuroradiology, Vito Fazzi Hospital, Lecce, ITA
| | - Emilio Lozupone
- Department of Neuroradiology, Vito Fazzi Hospital, Lecce, ITA
| |
Collapse
|
36
|
Associations between brain gene expression perturbations implicated by COVID-19 and psychiatric disorders. J Psychiatr Res 2023; 162:79-87. [PMID: 37105022 PMCID: PMC10043811 DOI: 10.1016/j.jpsychires.2023.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Background Currently, there is increasing evidence from clinic, epidemiology, as well as neuroimaging, demonstrating neuropsychiatric abnormalities in COVID-19, however, whether there were associations between brain changes caused by COVID-19 and genetic susceptibility of psychiatric disorders was still unknown. Methods In this study, we performed a meta-analysis to investigate these associations by combing single-cell RNA sequencing datasets of brain tissues of COVID-19 and genome-wide association study summary statistics of psychiatric disorders. Results The analysis demonstrated that among ten psychiatric disorders, gene expression perturbations implicated by COVID-19 in excitatory neurons of choroid plexus were significantly associated with schizophrenia. Conclusions Our analysis might provide insights for the underlying mechanism of the psychiatric consequence of COVID-19.
Collapse
|
37
|
Sher AA, Lao YT, Coombs KM. HLA-A, HSPA5, IGFBP5 and PSMA2 Are Restriction Factors for Zika Virus Growth in Astrocytic Cells. Viruses 2022; 15:97. [PMID: 36680137 PMCID: PMC9863221 DOI: 10.3390/v15010097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Zika virus (ZIKV), an arbo-flavivirus, is transmitted via Aeges aegyptii mosquitoes Following its major outbreaks in 2013, 2014 and 2016, WHO declared it a Public Health Emergency of International Concern. Symptoms of ZIKV infection include acute fever, conjunctivitis, headache, muscle & joint pain and malaise. Cases of its transmission also have been reported via perinatal, sexual and transfusion transmission. ZIKV pathologies include meningo-encephalitis and myelitis in the central nervous system (CNS) and Guillain-Barré syndrome and acute transient polyneuritis in the peripheral nervous system (PNS). Drugs like azithromycin have been tested as inhibitors of ZIKV infection but no vaccines or treatments are currently available. Astrocytes are the most abundant cells in the CNS and among the first cells in CNS infected by ZIKV; (2) Methods: We previously used SOMAScan proteomics to study ZIKV-infected astrocytic cells. Here, we use mass spectrometric analyses to further explain dysregulations in the cellular expression profile of glioblastoma astrocytoma U251 cells. We also knocked down (KD) some of the U251 cellular proteins using siRNAs and observed the impact on ZIKV replication and infectivity; (3) Results & Conclusions: The top ZIKV dysregulated cellular networks were antimicrobial response, cell death, and energy production while top dysregulated functions were antigen presentation, viral replication and cytopathic impact. Th1 and interferon signaling pathways were among the top dysregulated canonical pathways. siRNA-mediated KD of HLA-A, IGFBP5, PSMA2 and HSPA5 increased ZIKV titers and protein synthesis, indicating they are ZIKV restriction factors. ZIKV infection also restored HLA-A expression in HLA-A KD cells by 48 h post-infection, suggesting interactions between this gene product and ZIKV.
Collapse
Affiliation(s)
- Affan A. Sher
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ying Tenny Lao
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|