Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.007
Filter
2.
Front Immunol ; 15: 1400641, 2024.
Article in English | MEDLINE | ID: mdl-38933267

ABSTRACT

Background and objectives: B cell depleting anti-CD20 monoclonal antibodies (aCD20 mAbs) are highly effective in treatment of multiple sclerosis (MS) but fail to halt the formation of meningeal ectopic lymphoid tissue (mELT) in the murine model experimental autoimmune encephalomyelitis (EAE). While mELT can be examined in EAE, it is not accessible in vivo in MS patients. Our key objectives were to compare the immune cells in cerebrospinal fluid (CSF), which is accessible in patients, with those in mELT, and to study the effects of aCD20 mAbs on CSF and mELT in EAE. Methods: Applying single cell RNA sequencing, we compared gene expression profiles in immune cells from (1) CSF with mELT and (2) aCD20 mAbs treated with control treated mice in a spontaneous 2D2xTh EAE model. Results: The immune cell composition in CSF and mELT was very similar. Gene expression profiles and pathway enrichment analysis revealed no striking differences between the two compartments. aCD20 mAbs led not only to a virtually complete depletion of B cells in the CSF but also to a reduction of naïve CD4+ T cells and marked increase of macrophages. No remarkable differences in regulated genes or pathways were observed. Discussion: Our results suggest that immune cells in the CSF may serve as a surrogate for mELT in EAE. Future studies are required to confirm this in MS patients. The observed increase of macrophages in B cell depleted CSF is a novel finding and requires verification in CSF of aCD20 mAbs treated MS patients. Due to unresolved technical challenges, we were unable to study the effects of aCD20 mAbs on mELT. This should be addressed in future studies.


Subject(s)
B-Lymphocytes , Encephalomyelitis, Autoimmune, Experimental , Meninges , Single-Cell Analysis , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/cerebrospinal fluid , Mice , Meninges/immunology , Meninges/pathology , B-Lymphocytes/immunology , Female , Tertiary Lymphoid Structures/immunology , Mice, Inbred C57BL , Antibodies, Monoclonal/immunology , Transcriptome , Gene Expression Profiling , Antigens, CD20/immunology , Cerebrospinal Fluid/immunology , Disease Models, Animal , Multiple Sclerosis/immunology , Multiple Sclerosis/cerebrospinal fluid
3.
Science ; 384(6702): 1302-1303, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38900900

ABSTRACT

Breast cancer cells migrate from the bone marrow to the leptomeninges.


Subject(s)
Breast Neoplasms , Meningeal Neoplasms , Meninges , Animals , Female , Humans , Mice , Breast Neoplasms/pathology , Cell Movement , Meninges/pathology , Meningeal Neoplasms/secondary
4.
Science ; 384(6702): eadh5548, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38900896

ABSTRACT

The molecular mechanisms that regulate breast cancer cell (BCC) metastasis and proliferation within the leptomeninges (LM) are poorly understood, which limits the development of effective therapies. In this work, we show that BCCs in mice can invade the LM by abluminal migration along blood vessels that connect vertebral or calvarial bone marrow and meninges, bypassing the blood-brain barrier. This process is dependent on BCC engagement with vascular basement membrane laminin through expression of the neuronal pathfinding molecule integrin α6. Once in the LM, BCCs colocalize with perivascular meningeal macrophages and induce their expression of the prosurvival neurotrophin glial-derived neurotrophic factor (GDNF). Intrathecal GDNF blockade, macrophage-specific GDNF ablation, or deletion of the GDNF receptor neural cell adhesion molecule (NCAM) from BCCs inhibits breast cancer growth within the LM. These data suggest integrin α6 and the GDNF signaling axis as new therapeutic targets against breast cancer LM metastasis.


Subject(s)
Bone Neoplasms , Breast Neoplasms , Integrin alpha6 , Meningeal Neoplasms , Meninges , Neural Pathways , Animals , Female , Humans , Mice , Basement Membrane/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Integrin alpha6/metabolism , Laminin/metabolism , Macrophages/metabolism , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/secondary , Meninges/pathology , Neoplasm Invasiveness , Neural Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/genetics , Signal Transduction , Neural Pathways/metabolism , Mice, SCID , Mice, Knockout
5.
J Neuroinflammation ; 21(1): 165, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937750

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a significant risk factor for Alzheimer's disease (AD), and accumulating evidence supports a role for adaptive immune B and T cells in both TBI and AD pathogenesis. We previously identified B cell and major histocompatibility complex class II (MHCII)-associated invariant chain peptide (CLIP)-positive B cell expansion after TBI. We also showed that antagonizing CLIP binding to the antigen presenting groove of MHCII after TBI acutely reduced CLIP + splenic B cells and was neuroprotective. The current study investigated the chronic effects of antagonizing CLIP in the 5xFAD Alzheimer's mouse model, with and without TBI. METHODS: 12-week-old male wild type (WT) and 5xFAD mice were administered either CLIP antagonist peptide (CAP) or vehicle, once at 30 min after either sham or a lateral fluid percussion injury (FPI). Analyses included flow cytometric analysis of immune cells in dural meninges and spleen, histopathological analysis of the brain, magnetic resonance diffusion tensor imaging, cerebrovascular analysis, and assessment of motor and neurobehavioral function over the ensuing 6 months. RESULTS: 9-month-old 5xFAD mice had significantly more CLIP + B cells in the meninges compared to age-matched WT mice. A one-time treatment with CAP significantly reduced this population in 5xFAD mice. Importantly, CAP also improved some of the immune, histopathological, and neurobehavioral impairments in 5xFAD mice over the ensuing six months. Although FPI did not further elevate meningeal CLIP + B cells, it did negate the ability of CAP to reduce meningeal CLIP + B cells in the 5xFAD mice. FPI at 3 months of age exacerbated some aspects of AD pathology in 5xFAD mice, including further reducing hippocampal neurogenesis, increasing plaque deposition in CA3, altering microgliosis, and disrupting the cerebrovascular structure. CAP treatment after injury ameliorated some but not all of these FPI effects.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , B-Lymphocytes , Brain Injuries, Traumatic , Histocompatibility Antigens Class II , Mice, Transgenic , Animals , Mice , Male , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/drug therapy , Histocompatibility Antigens Class II/metabolism , B-Lymphocytes/drug effects , Meninges/pathology , Meninges/drug effects , Amyloid beta-Protein Precursor/genetics , Alzheimer Disease/pathology , Alzheimer Disease/drug therapy , Humans , Disease Models, Animal , Presenilin-1/genetics , Mice, Inbred C57BL
6.
Nat Commun ; 15(1): 5404, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926356

ABSTRACT

B cells and T cells collaborate in multiple sclerosis (MS) pathogenesis. IgH[MOG] mice possess a B cell repertoire skewed to recognize myelin oligodendrocyte glycoprotein (MOG). Here, we show that upon immunization with the T cell-obligate autoantigen, MOG[35-55], IgH[MOG] mice develop rapid and exacerbated experimental autoimmune encephalomyelitis (EAE) relative to wildtype (WT) counterparts, characterized by aggregation of T and B cells in the IgH[MOG] meninges and by CD4+ T helper 17 (Th17) cells in the CNS. Production of the Th17 maintenance factor IL-23 is observed from IgH[MOG] CNS-infiltrating and meningeal B cells, and in vivo blockade of IL-23p19 attenuates disease severity in IgH[MOG] mice. In the CNS parenchyma and dura mater of IgH[MOG] mice, we observe an increased frequency of CD4+PD-1+CXCR5- T cells that share numerous characteristics with the recently described T peripheral helper (Tph) cell subset. Further, CNS-infiltrating B and Tph cells from IgH[MOG] mice show increased reactive oxygen species (ROS) production. Meningeal inflammation, Tph-like cell accumulation in the CNS and B/Tph cell production of ROS were all reduced upon p19 blockade. Altogether, MOG-specific B cells promote autoimmune inflammation of the CNS parenchyma and meninges in an IL-23-dependent manner.


Subject(s)
Autoimmunity , B-Lymphocytes , CD4-Positive T-Lymphocytes , Encephalomyelitis, Autoimmune, Experimental , Interleukin-23 , Myelin-Oligodendrocyte Glycoprotein , Animals , Female , Mice , Autoimmunity/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-23/immunology , Interleukin-23/metabolism , Meninges/immunology , Meninges/pathology , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Myelin Sheath/immunology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , Th17 Cells/immunology
7.
Cell Rep Med ; 5(6): 101606, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38866016

ABSTRACT

Leptomeningeal disease (LMD) remains a rapidly lethal complication for late-stage melanoma patients. Here, we characterize the tumor microenvironment of LMD and patient-matched extra-cranial metastases using spatial transcriptomics in a small number of clinical specimens (nine tissues from two patients) with extensive in vitro and in vivo validation. The spatial landscape of melanoma LMD is characterized by a lack of immune infiltration and instead exhibits a higher level of stromal involvement. The tumor-stroma interactions at the leptomeninges activate tumor-promoting signaling, mediated through upregulation of SERPINA3. The meningeal stroma is required for melanoma cells to survive in the cerebrospinal fluid (CSF) and promotes MAPK inhibitor resistance. Knocking down SERPINA3 or inhibiting the downstream IGR1R/PI3K/AKT axis results in tumor cell death and re-sensitization to MAPK-targeting therapy. Our data provide a spatial atlas of melanoma LMD, identify the tumor-promoting role of meningeal stroma, and demonstrate a mechanism for overcoming microenvironment-mediated drug resistance in LMD.


Subject(s)
Melanoma , Meningeal Neoplasms , Stromal Cells , Tumor Microenvironment , Melanoma/genetics , Melanoma/pathology , Humans , Tumor Microenvironment/genetics , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Animals , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic , Transcriptome/genetics , Gene Expression Profiling , Meninges/pathology , Meninges/metabolism , Drug Resistance, Neoplasm/genetics , Signal Transduction , Female
8.
J Neuroinflammation ; 21(1): 135, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802931

ABSTRACT

Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.


Subject(s)
Brain Injuries, Traumatic , Meninges , Neuroinflammatory Diseases , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Humans , Animals , Meninges/immunology , Meninges/pathology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Neuroimmunomodulation/physiology , Neuroimmunomodulation/immunology
9.
PLoS One ; 19(5): e0300298, 2024.
Article in English | MEDLINE | ID: mdl-38809920

ABSTRACT

BACKGROUND/PURPOSE: Leptomeningeal enhancement (LME) on post-contrast FLAIR is described as a potential biomarker of meningeal inflammation in multiple sclerosis (MS). Here we report an assessment of the impact of MRI field strength and acquisition timing on meningeal contrast enhancement (MCE). METHODS: This was a cross-sectional, observational study of 95 participants with MS and 17 healthy controls (HC) subjects. Each participant underwent an MRI of the brain on both a 7 Tesla (7T) and 3 Tesla (3T) MRI scanner. 7T protocols included a FLAIR image before, soon after (Gd+ Early 7T FLAIR), and 23 minutes after gadolinium (Gd+ Delayed 7T FLAIR). 3T protocol included FLAIR before and 21 minutes after gadolinium (Gd+ Delayed 3T FLAIR). RESULTS: LME was seen in 23.3% of participants with MS on Gd+ Delayed 3T FLAIR, 47.4% on Gd+ Early 7T FLAIR (p = 0.002) and 57.9% on Gd+ Delayed 7T FLAIR (p < 0.001 and p = 0.008, respectively). The count and volume of LME, leptomeningeal and paravascular enhancement (LMPE), and paravascular and dural enhancement (PDE) were all highest for Gd+ Delayed 7T FLAIR and lowest for Gd+ Delayed 3T FLAIR. Non-significant trends were seen for higher proportion, counts, and volumes for LME and PDE in MS compared to HCs. The rate of LMPE was different between MS and HCs on Gd+ Delayed 7T FLAIR (98.9% vs 82.4%, p = 0.003). MS participants with LME on Gd+ Delayed 7T FLAIR were older (47.6 (10.6) years) than those without (42.0 (9.7), p = 0.008). CONCLUSION: 7T MRI and a delay after contrast injection increased sensitivity for all forms of MCE. However, the lack of difference between groups for LME and its association with age calls into question its relevance as a biomarker of meningeal inflammation in MS.


Subject(s)
Contrast Media , Gadolinium , Magnetic Resonance Imaging , Meninges , Multiple Sclerosis , Humans , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Female , Magnetic Resonance Imaging/methods , Male , Adult , Meninges/diagnostic imaging , Meninges/pathology , Cross-Sectional Studies , Middle Aged , Gadolinium/administration & dosage , Case-Control Studies , Brain/diagnostic imaging , Brain/pathology , Clinical Relevance
10.
Cell Mol Life Sci ; 81(1): 192, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652179

ABSTRACT

BACKGROUND:  Alzheimer's disease (AD) is pathologically characterized by the abnormal accumulation of Aß and tau proteins. There has long been a keen interest among researchers in understanding how Aß and tau are ultimately cleared in the brain. The discovery of this glymphatic system introduced a novel perspective on protein clearance and it gained recognition as one of the major brain clearance pathways for clearing these pathogenic proteins in AD. This finding has sparked interest in exploring the potential contribution of the glymphatic/meningeal lymphatic system in AD. Furthermore, there is a growing emphasis and discussion regarding the possibility that activating the glymphatic/meningeal lymphatic system could serve as a novel therapeutic strategy against AD. OBJECTIVES:  Given this current research trend, the primary focus of this comprehensive review is to highlight the role of the glymphatic/meningeal lymphatic system in the pathogenesis of AD. The discussion will encompass future research directions and prospects for treatment in relation to the glymphatic/meningeal lymphatic system.


Subject(s)
Alzheimer Disease , Glymphatic System , Lymphatic System , Meninges , Proteostasis , Animals , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Glymphatic System/metabolism , Glymphatic System/pathology , Lymphatic System/metabolism , Lymphatic System/pathology , Meninges/metabolism , Meninges/pathology , tau Proteins/metabolism
11.
Neuroradiology ; 66(8): 1335-1344, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38658472

ABSTRACT

PURPOSE: To avoid contrast administration in spontaneous intracranial hypotension (SIH), some studies suggest accepting diffuse pachymeningeal hyperintensity (DPMH) on non-contrast fluid-attenuated inversion recovery (FLAIR) as an equivalent sign to diffuse pachymeningeal enhancement (DPME) on contrast-enhanced T1WI (T1ce), despite lacking thorough performance metrics. This study aimed to comprehensively explore its feasibility. METHODS: In this single-center retrospective study, between April 2021 and November 2023, brain MRI examinations of 43 patients clinically diagnosed with SIH were assessed using 1.5 and 3.0 Tesla MRI scanners. Two radiologists independently assessed the presence or absence of DPMH on FLAIR and DPME on T1ce, with T1ce serving as a gold-standard for pachymeningeal thickening. The contribution of the subdural fluid collections to DPMH was investigated with quantitative measurements. Using Cohen's kappa statistics, interobserver agreement was assessed. RESULTS: In 39 out of 43 patients (90.7%), pachymeningeal thickening was observed on T1ce. FLAIR sequence produced an accuracy, sensitivity, specificity, positive predictive value, and negative predictive value of 72.1%, 71.8%, 75.0%, 96.6%, and 21.4% respectively, for determining pachymeningeal thickening. FLAIR identified pachymeningeal thickening in 28 cases; however, among these, 21 cases (75%) revealed that the pachymeningeal hyperintense signal was influenced by subdural fluid collections. False-negative rate for FLAIR was 28.2% (11/39). CONCLUSION: The lack of complete correlation between FLAIR and T1ce in identifying pachymeningeal thickening highlights the need for caution in removing contrast agent administration from the MRI protocol of SIH patients, as it reveals a major criterion (i.e., pachymeningeal enhancement) of Bern score.


Subject(s)
Contrast Media , Intracranial Hypotension , Magnetic Resonance Imaging , Meninges , Humans , Female , Male , Intracranial Hypotension/diagnostic imaging , Magnetic Resonance Imaging/methods , Retrospective Studies , Middle Aged , Adult , Meninges/diagnostic imaging , Meninges/pathology , Aged , Sensitivity and Specificity , Feasibility Studies , Image Enhancement/methods
12.
J Neurooncol ; 167(1): 39-47, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38294637

ABSTRACT

BACKGROUND: Leptomeningeal disease (LMD) secondary to high grade glioma (HGG), such as glioblastoma (GBM), are characterized by the spread of tumor cells to the leptomeninges which further complicates treatment approaches. Intrathecal (IT) chemotherapy has surfaced as a potential strategy to bypass the blood-brain barrier and address the challenges posed by disseminated disease. Here, we present a review of the safety and efficacy of IT chemotherapy in the treatment of LMD secondary to HGG. METHODS: A systematic review following PRISMA guidelines was conducted searching PubMed and Embase from January 1995 to September 2022 using specified terms related to IT chemotherapy for LMD. Included articles involved patients diagnosed with LMD from HGG, treated with intrathecal chemotherapy, and provided survival data. Data, including demographics, tumor characteristics, treatment, and survival information, were collected and independently extracted. RESULTS: A total of 68 patients across 10 clinical studies were diagnosed with LMD from HGG and included in the review. Among these patients, the average age at diagnosis was 44.2 years. GBM was the most common tumor type (n = 58, 85.3%). A majority of the patients presented with recurrent disease (n = 29, 60.4%). The review encompassed various IT chemotherapy regimens, including mafosfamide, thio-TEPA, 5-fluoro-2'-deoxyuridine (FdUrd), methotrexate (MTX), and cytarabine; however, dosages and frequencies were inconsistently reported. The mean progression-free survival (PFS) and overall survival (OS) for this cohort were 7.5 months and 11.7 months, respectively. Common side effects of IT chemotherapy included headaches, nausea, and vomiting, with more severe complications such as myelotoxicity, disseminated intravascular coagulopathy, meningitis, and gastrointestinal toxicity reported in some cases. CONCLUSION: LMD continues to be an uncommon complication associated with HGG with a poor prognosis. This article provides an overview of the presently available literature on IT chemotherapy for LMD secondary to HGG, and their respective treatment protocols with overall survival attributes. Additional research is warranted to ascertain how to maximize the potential efficacy of IT chemotherapy as a treatment option.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Adult , Brain Neoplasms/pathology , Glioma/complications , Glioma/drug therapy , Glioblastoma/drug therapy , Thiotepa/therapeutic use , Meninges/pathology
13.
Eur J Radiol ; 171: 111299, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38237519

ABSTRACT

PURPOSE: The purpose of this study is to investigate whether the presence and pattern of enhancement at the internal acoustic canal (IAC) could help in discriminating between leptomeningeal carcinomatosis (LCa) and meningeal inflammation/infection (MMI). METHODS: Magnetic resonance (MR) images of patients with leptomeningeal enhancement were retrospectively evaluated. MR images of the LCa group (n = 33), MMI group (n = 19) and control group (n = 33) were evaluated for the presence, type (moderate/prominent), and localization (unilateral/bilateral) of the IAC enhancement. RESULTS: The presence of IAC enhancement was significantly more common in patients with LCa (p < 0.001). In 73.7 % of patients with MMI, no contrast enhancement was observed in the IAC. In patients with contrast enhancement in the IAC, the risk of LCa in the etiology is 20 times greater than the risk of having MMI. Seventy-five percent of the IAC enhancement seen in LCa patients and 20 % of the IAC enhancements seen in MMI patients was bilateral. This difference was statistically significant (p = 0.029). CONCLUSION: Intense contrast enhancement of the IAC can be a marker for LCa.


Subject(s)
Meningeal Carcinomatosis , Humans , Meningeal Carcinomatosis/diagnostic imaging , Meningeal Carcinomatosis/pathology , Retrospective Studies , Meninges/pathology , Inflammation/diagnostic imaging , Inflammation/pathology , Magnetic Resonance Imaging/methods
14.
Brain ; 147(7): 2566-2578, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38289855

ABSTRACT

Compartmentalized meningeal inflammation is thought to represent one of the key players in the pathogenesis of cortical demyelination in multiple sclerosis. PET targeting the 18 kDa mitochondrial translocator protein (TSPO) is a molecular-specific approach to quantifying immune cell-mediated density in the cortico-meningeal tissue compartment in vivo. This study aimed to characterize cortical and meningeal TSPO expression in a heterogeneous cohort of multiple sclerosis cases using in vivo simultaneous MR-PET with 11C-PBR28, a second-generation TSPO radioligand, and ex vivo immunohistochemistry. Forty-nine multiple sclerosis patients (21 with secondary progressive and 28 with relapsing-remitting multiple sclerosis) with mixed or high affinity binding for 11C-PBR28 underwent 90-min 11C-PBR28 simultaneous MR-PET. Tracer binding was measured using 60-90 min normalized standardized uptake value ratios sampled at mid-cortical depth and ∼3 mm above the pial surface. Data in multiple sclerosis patients were compared to 21 age-matched healthy controls. To characterize the nature of 11C-PBR28 PET uptake, the meningeal and cortical lesion cellular expression of TSPO was further described in post-mortem brain tissue from 20 cases with secondary progressive multiple sclerosis and five age-matched healthy donors. Relative to healthy controls, patients with multiple sclerosis exhibited abnormally increased TSPO signal in the cortex and meningeal tissue, diffusively in progressive disease and more localized in relapsing-remitting multiple sclerosis. In multiple sclerosis, increased meningeal TSPO levels were associated with increased Expanded Disability Status Scale scores (P = 0.007, by linear regression). Immunohistochemistry, validated using in situ sequencing analysis, revealed increased TSPO expression in the meninges and adjacent subpial cortical lesions of post-mortem secondary progressive multiple sclerosis cases relative to control tissue. In these cases, increased TSPO expression was related to meningeal inflammation. Translocator protein immunostaining was detected on meningeal MHC-class II+ macrophages and cortical-activated MHC-class II+ TMEM119+ microglia. In vivo arterial blood data and neuropathology showed that endothelial binding did not significantly account for increased TSPO cortico-meningeal expression in multiple sclerosis. Our findings support the use of TSPO-PET in multiple sclerosis for imaging in vivo inflammation in the cortico-meningeal brain tissue compartment and provide in vivo evidence implicating meningeal inflammation in the pathogenesis of the disease.


Subject(s)
Meninges , Multiple Sclerosis , Positron-Emission Tomography , Receptors, GABA , Humans , Receptors, GABA/metabolism , Receptors, GABA/genetics , Female , Male , Middle Aged , Adult , Positron-Emission Tomography/methods , Meninges/metabolism , Meninges/diagnostic imaging , Meninges/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Aged , Cerebral Cortex/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/pathology , Magnetic Resonance Imaging , Multiple Sclerosis, Chronic Progressive/metabolism , Multiple Sclerosis, Chronic Progressive/diagnostic imaging , Multiple Sclerosis, Chronic Progressive/pathology , Acetamides , Pyridines
15.
Invest Radiol ; 59(3): 243-251, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37493285

ABSTRACT

BACKGROUND: Leptomeningeal contrast enhancement (LME) on T2-weighted Fluid-Attenuated Inversion Recovery (T2-FLAIR) MRI is a reported marker of leptomeningeal inflammation, which is known to be associated with progression of multiple sclerosis (MS). However, this MRI approach, as typically implemented on clinical 3-tesla (T) systems, detects only a few enhancing foci in ~25% of patients and has thus been criticized as poorly sensitive. PURPOSE: To compare an optimized 3D real-reconstruction inversion recovery (Real-IR) MRI sequence on a clinical 3 T scanner to T2-FLAIR for prevalence, characteristics, and clinical/radiological correlations of LME. MATERIALS AND METHODS: We obtained 3D T2-FLAIR and Real-IR scans before and after administration of standard-dose gadobutrol in 177 scans of 154 participants (98 women, 64%; mean ± SD age: 49 ± 12 years), including 124 with an MS-spectrum diagnosis, 21 with other neurological and/or inflammatory disorders, and 9 without neurological history. We calculated contrast-to-noise ratios (CNR) in 20 representative LME foci and determined association of LME with cortical lesions identified at 7 T (n = 19), paramagnetic rim lesions (PRL) at 3 T (n = 105), and clinical/demographic data. RESULTS: We observed focal LME in 73% of participants on Real-IR (70% in established MS, 33% in healthy volunteers, P < 0.0001), compared to 33% on T2-FLAIR (34% vs. 11%, P = 0.0002). Real-IR showed 3.7-fold more LME foci than T2-FLAIR ( P = 0.001), including all T2-FLAIR foci. LME CNR was 2.5-fold higher by Real-IR ( P < 0.0001). The major determinant of LME status was age. Although LME was not associated with cortical lesions, the number of PRL was associated with the number of LME foci on both T2-FLAIR ( P = 0.003) and Real-IR ( P = 0.0003) after adjusting for age, sex, and white matter lesion volume. CONCLUSIONS: Real-IR a promising tool to detect, characterize, and understand the significance of LME in MS. The association between PRL and LME highlights a possible role of the leptomeninges in sustaining chronic inflammation.


Subject(s)
Multiple Sclerosis , Humans , Female , Adult , Middle Aged , Multiple Sclerosis/pathology , Magnetic Resonance Imaging , Meninges/diagnostic imaging , Meninges/pathology , Inflammation/pathology
16.
Semin Neurol ; 43(6): 859-866, 2023 12.
Article in English | MEDLINE | ID: mdl-37989214

ABSTRACT

Leptomeningeal metastases/diseases (LMDs) are a late-stage complication of solid tumor or hematologic malignancies. LMD is spread of cancer cells to the layers of the leptomeninges (pia and arachnoid maters) and subarachnoid space seen in 3 to 5% of cancer patients. It is a disseminated disease which carries with it significant neurologic morbidity and mortality. Our understanding of disease pathophysiology is currently lacking; however, advances are being made. As our knowledge of disease pathogenesis has improved, treatment strategies have evolved. Mainstays of treatment such as radiotherapy have changed from involved-field radiotherapy strategies to proton craniospinal irradiation which has demonstrated promising results in recent clinical trials. Systemic treatment strategies have also improved from more traditional chemotherapeutics with limited central nervous system (CNS) penetration to more targeted therapies with better CNS tumor response. Many challenges remain from earlier clinical detection of disease through improvement of active treatment options, but we are getting closer to meaningful treatment.


Subject(s)
Meningeal Carcinomatosis , Neoplasms , Humans , Meningeal Carcinomatosis/therapy , Meningeal Carcinomatosis/pathology , Meninges/pathology , Biomarkers
17.
Radiographics ; 43(9): e230039, 2023 08.
Article in English | MEDLINE | ID: mdl-37535461

ABSTRACT

Meningeal lesions can be caused by various conditions and pose diagnostic challenges. The authors review the anatomy of the meninges in the brain and spinal cord to provide a better understanding of the localization and extension of these diseases and summarize the clinical and imaging features of various conditions that cause dural and/or leptomeningeal enhancing lesions. These conditions include infectious meningitis (bacterial, tuberculous, viral, and fungal), autoimmune diseases (vasculitis, connective tissue diseases, autoimmune meningoencephalitis, Vogt-Koyanagi-Harada disease, neuro-Behçet syndrome, Susac syndrome, and sarcoidosis), primary and secondary tumors (meningioma, diffuse leptomeningeal glioneuronal tumor, melanocytic tumors, and lymphoma), tumorlike diseases (histiocytosis and immunoglobulin G4-related diseases), medication-induced diseases (immune-related adverse effects and posterior reversible encephalopathy syndrome), and other conditions (spontaneous intracranial hypotension, amyloidosis, and moyamoya disease). Although meningeal lesions may manifest with nonspecific imaging findings, correct diagnosis is important because the treatment strategy varies among these diseases. ©RSNA, 2023 Online supplemental material and the slide presentation from the RSNA Annual Meeting are available for this article. Quiz questions for this article are available through the Online Learning Center.


Subject(s)
Meningeal Neoplasms , Meningitis , Posterior Leukoencephalopathy Syndrome , Sarcoidosis , Humans , Posterior Leukoencephalopathy Syndrome/complications , Posterior Leukoencephalopathy Syndrome/pathology , Meninges/pathology , Meningitis/diagnosis , Meningitis/etiology , Meningitis/therapy , Neuroimaging , Sarcoidosis/pathology , Meningeal Neoplasms/pathology , Magnetic Resonance Imaging/methods
18.
Mol Neurodegener ; 18(1): 55, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37580702

ABSTRACT

Alzheimer's disease (AD) is an aging-related form of dementia associated with the accumulation of pathological aggregates of amyloid beta and neurofibrillary tangles in the brain. These phenomena are accompanied by exacerbated inflammation and marked neuronal loss, which altogether contribute to accelerated cognitive decline. The multifactorial nature of AD, allied to our still limited knowledge of its etiology and pathophysiology, have lessened our capacity to develop effective treatments for AD patients. Over the last few decades, genome wide association studies and biomarker development, alongside mechanistic experiments involving animal models, have identified different immune components that play key roles in the modulation of brain pathology in AD, affecting its progression and severity. As we will relay in this review, much of the recent efforts have been directed to better understanding the role of brain innate immunity, and particularly of microglia. However, and despite the lack of diversity within brain resident immune cells, the brain border tissues, especially the meninges, harbour a considerable number of different types and subtypes of adaptive and innate immune cells. Alongside microglia, which have taken the centre stage as important players in AD research, there is new and exciting evidence pointing to adaptive immune cells, namely T and B cells found in the brain and its meninges, as important modulators of neuroinflammation and neuronal (dys)function in AD. Importantly, a genuine and functional lymphatic vascular network is present around the brain in the outermost meningeal layer, the dura. The meningeal lymphatics are directly connected to the peripheral lymphatic system in different mammalian species, including humans, and play a crucial role in preserving a "healthy" immune surveillance of the CNS, by shaping immune responses, not only locally at the meninges, but also at the level of the brain tissue. In this review, we will provide a comprehensive view on our current knowledge about the meningeal lymphatic vasculature, emphasizing its described roles in modulating CNS fluid and macromolecule drainage, meningeal and brain immunity, as well as glial and neuronal function in aging and in AD.


Subject(s)
Alzheimer Disease , Animals , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Genome-Wide Association Study , Meninges/pathology , Meninges/physiology , Lymphatic System/metabolism , Lymphatic System/pathology , Brain/metabolism , Mammals/metabolism
19.
Nat Rev Neurol ; 19(8): 461-476, 2023 08.
Article in English | MEDLINE | ID: mdl-37400550

ABSTRACT

Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.


Subject(s)
Multiple Sclerosis , Humans , Gray Matter/metabolism , Gray Matter/pathology , Cerebral Cortex/pathology , Meninges/metabolism , Meninges/pathology , Inflammation , Disease Progression
20.
J Vet Diagn Invest ; 35(5): 573-576, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37382287

ABSTRACT

Neurologic disease associated with migration of plant material is reported infrequently in dogs. Here we describe meningoencephalomyelitis associated with foreign plant material in a 2-y-old castrated male West Highland White Terrier dog with acute neck pain. Magnetic resonance imaging revealed spinal meningeal contrast enhancement. Although clinical signs improved after treatment with steroids, the dog was readmitted for further evaluation 3-mo later and was euthanized after generalized epileptic seizures. Autopsy findings consisted of coalescing, pus-filled, neuroparenchymal cavitations surrounded by hemorrhage in the left caudal colliculus and rostral left cerebellar hemisphere. Histologically, lesions consisted of necrosis and suppuration, which surrounded a 1 × 2-mm foreign body morphologically consistent with plant material and clusters of gram-positive bacterial cocci. Affected areas were surrounded by reactive astrocytes, fibrous connective tissue, and mixed inflammatory infiltrates. Areas of hemorrhage and infiltration by neutrophils and foamy macrophages with fibrinoid change of small capillaries were observed in the adjacent neuroparenchyma. The inflammation extended to the perivascular spaces in the leptomeninges (mesencephalon, cerebellum, and brainstem, and spinal cord) and spinal central canal. Anaerobic bacterial culture of frozen samples of cerebellum yielded heavy growth of Bacteroides pyogenes.


Subject(s)
Dog Diseases , Meningoencephalitis , Dogs , Animals , Meningoencephalitis/diagnosis , Meningoencephalitis/veterinary , Dog Diseases/diagnosis , Spinal Cord/pathology , Meninges/pathology , Cerebellum/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...