Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Immunol ; 25(3): 432-447, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38409259

ABSTRACT

Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.


Subject(s)
Astrocytes , Multiple Sclerosis , Animals , Humans , Mice , Anti-Inflammatory Agents , Disease Models, Animal , Epigenesis, Genetic , Heparin-binding EGF-like Growth Factor/genetics , Inflammation , Proteomics
2.
Nature ; 607(7920): 776-783, 2022 07.
Article in English | MEDLINE | ID: mdl-35859176

ABSTRACT

Mutations of the ADAR1 gene encoding an RNA deaminase cause severe diseases associated with chronic activation of type I interferon (IFN) responses, including Aicardi-Goutières syndrome and bilateral striatal necrosis1-3. The IFN-inducible p150 isoform of ADAR1 contains a Zα domain that recognizes RNA with an alternative left-handed double-helix structure, termed Z-RNA4,5. Hemizygous ADAR1 mutations in the Zα domain cause type I IFN-mediated pathologies in humans2,3 and mice6-8; however, it remains unclear how the interaction of ADAR1 with Z-RNA prevents IFN activation. Here we show that Z-DNA-binding protein 1 (ZBP1), the only other protein in mammals known to harbour Zα domains9, promotes type I IFN activation and fatal pathology in mice with impaired ADAR1 function. ZBP1 deficiency or mutation of its Zα domains reduced the expression of IFN-stimulated genes and largely prevented early postnatal lethality in mice with hemizygous expression of ADAR1 with mutated Zα domain (Adar1mZα/- mice). Adar1mZα/- mice showed upregulation and impaired editing of endogenous retroelement-derived complementary RNA reads, which represent a likely source of Z-RNAs activating ZBP1. Notably, ZBP1 promoted IFN activation and severe pathology in Adar1mZα/- mice in a manner independent of RIPK1, RIPK3, MLKL-mediated necroptosis and caspase-8-dependent apoptosis, suggesting a novel mechanism of action. Thus, ADAR1 prevents endogenous Z-RNA-dependent activation of pathogenic type I IFN responses by ZBP1, suggesting that ZBP1 could contribute to type I interferonopathies caused by ADAR1 mutations.


Subject(s)
Adenosine Deaminase , Interferon Type I , RNA-Binding Proteins , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Apoptosis , Caspase 8/metabolism , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Mice , Mutation , Necroptosis , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Brain ; 147(3): 839-848, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38123517

ABSTRACT

Intrathecal IgM production in multiple sclerosis is associated with a worse disease course. To investigate pathogenic relevance of autoreactive IgM in multiple sclerosis, CSF from two independent cohorts, including multiple sclerosis patients and controls, were screened for antibody binding to induced pluripotent stem cell-derived neurons and astrocytes, and a panel of CNS-related cell lines. IgM binding to a primitive neuro-ectodermal tumour cell line discriminated 10% of multiple sclerosis donors from controls. Transcriptomes of single IgM producing CSF B cells from patients with cell-binding IgM were sequenced and used to produce recombinant monoclonal antibodies for characterization and antigen identification. We produced five cell-binding recombinant IgM antibodies, of which one, cloned from an HLA-DR + plasma-like B cell, mediated antigen-dependent complement activation. Immunoprecipitation and mass spectrometry, and biochemical and transcriptome analysis of the target cells identified the iron transport scavenger protein SCARA5 as the antigen target of this antibody. Intrathecal injection of a SCARA5 antibody led to an increased T cell infiltration in an experimental autoimmune encephalomyelitis (EAE) model. CSF IgM might contribute to CNS inflammation in multiple sclerosis by binding to cell surface antigens like SCARA5 and activating complement, or by facilitating immune cell migration into the brain.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Immunoglobulin M , Multiple Sclerosis , Scavenger Receptors, Class A , Animals , Humans , Antibodies, Monoclonal , Cell Line, Tumor , Immunoglobulin M/cerebrospinal fluid , Membrane Transport Proteins , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Scavenger Receptors, Class A/immunology
4.
Proc Natl Acad Sci U S A ; 119(31): e2205042119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881799

ABSTRACT

Dimethyl fumarate (DMF) is an immunomodulatory treatment for multiple sclerosis (MS). Despite its wide clinical use, the mechanisms underlying clinical response are not understood. This study aimed to reveal immune markers of therapeutic response to DMF treatment in MS. For this purpose, we prospectively collected peripheral blood mononuclear cells (PBMCs) from a highly characterized cohort of 44 individuals with MS before and at 12 and 48 wk of DMF treatment. Single cells were profiled using high-dimensional mass cytometry. To capture the heterogeneity of different immune subsets, we adopted a bioinformatic multipanel approach that allowed cell population-cluster assignment of more than 50 different parameters, including lineage and activation markers as well as chemokine receptors and cytokines. Data were further analyzed in a semiunbiased fashion implementing a supervised representation learning approach to capture subtle longitudinal immune changes characteristic for therapy response. With this approach, we identified a population of memory T helper cells expressing high levels of neuroinflammatory cytokines (granulocyte-macrophage colony-stimulating factor [GM-CSF], interferon γ [IFNγ]) as well as CXCR3, whose abundance correlated with treatment response. Using spectral flow cytometry, we confirmed these findings in a second cohort of patients. Serum neurofilament light-chain levels confirmed the correlation of this immune cell signature with axonal damage. The identified cell population is expanded in peripheral blood under natalizumab treatment, substantiating a specific role in treatment response. We propose that depletion of GM-CSF-, IFNγ-, and CXCR3-expressing T helper cells is the main mechanism of action of DMF and allows monitoring of treatment response.


Subject(s)
Biomarkers, Pharmacological , Cytokines , Dimethyl Fumarate , Immunosuppressive Agents , Multiple Sclerosis , T-Lymphocytes, Helper-Inducer , Biomarkers, Pharmacological/metabolism , Cytokines/metabolism , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interferon-gamma/metabolism , Lymphocyte Depletion , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Single-Cell Analysis , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
5.
Eur J Immunol ; 53(10): e2250234, 2023 10.
Article in English | MEDLINE | ID: mdl-37505465

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Humans , Mice , Central Nervous System , Multiple Sclerosis/pathology , Spinal Cord , Myeloid Cells/metabolism , Mice, Inbred C57BL
6.
J Neurooncol ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900237

ABSTRACT

PURPOSE: Mesenchymal stromal cells (MSCs) within the glioblastoma microenvironment have been shown to promote tumor progression. Tumor Treating Fields (TTFields) are alternating electric fields with low intensity and intermediate frequency that exhibit anti-tumorigenic effects. While the effects of TTFields on glioblastoma cells have been studied previously, nothing is known about the influence of TTFields on MSCs. METHODS: Single-cell RNA sequencing and immunofluorescence staining were employed to identify glioblastoma-associated MSCs in patient samples. Proliferation and clonogenic survival of human bone marrow-derived MSCs were assessed after TTFields in vitro. MSC' characteristic surface marker expression was determined using flow cytometry, while multi-lineage differentiation potential was examined with immunohistochemistry. Apoptosis was quantified based on caspase-3 and annexin-V/7-AAD levels in flow cytometry, and senescence was assessed with ß-galactosidase staining. MSCs' migratory potential was evaluated with Boyden chamber assays. RESULTS: Single-cell RNA sequencing and immunofluorescence showed the presence of glioblastoma-associated MSCs in patient samples. TTFields significantly reduced proliferation and clonogenic survival of human bone marrow-derived MSCs by up to 60% and 90%, respectively. While the characteristic surface marker expression and differentiation capacity were intact after TTFields, treatment resulted in increased apoptosis and senescence. Furthermore, TTFields significantly reduced MSCs' migratory capacity. CONCLUSION: We could demonstrate the presence of tumor-associated MSCs in glioblastoma patients, providing a rationale to study the impact of TTFields on MSCs. TTFields considerably increase apoptosis and senescence in MSCs, resulting in impaired survival and migration. The results provide a basis for further analyses on the role of MSCs in glioblastoma patients receiving TTFields.

7.
Ann Neurol ; 91(5): 676-681, 2022 05.
Article in English | MEDLINE | ID: mdl-35170072

ABSTRACT

Treatment with dimethyl fumarate (DMF) leads to lymphopenia and infectious complications in a subset of patients with multiple sclerosis (MS). Here, we aimed to reveal immune markers of DMF-associated lymphopenia. This prospective observational study longitudinally assessed 31 individuals with MS by single-cell mass cytometry before and after 12 and 48 weeks of DMF therapy. Employing a neural network-based representation learning approach, we identified a CCR4-expressing T helper cell population negatively associated with relevant lymphopenia. CCR4-expressing T helper cells represent a candidate prognostic biomarker for the development of relevant lymphopenia in patients undergoing DMF treatment. ANN NEUROL 2022;91:676-681.


Subject(s)
Lymphopenia , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Dimethyl Fumarate/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Lymphopenia/chemically induced , Multiple Sclerosis/chemically induced , Multiple Sclerosis/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Prospective Studies
8.
Brain ; 144(10): 3126-3141, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34849598

ABSTRACT

Dimethyl fumarate, an approved treatment for relapsing-remitting multiple sclerosis, exerts pleiotropic effects on immune cells as well as CNS resident cells. Here, we show that dimethyl fumarate exerts a profound alteration of the metabolic profile of human CD4+ as well as CD8+ T cells and restricts their antioxidative capacities by decreasing intracellular levels of the reactive oxygen species scavenger glutathione. This causes an increase in mitochondrial reactive oxygen species levels accompanied by an enhanced mitochondrial stress response, ultimately leading to impaired mitochondrial function. Enhanced mitochondrial reactive oxygen species levels not only result in enhanced T-cell apoptosis in vitro as well as in dimethyl fumarate-treated patients, but are key for the well-known immunomodulatory effects of dimethyl fumarate both in vitro and in an animal model of multiple sclerosis, i.e. experimental autoimmune encephalomyelitis. Indeed, dimethyl fumarate immune-modulatory effects on T cells were completely abrogated by pharmacological interference of mitochondrial reactive oxygen species production. These data shed new light on dimethyl fumarate as bona fide immune-metabolic drug that targets the intracellular stress response in activated T cells, thereby restricting mitochondrial function and energetic capacity, providing novel insight into the role of oxidative stress in modulating cellular immune responses and T cell-mediated autoimmunity.


Subject(s)
Antioxidants/pharmacology , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dimethyl Fumarate/pharmacology , Immunosuppressive Agents/pharmacology , Adult , Animals , Antioxidants/therapeutic use , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cohort Studies , Dimethyl Fumarate/therapeutic use , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Young Adult
9.
Mult Scler ; 25(12): 1682-1685, 2019 10.
Article in English | MEDLINE | ID: mdl-31208265

ABSTRACT

BACKGROUND: Progressive multifocal leukoencephalopathy (PML) is the main safety concern for dimethyl fumarate (DMF) treatment in persons with multiple sclerosis (pwMS). Risk stratification under DMF is currently based on age above 50 years and prolonged lymphopenia below 500 cells/µL. OBJECTIVE: To report a case of PML under DMF without severe lymphopenia or immunosenescence. METHODS: Case report. RESULTS: A 39-year-old female pwMS developed DMF-associated oligosymptomatic PML. The patient had not experienced any repeated lymphocyte counts below 800 cells/µL and was 15 years younger than previously described cases. CONCLUSION: Despite risk stratification, vigilance for PML is advised in all pwMS under DMF. Severe CD8-lymphopenia is a common feature of all published DMF-associated cases.


Subject(s)
Dimethyl Fumarate/pharmacology , Leukoencephalopathy, Progressive Multifocal/drug therapy , Lymphopenia/drug therapy , Multiple Sclerosis/drug therapy , Adult , Female , Humans , Immunosuppressive Agents/pharmacology , Lymphocyte Count/methods , Lymphocytes/drug effects , Lymphopenia/diagnosis , Multiple Sclerosis/diagnosis
10.
J Autoimmun ; 86: 39-50, 2018 01.
Article in English | MEDLINE | ID: mdl-28958667

ABSTRACT

INTRODUCTION: The mode of action of dimethyl fumarate (DMF), an immunomodulatory treatment for relapsing-remitting multiple sclerosis (RRMS), has not yet been fully elucidated. While in-vitro experiments and animal studies suggest effects on immune cell survival, proliferation, migration and oxidative stress response, corresponding observations from human studies are lacking. This study aims to characterize ex-vivo and in-vivo effects in a cohort of DMF treated RRMS patients. METHODS: Blood samples were collected from twenty well-characterized RRMS patients at baseline and after 3, 6 and 12 months of DMF treatment and an age- and gender-matched cohort of 20 healthy individuals at 0 and 3 months. Leukocyte subpopulations, immunoglobulin levels and cytokine secretion were measured. T cells were assessed for their levels of reactive oxygen species (ROS), metabolic status and their proliferative capacity. Levels of antioxidants were determined in serum by mass spectrometry. Responses of monocyte activation markers as well as NFkB and MAPK pathways to DMF were analysed. RESULTS: Upon DMF treatment, all lymphocyte subpopulations dropped significantly over the course of 12 months with cytotoxic and effector T cells being affected most significantly. DMF induced cell death and inhibited proliferation of T cells in-vitro. Interestingly, this anti-proliferative effect decreased under treatment. In-vivo DMF treatment led to decreased T cell glycolysis and higher turn-over of antioxidants. In line with these results a significant increase of cytosolic ROS levels after 3 months treatment was detected in T cells. In-vitro DMF treatment reduced NFkB (p65) translocation to the nucleus and MAPK (p38) levels decreased upon stimulation with monomethyl fumarate (MMF) in-vitro and ex-vivo. Consequently, the expression of co-stimulatory molecules like CD40 and CD150 was decreased in antigen presenting cells both in-vitro and ex-vivo. CONCLUSION: This study translates knowledge from in-vitro and animal studies on DMF into the clinical setting. Our data suggest that DMF not only alters lymphocyte composition, but also has profound effects on proliferation and induces oxidative stress in T cells. It also acts on innate immunity by reducing the activation status of antigen presenting cells (APCs) via NFkB and MAPK inactivation.


Subject(s)
Antigen-Presenting Cells/immunology , Dimethyl Fumarate/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Adult , Antigen Presentation , Antigen-Presenting Cells/drug effects , Cell Death , Cell Proliferation , Cells, Cultured , Cohort Studies , Female , Glycolysis , Humans , Immunity, Innate , Male , Middle Aged , NF-kappa B/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Helper-Inducer/drug effects
12.
Neurooncol Adv ; 6(1): vdae093, 2024.
Article in English | MEDLINE | ID: mdl-38946879

ABSTRACT

Background: Primary CNS lymphoma (PCNSL) and glioblastoma (GBM) both represent frequent intracranial malignancies with differing clinical management. However, distinguishing PCNSL from GBM with conventional MRI can be challenging when atypical imaging features are present. We employed advanced dMRI for noninvasive characterization of the microstructure of PCNSL and differentiation from GBM as the most frequent primary brain malignancy. Methods: Multiple dMRI metrics including Diffusion Tensor Imaging, Neurite Orientation Dispersion and Density Imaging, and Diffusion Microstructure Imaging were extracted from the contrast-enhancing tumor component in 10 PCNSL and 10 age-matched GBM on 3T MRI. Imaging findings were correlated with cell density and axonal markers obtained from histopathology. Results: We found significantly increased intra-axonal volume fractions (V-intra and intracellular volume fraction) and microFA in PCNSL compared to GBM (all P < .001). In contrast, mean diffusivity (MD), axial diffusivity (aD), and microADC (all P < .001), and also free water fractions (V-CSF and V-ISO) were significantly lower in PCNSL (all P < .01). Receiver-operating characteristic analysis revealed high predictive values regarding the presence of a PCNSL for MD, aD, microADC, V-intra, ICVF, microFA, V-CSF, and V-ISO (area under the curve [AUC] in all >0.840, highest for MD and ICVF with an AUC of 0.960). Comparative histopathology between PCNSL and GBM revealed a significantly increased cell density in PCNSL and the presence of axonal remnants in a higher proportion of samples. Conclusions: Advanced diffusion imaging enables the characterization of the microstructure of PCNSL and reliably distinguishes PCNSL from GBM. Both imaging and histopathology revealed a relatively increased cell density and a preserved axonal microstructure in PCNSL.

13.
AJNR Am J Neuroradiol ; 44(11): 1262-1269, 2023 11.
Article in English | MEDLINE | ID: mdl-37884304

ABSTRACT

BACKGROUND AND PURPOSE: Glioblastomas and metastases are the most common malignant intra-axial brain tumors in adults and can be difficult to distinguish on conventional MR imaging due to similar imaging features. We used advanced diffusion techniques and structural histopathology to distinguish these tumor entities on the basis of microstructural axonal and fibrillar signatures in the contrast-enhancing tumor component. MATERIALS AND METHODS: Contrast-enhancing tumor components were analyzed in 22 glioblastomas and 21 brain metastases on 3T MR imaging using DTI-fractional anisotropy, neurite orientation dispersion and density imaging-orientation dispersion, and diffusion microstructural imaging-micro-fractional anisotropy. Available histopathologic specimens (10 glioblastomas and 9 metastases) were assessed for the presence of axonal structures and scored using 4-level scales for Bielschowsky staining (0: no axonal structures, 1: minimal axonal fragments preserved, 2: decreased axonal density, 3: no axonal loss) and glial fibrillary acid protein expression (0: no glial fibrillary acid protein positivity, 1: limited expression, 2: equivalent to surrounding parenchyma, 3: increased expression). RESULTS: When we compared glioblastomas and metastases, fractional anisotropy was significantly increased and orientation dispersion was decreased in glioblastomas (each P < .001), with a significant shift toward increased glial fibrillary acid protein and Bielschowsky scores. Positive associations of fractional anisotropy and negative associations of orientation dispersion with glial fibrillary acid protein and Bielschowsky scores were revealed, whereas no association between micro-fractional anisotropy with glial fibrillary acid protein and Bielschowsky scores was detected. Receiver operating characteristic curves revealed high predictive values of both fractional anisotropy (area under the curve = 0.8463) and orientation dispersion (area under the curve = 0.8398) regarding the presence of a glioblastoma. CONCLUSIONS: Diffusion imaging fractional anisotropy and orientation dispersion metrics correlated with histopathologic markers of directionality and may serve as imaging biomarkers in contrast-enhancing tumor components.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Humans , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Diffusion Tensor Imaging/methods , Glial Fibrillary Acidic Protein , Magnetic Resonance Imaging/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology
14.
Cancers (Basel) ; 14(19)2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36230839

ABSTRACT

Glioblastomas are the most common primary brain tumors. Despite extensive clinical and molecular insights into these tumors, the prognosis remains dismal. While targeted immunotherapies have shown remarkable success across different non-brain tumor entities, they failed to show efficacy in glioblastomas. These failures prompted the field to reassess the idiosyncrasies of the glioblastoma microenvironment. Several high-dimensional single-cell RNA sequencing studies generated remarkable findings about glioblastoma-associated immune cells. To build on the collective strength of these studies, we integrated several murine and human datasets that profiled glioblastoma-associated immune cells at different time points. We integrated these datasets and utilized state-of-the-art algorithms to investigate them in a hypothesis-free, purely exploratory approach. We identified a robust accumulation of a natural killer cell subset that was characterized by a downregulation of activation-associated genes with a concomitant upregulation of apoptosis genes. In both species, we found a robust upregulation of the Lymphotoxin-ß gene, a cytokine from the TNF superfamily and a key factor for the development of adaptive immunity. Further validation analyses uncovered a correlation of lymphotoxin signaling with mesenchymal-like glioblastoma regions in situ and in TCGA and CGGA glioblastoma cohorts. In summary, we identify lymphotoxin signaling as a potential therapeutic target in glioblastoma-associated natural killer cells.

15.
Cancers (Basel) ; 15(1)2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36612127

ABSTRACT

Although the free water content within the perilesional T2 hyperintense region should differ between glioblastomas (GBM) and brain metastases based on histological differences, the application of classical MR diffusion models has led to inconsistent results regarding the differentiation between these two entities. Whereas diffusion tensor imaging (DTI) considers the voxel as a single compartment, multicompartment approaches such as neurite orientation dispersion and density imaging (NODDI) or the recently introduced diffusion microstructure imaging (DMI) allow for the calculation of the relative proportions of intra- and extra-axonal and also free water compartments in brain tissue. We investigate the potential of water-sensitive DTI, NODDI and DMI metrics to detect differences in free water content of the perilesional T2 hyperintense area between histopathologically confirmed GBM and brain metastases. Respective diffusion metrics most susceptible to alterations in the free water content (MD, V-ISO, V-CSF) were extracted from T2 hyperintense perilesional areas, normalized and compared in 24 patients with GBM and 25 with brain metastases. DTI MD was significantly increased in metastases (p = 0.006) compared to GBM, which was corroborated by an increased DMI V-CSF (p = 0.001), while the NODDI-derived ISO-VF showed only trend level increase in metastases not reaching significance (p = 0.060). In conclusion, diffusion MRI metrics are able to detect subtle differences in the free water content of perilesional T2 hyperintense areas in GBM and metastases, whereas DMI seems to be superior to DTI and NODDI.

16.
Gut Microbes ; 14(1): 2147055, 2022.
Article in English | MEDLINE | ID: mdl-36398902

ABSTRACT

Mounting evidence points towards a pivotal role of gut microbiota in multiple sclerosis (MS) pathophysiology. Yet, whether disease-modifying treatments alter microbiota composition and whether microbiota shape treatment response and side-effects remain unclear. In this prospective observational pilot study, we assessed the effect of dimethyl fumarate (DMF) on gut microbiota and on host/microbial metabolomics in a cohort of 20 MS patients. Combining state-of-the-art microbial sequencing, metabolome mass spectrometry, and computational analysis, we identified longitudinal changes in gut microbiota composition under DMF-treatment and an increase in citric acid cycle metabolites. Notably, DMF-induced lymphopenia, a clinically relevant safety concern, was correlated with distinct baseline microbiome signatures in MS patients. We identified gastrointestinal microbiota as a key therapeutic target for metabolic properties of DMF. By characterizing gut microbial composition as a candidate risk factor for DMF-induced lymphopenia, we provide novel insights into the role of microbiota in mediating clinical side-effects.


Subject(s)
Gastrointestinal Microbiome , Lymphopenia , Multiple Sclerosis , Humans , Dimethyl Fumarate/adverse effects , Multiple Sclerosis/drug therapy , Prospective Studies , Lymphopenia/chemically induced , Risk Factors
17.
Cancers (Basel) ; 14(5)2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35267463

ABSTRACT

Purpose: Glioblastomas (GBM) and brain metastases are often difficult to differentiate in conventional MRI. Diffusion microstructure imaging (DMI) is a novel MR technique that allows the approximation of the distribution of the intra-axonal compartment, the extra-axonal cellular, and the compartment of interstitial/free water within the white matter. We hypothesize that alterations in the T2 hyperintense areas surrounding contrast-enhancing tumor components may be used to differentiate GBM from metastases. Methods: DMI was performed in 19 patients with glioblastomas and 17 with metastatic lesions. DMI metrics were obtained from the T2 hyperintense areas surrounding contrast-enhancing tumor components. Resected brain tissue was assessed in six patients in each group for features of an edema pattern and tumor infiltration in the perilesional interstitium. Results: Within the perimetastatic T2 hyperintensities, we observed a significant increase in free water (p < 0.001) and a decrease in both the intra-axonal (p = 0.006) and extra-axonal compartments (p = 0.024) compared to GBM. Perilesional free water fraction was discriminative regarding the presence of GBM vs. metastasis with a ROC AUC of 0.824. Histologically, features of perilesional edema were present in all assessed metastases and absent or marginal in GBM. Conclusion: Perilesional T2 hyperintensities in brain metastases and GBM differ significantly in DMI-values. The increased free water fraction in brain metastases suits the histopathologically based hypothesis of perimetastatic vasogenic edema, whereas in glioblastomas there is additional tumor infiltration.

18.
Article in English | MEDLINE | ID: mdl-33589541

ABSTRACT

OBJECTIVE: To identify an MS-specific immune cell population by deep immune phenotyping and relate it to soluble signaling molecules in CSF. METHODS: We analyzed surface expression of 22 markers in paired blood/CSF samples from 39 patients using mass cytometry (cytometry by time of flight). We also measured the concentrations of 296 signaling molecules in CSF using proximity extension assay. Results were analyzed using highly automated unsupervised algorithmic informatics. RESULTS: Mass cytometry objectively identified a B-cell population characterized by the expression of CD49d, CD69, CD27, CXCR3, and human leukocyte antigen (HLA)-DR as clearly associated with MS. Concentrations of the B cell-related factors, notably FCRL2, were increased in MS CSF, especially in early stages of the disease. The B-cell trophic factor B cell activating factor (BAFF) was decreased in MS. Proteins involved in neural plasticity were also reduced in MS. CONCLUSION: When analyzed without a priori assumptions, both the soluble and the cellular compartments of the CSF in MS were characterized by markers related to B cells, and the strongest candidate for an MS-specific cell type has a B-cell phenotype.


Subject(s)
B-Cell Activating Factor/cerebrospinal fluid , B-Lymphocytes/cytology , Biomarkers/cerebrospinal fluid , Multiple Sclerosis/immunology , Adult , B-Lymphocytes/immunology , Biomarkers/analysis , Female , Humans , Male , Middle Aged , Phenotype
19.
Ther Adv Neurol Disord ; 12: 1756286419833574, 2019.
Article in English | MEDLINE | ID: mdl-30873219

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS). Despite improvements of immunomodulatory therapies in relapsing-remitting MS, the pathomechanisms of progressive disease are poorly understood and therapeutically addressed to date. A pathophysiological role for proteins encoded by human endogenous retroviruses (HERVs) has been proposed. GNbAC1 is a monoclonal antibody directed against the envelope protein of a HERV with postulated involvement in MS. METHODS: This review addresses the treatment concept of GNbAC1, the design, preclinical and clinical development of the antibody, as published by November 2018. All four in-human trials (of which two addressed MS) are discussed. CONCLUSION: The treatment concept of GNbAC1 is appealing but remains controversial due to conflicting results regarding the hypothesized underlying pathomechanism. Anticipated immunomodulatory effects were not observed in clinical or pharmacodynamic analyses of the currently available data. However, a magnetic resonance imaging sign compatible with the remyelinating potential of GNbAC1 encouraged further development of this antibody in progressive MS. No relevant issues with tolerability or safety have been described to date.

20.
Mult Scler Relat Disord ; 31: 38-40, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30901703

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) and psoriasis vulgaris (PV) are two disorders with autoimmune pathophysiology and a supposed altered T helper (TH) cell response. OBJECTIVE: To report a case of concomitant relapsing remitting MS and PV treated with different immunomodulatory medications, particularly secukinumab and rituximab. METHODS: Case report. RESULTS: In a 68-year-old woman diagnosed with MS and PV, secukinumab alleviated the dermatological condition, but could not control neuroinflammation. Rituximab treatment halted MS activity, but led to a flare-up of dermatological inflammation. CONCLUSION: The presented case suggests that the pathomechanisms of MS and PV differ regarding involvement of TH and B cells with implications for therapeutic approaches.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunologic Factors/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Psoriasis/drug therapy , Rituximab/therapeutic use , Aged , Female , Humans , Multiple Sclerosis, Relapsing-Remitting/complications , Multiple Sclerosis, Relapsing-Remitting/immunology , Psoriasis/complications , Psoriasis/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL