Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Nat Commun ; 15(1): 3969, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730242

Encephalitis is a rare and potentially fatal manifestation of herpes simplex type 1 infection. Following genome-wide genetic analyses, we identified a previously uncharacterized and very rare heterozygous variant in the E3 ubiquitin ligase WWP2, in a 14-month-old girl with herpes simplex encephalitis. The p.R841H variant (NM_007014.4:c.2522G > A) impaired TLR3 mediated signaling in inducible pluripotent stem cells-derived neural precursor cells and neurons; cells bearing this mutation were also more susceptible to HSV-1 infection compared to control cells. The p.R841H variant increased TRIF ubiquitination in vitro. Antiviral immunity was rescued following the correction of p.R841H by CRISPR-Cas9 technology. Moreover, the introduction of p.R841H in wild type cells reduced such immunity, suggesting that this mutation is linked to the observed phenotypes.


Encephalitis, Herpes Simplex , Herpesvirus 1, Human , Mutation , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Female , Encephalitis, Herpes Simplex/genetics , Infant , Herpesvirus 1, Human/genetics , Induced Pluripotent Stem Cells/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Ubiquitination , Neurons/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/virology , CRISPR-Cas Systems
2.
Brain ; 147(3): 839-848, 2024 03 01.
Article En | MEDLINE | ID: mdl-38123517

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.


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
3.
Microbiol Spectr ; 11(6): e0275123, 2023 Dec 12.
Article En | MEDLINE | ID: mdl-37815349

IMPORTANCE: Progressive multifocal leukoencephalopathy is a crimpling demyelinating disease of the central nervous system caused by JC polyomavirus (JCPyV). Much about JCPyV propagation in the brain remains obscure because of a lack of proper animal models to study the virus in the context of the disease, thus hampering efforts toward the development of new antiviral strategies. Here, having established a robust and representative model of JCPyV infection in human-induced pluripotent stem cell-derived astrocytes, we are able to fully characterize the effect of JCPyV on the biology of the cells and show that the proteomic signature observed for JCPyV-infected astrocytes is extended to extracellular vesicles (EVs). These data suggest that astrocyte-derived EVs found in body fluids might serve as a rich source of information relevant to JCPyV infection in the brain, opening avenues toward better understanding the pathogenesis of the virus and, ultimately, the identification of new antiviral targets.


Extracellular Vesicles , JC Virus , Polyomavirus Infections , Animals , Humans , JC Virus/physiology , Astrocytes , Proteomics , Antiviral Agents
4.
Fluids Barriers CNS ; 20(1): 76, 2023 Oct 24.
Article En | MEDLINE | ID: mdl-37875964

BACKGROUND: As a consequence of SARS-CoV-2 infection various neurocognitive and neuropsychiatric symptoms can appear, which may persist for several months post infection. However, cell type-specific routes of brain infection and underlying mechanisms resulting in neuroglial dysfunction are not well understood. METHODS: Here, we investigated the susceptibility of cells constituting the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) of the choroid plexus (ChP) to SARS-CoV-2 infection using human induced pluripotent stem cell (hiPSC)-derived cellular models and a ChP papilloma-derived epithelial cell line as well as ChP tissue from COVID-19 patients, respectively. RESULTS: We noted a differential infectibility of hiPSC-derived brain microvascular endothelial cells (BMECs) depending on the differentiation method. Extended endothelial culture method (EECM)-BMECs characterized by a complete set of endothelial markers, good barrier properties and a mature immune phenotype were refractory to SARS-CoV-2 infection and did not exhibit an activated phenotype after prolonged SARS-CoV-2 inoculation. In contrast, defined medium method (DMM)-BMECs, characterized by a mixed endothelial and epithelial phenotype and excellent barrier properties were productively infected by SARS-CoV-2 in an ACE2-dependent manner. hiPSC-derived brain pericyte-like cells (BPLCs) lacking ACE2 expression were not susceptible to SARS-CoV-2 infection. Furthermore, the human choroid plexus papilloma-derived epithelial cell line HIBCPP, modeling the BCSFB was productively infected by SARS-CoV-2 preferentially from the basolateral side, facing the blood compartment. Assessment of ChP tissue from COVID-19 patients by RNA in situ hybridization revealed SARS-CoV-2 transcripts in ChP epithelial and ChP stromal cells. CONCLUSIONS: Our study shows that the BCSFB of the ChP rather than the BBB is susceptible to direct SARS-CoV-2 infection. Thus, neuropsychiatric symptoms because of COVID-19 may rather be associated with dysfunction of the BCSFB than the BBB. Future studies should consider a role of the ChP in underlying neuropsychiatric symptoms following SARS-CoV-2 infection.


COVID-19 , Induced Pluripotent Stem Cells , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , SARS-CoV-2/metabolism , Pericytes/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Induced Pluripotent Stem Cells/metabolism , Epithelial Cells/metabolism , Choroid Plexus/metabolism
5.
Cells ; 12(7)2023 03 23.
Article En | MEDLINE | ID: mdl-37048058

Tauopathies are neurodegenerative disorders involving the accumulation of tau isoforms in cell subpopulations such as astrocytes. The origins of the 3R and 4R isoforms of tau that accumulate in astrocytes remain unclear. Extracellular vesicles (EVs) were isolated from primary neurons overexpressing 1N3R or 1N4R tau or from human brain extracts (progressive supranuclear palsy or Pick disease patients or controls) and characterized (electron microscopy, nanoparticle tracking analysis (NTA), proteomics). After the isolated EVs were added to primary astrocytes or human iPSC-derived astrocytes, tau transfer and mitochondrial system function were evaluated (ELISA, immunofluorescence, MitoTracker staining). We demonstrated that neurons in which 3R or 4R tau accumulated had the capacity to transfer tau to astrocytes and that EVs were essential for the propagation of both isoforms of tau. Treatment with tau-containing EVs disrupted the astrocytic mitochondrial system, altering mitochondrial morphology, dynamics, and redox state. Although similar levels of 3R and 4R tau were transferred, 3R tau-containing EVs were significantly more damaging to astrocytes than 4R tau-containing EVs. Moreover, EVs isolated from the brain fluid of patients with different tauopathies affected mitochondrial function in astrocytes derived from human iPSCs. Our data indicate that tau pathology spreads to surrounding astrocytes via EVs-mediated transfer and modifies their function.


Tauopathies , tau Proteins , Humans , tau Proteins/metabolism , Astrocytes/metabolism , Tauopathies/pathology , Brain/metabolism , Protein Isoforms/metabolism
6.
Article En | MEDLINE | ID: mdl-36717268

BACKGROUND AND OBJECTIVE: Depleting CD20+ B cells is the primary mechanism by which ocrelizumab (OCRE) is efficient in persons with multiple sclerosis (pwMS). However, the exact role of OCRE on other immune cell subsets directly or indirectly remains elusive. The purpose of this study is to characterize the dynamics of peripheral immune cells of pwMS on OCRE. METHODS: We collected blood samples from 38 pwMS before OCRE onset (T0) and at 6 and 12 months (T6, T12) after initiation. To cover the immune cell diversity, using mass cytometry time of flight, we designed a 38-parameter panel to analyze B, T, and innate immune cell markers and CNS migratory markers. In parallel, viral-specific CD8+ T-cell responses were assessed by the quantification of interferon-γ secretion using the enzyme-linked immunospot assay on cytomegalovirus, Epstein-Barr virus, and influenza stimulations. RESULTS: Beside B-cell depletion, we observed a loss in memory CD8+CD20+ and central memory CD8+ T cells but not in CD4+CD20+ T cells already at T6 and T12 (p < 0.001). The loss of memory CD8+ T cells correlated with a lower CXCR3 expression (p < 0.001) and CNS-related LFA-1 integrin expression (p < 0.001) as well as a reduced antiviral cellular immune response observed at both time points (p < 0.001). Of note, we did not observe major changes in the phenotype of the other cell types studied. Seven of 38 (18.4%) patients in our cohort presented with infections while on OCRE; 4 of which were switched from dimethyl fumarate. Finally, using a mixed linear model on mass cytometry data, we demonstrated that the immunomodulation induced by previous disease-modifying therapies (DMTs) was prolonged over the period of the study. DISCUSSION: In addition to its well-known role on B cells, our data suggest that OCRE also acts on CD8+ T cells by depleting the memory compartment. These changes in CD8+ T cells may be an asset in the action of OCRE on MS course but might also contribute to explain the increased occurrence of infections in these patients. Finally, although more data are needed to confirm this observation, it suggests that clinicians should pay a special attention to an increased infection risk in pwMS switched from other DMTs to OCRE.


Epstein-Barr Virus Infections , Multiple Sclerosis , Humans , CD8-Positive T-Lymphocytes , Herpesvirus 4, Human , Epstein-Barr Virus Infections/metabolism , Longitudinal Studies , Phenotype
7.
STAR Protoc ; 3(3): 101620, 2022 09 16.
Article En | MEDLINE | ID: mdl-36035798

This protocol describes the generation and characterization of human induced pluripotent stem cells (hiPSCs) from erythroblasts. A key difference with classical protocols is the reprogramming of erythroblasts from a simple blood draw as opposed to fibroblasts/keratinocytes, which requires a biopsy. Moreover, working with erythroblasts ensures that no recombination of the TCR/BCR genes occurs, as opposed to T cells and whole peripheral blood mononuclear cells-based approaches. Last, this approach uses non-integrative episomes ensuring no integration of transgenes into the hiPSCs genome. For complete details on the use and execution of this protocol, please refer to Perriot et al. (2018).


Induced Pluripotent Stem Cells , Cellular Reprogramming , Erythroblasts , Humans , Leukocytes, Mononuclear , Transgenes
8.
Brain ; 145(12): 4334-4348, 2022 12 19.
Article En | MEDLINE | ID: mdl-35085379

Blood-brain barrier (BBB) breakdown and immune cell infiltration into the CNS are early hallmarks of multiple sclerosis (MS). The mechanisms leading to BBB dysfunction are incompletely understood and generally thought to be a consequence of neuroinflammation. Here, we have challenged this view and asked if intrinsic alterations in the BBB of MS patients contribute to MS pathogenesis. To this end, we made use of human induced pluripotent stem cells derived from healthy controls and MS patients and differentiated them into brain microvascular endothelial cell (BMEC)-like cells as in vitro model of the BBB. MS-derived BMEC-like cells showed impaired junctional integrity, barrier properties and efflux pump activity when compared to healthy controls. Also, MS-derived BMEC-like cells displayed an inflammatory phenotype with increased adhesion molecule expression and immune cell interactions. Activation of Wnt/ß-catenin signalling in MS-derived endothelial progenitor cells enhanced barrier characteristics and reduced the inflammatory phenotype. Our study provides evidence for an intrinsic impairment of BBB function in MS patients that can be modelled in vitro. Human iPSC-derived BMEC-like cells are thus suitable to explore the molecular underpinnings of BBB dysfunction in MS and will assist in the identification of potential novel therapeutic targets for BBB stabilization.


Induced Pluripotent Stem Cells , Multiple Sclerosis , Humans , Blood-Brain Barrier/pathology , Multiple Sclerosis/pathology , Cells, Cultured , Induced Pluripotent Stem Cells/metabolism , Brain/physiology
9.
STAR Protoc ; 2(4): 100902, 2021 12 17.
Article En | MEDLINE | ID: mdl-34746863

This protocol describes how to obtain human astrocytes from human-induced pluripotent stem cells (hiPSCs) in chemically defined media, without the use of fetal bovine serum (FBS). FBS eases the differentiation of astrocytes but also deeply alters their phenotype, as compared with their in vivo counterparts. Our protocol generates hiPSC-derived astrocytes displaying a phenotype and functions similar to human primary astrocytes, including adequate response to inflammation, neurotransmitter uptake, and trophic support to neurons. For complete details on the use and execution of this protocol, please refer to Perriot et al. (2018).


Astrocytes/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Calcium/metabolism , Cells, Cultured , Culture Media/metabolism , Glutamic Acid/metabolism , Humans
10.
Article En | MEDLINE | ID: mdl-34135107

OBJECTIVE: Coronavirus disease (COVID-19) has been associated with a large variety of neurologic disorders. However, the mechanisms underlying these neurologic complications remain elusive. In this study, we aimed at determining whether neurologic symptoms were caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) direct infection or by either systemic or local proinflammatory mediators. METHODS: In this cross-sectional study, we checked for SARS-CoV-2 RNA by quantitative reverse transcription PCR, SARS-CoV-2-specific antibodies, and 49 cytokines/chemokines/growth factors (by Luminex) in the CSF +/- sera of a cohort of 22 COVID-19 patients with neurologic presentation and 55 neurologic control patients (inflammatory neurologic disorder [IND], noninflammatory neurologic disorder, and MS). RESULTS: We detected anti-SARS-CoV-2 immunoglobulin G in patients with severe COVID-19 with signs of intrathecal synthesis for some of them. Of the 4 categories of tested patients, the CSF of IND exhibited the highest level of cytokines, chemokines, and growth factors. By contrast, patients with COVID-19 did not present overall upregulation of inflammatory mediators in the CSF. However, patients with severe COVID-19 (intensive care unit patients) exhibited higher concentrations of CCL2, CXCL8, and vascular endothelium growth factor A (VEGF-A) in the CSF than patients with a milder form of COVID-19. In addition, we could show that intrathecal CXCL8 synthesis was linked to an elevated albumin ratio and correlated with the increase of peripheral inflammation (serum hepatocyte growth factor [HGF] and CXCL10). CONCLUSIONS: Our results do not indicate active replication of SARS-CoV-2 in the CSF or signs of massive inflammation in the CSF compartment but highlight a specific impairment of the neurovascular unit linked to intrathecal production of CXCL8.


Brain Diseases/etiology , COVID-19/complications , Cytokines/cerebrospinal fluid , Inflammation/etiology , Neurovascular Coupling , SARS-CoV-2/pathogenicity , Adult , Aged , Aged, 80 and over , Antibodies, Viral/cerebrospinal fluid , Brain Diseases/cerebrospinal fluid , Brain Diseases/immunology , Brain Diseases/physiopathology , COVID-19/cerebrospinal fluid , COVID-19/immunology , Critical Care , Cross-Sectional Studies , Cytokines/blood , Electroencephalography , Female , Humans , Immunoglobulin G/cerebrospinal fluid , Inflammation/cerebrospinal fluid , Inflammation/immunology , Interleukin-8/cerebrospinal fluid , Male , Middle Aged , Neurovascular Coupling/immunology , SARS-CoV-2/immunology , Severity of Illness Index , Young Adult
11.
FASEB J ; 34(12): 16693-16715, 2020 12.
Article En | MEDLINE | ID: mdl-33124083

Human induced pluripotent stem cell (hiPSC)-derived blood-brain barrier (BBB) models established to date lack expression of key adhesion molecules involved in immune cell migration across the BBB in vivo. Here, we introduce the extended endothelial cell culture method (EECM), which differentiates hiPSC-derived endothelial progenitor cells to brain microvascular endothelial cell (BMEC)-like cells with good barrier properties and mature tight junctions. Importantly, EECM-BMEC-like cells exhibited constitutive cell surface expression of ICAM-1, ICAM-2, and E-selectin. Pro-inflammatory cytokine stimulation increased the cell surface expression of ICAM-1 and induced cell surface expression of P-selectin and VCAM-1. Co-culture of EECM-BMEC-like cells with hiPSC-derived smooth muscle-like cells or their conditioned medium further increased the induction of VCAM-1. Functional expression of endothelial ICAM-1 and VCAM-1 was confirmed by T-cell interaction with EECM-BMEC-like cells. Taken together, we introduce the first hiPSC-derived BBB model that displays an adhesion molecule phenotype that is suitable for the study of immune cell interactions.


Blood-Brain Barrier/diagnostic imaging , Cell Communication/physiology , Induced Pluripotent Stem Cells/cytology , Adult , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/metabolism , Cell Adhesion Molecules/metabolism , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , Middle Aged , Vascular Cell Adhesion Molecule-1/metabolism
12.
Stem Cell Reports ; 11(5): 1199-1210, 2018 11 13.
Article En | MEDLINE | ID: mdl-30409508

Recent studies highlighted the importance of astrocytes in neuroinflammatory diseases, interacting closely with other CNS cells but also with the immune system. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still poorly characterized. Here, we develop a serum-free protocol to differentiate human induced pluripotent stem cells (hiPSCs) into astrocytes. Gene expression and functional assays show that our protocol consistently yields a highly enriched population of resting mature astrocytes across the 13 hiPSC lines differentiated. Using this model, we first highlight the importance of serum-free media for astrocyte culture to generate resting astrocytes. Second, we assess the astrocytic response to IL-1ß, TNF-α, and IL-6, all cytokines important in neuroinflammation, such as multiple sclerosis. Our study reveals very specific profiles of reactive astrocytes depending on the triggering stimulus. This model provides ideal conditions for in-depth and unbiased characterization of astrocyte reactivity in neuroinflammatory conditions.


Astrocytes/pathology , Cytokines/pharmacology , Induced Pluripotent Stem Cells/pathology , Multiple Sclerosis/pathology , Astrocytes/drug effects , Astrocytes/metabolism , Case-Control Studies , Cell Differentiation/drug effects , Cells, Cultured , Culture Media, Serum-Free , Humans , Inflammation Mediators/metabolism , Multiple Sclerosis/genetics , Phenotype , Remyelination/drug effects , Transcriptome/drug effects , Transcriptome/genetics
13.
Neurol Neuroimmunol Neuroinflamm ; 4(6): e401, 2017 Nov.
Article En | MEDLINE | ID: mdl-29075657

OBJECTIVE: To evaluate the long-term effects of treatments used in MS on the T-cell trafficking profile. METHODS: We enrolled 83 patients with MS under fingolimod (FTY), natalizumab (NTZ), dimethyl fumarate (DMF), or other disease-modifying treatments (DMTs). Blood was drawn before treatment onset and up to 36-48 months. The ex vivo expression of CNS-related integrins (α4ß1 and αL subunit of LFA-1) and the gut-related integrin (α4ß7) was assessed using flow cytometry on CD4+ and CD8+ T cells. The adhesion profiles of CD3+ T cells to specific integrin ligands (vascular cell adhesion molecule-1 [VCAM-1], intercellular adhesion molecule-1 [ICAM-1], and mucosal vascular addressin cell adhesion molecule-1 [MAdCAM-1]) were measured in vitro before and after 12 and 36-48 months. RESULTS: NTZ decreased the frequency of α4ß1+ and α4ß7+ integrin expressing T cells and the binding of these cells to VCAM-1 and MAdCAM-1, respectively. After 12 months, DMF induced a decreased frequency of αLhighCD4+ T cells combined with reduced binding to ICAM-1. By contrast, with FTY, there was a doubling of the frequency of α4ß1+ and αLhigh, but a decreased frequency of α4ß7+ T cells. Strikingly, the binding of α4ß1+, α4ß7+, and to a lesser extent of αLhigh T cells to VCAM-1, MAdCAM-1, and ICAM-1, respectively, was decreased at month 12 under FTY treatment. The presence of manganese partially restored the binding of these T cells to VCAM-1 in vitro, suggesting that FTY interferes with integrin activation. CONCLUSIONS: In addition to NTZ, DMF and FTY but not other tested DMTs may also decrease T-cell-mediated immune surveillance of the CNS. Whether this mechanism may contribute to the onset of CNS opportunistic infections remains to be shown.

...