Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Epilepsia Open ; 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38017663

ABSTRACT

OBJECTIVE: To report the interim results of the PERPRISE study (Study 509; NCT04202159), which is evaluating perampanel as the only adjunctive anti-seizure medication (ASM) in adults with focal to bilateral tonic-clonic seizures (FBTCS) or primary generalized tonic-clonic seizures (GTCS). METHODS: PERPRISE is an ongoing 12-month multicenter, prospective, observational, non-interventional study of perampanel in a real-world setting in Germany. Patients are aged ≥18 years with FBTCS or GTCS due to focal or idiopathic generalized epilepsy. Perampanel, as an adjunctive therapy to ASM monotherapy ('add-on therapy') or as a substitute for one ASM in dual therapy ('substitution therapy'), is prescribed in line with its SmPC. The Interim Analysis Set comprises the first 100 patients who received ≥1 dose of perampanel and attended or discontinued prior to the ~6-month visit. Interim endpoints include retention rate, measures of effects on seizure frequency, and treatment-emergent adverse events (TEAEs). RESULTS: One hundred patients were included in the Interim Analysis Set (add-on, n = 43 [43.0%]; substitution, n = 55 [55.0%]; unknown, n = 2). The 6-month retention rate was 78.0% (add-on, 83.7%; substitution, 72.7%). For the overall population with GTCS and/or FBTCS, seizure-freedom rate at 6 months was 58.8% (add-on, 72.2%; substitution, 47.9%) and 50% responder rate at 6 months was 82.6% (add-on, 89.2%; substitution, 76.6%). Retention rates and seizure outcomes were better with perampanel as an early-line treatment than as a late-line treatment. TEAEs were reported by 48 patients (48.0%), most commonly dizziness (n = 9), fatigue (n = 7), and irritability (n = 7). Sixteen patients (16.0%) withdrew from perampanel treatment due to TEAEs. SIGNIFICANCE: The interim analysis of PERPRISE offers insight into the real-world use of perampanel in Germany, including for the first time, clinical practice data from patients with GTCS and switching ASMs within a dual therapy. Further data from PERPRISE will be of value to inform clinical decision-making in this patient cohort. PLAIN LANGUAGE SUMMARY: Patients with epilepsy often take more than one medication for seizure control. This 12month study looked at patients in Germany receiving perampanel as only add-on medication. The interim analysis shows, that at 6 months, over 70% of the 100 patients continued to use perampanel; 59% experienced no seizures during treatment with perampanel, and in 83%, seizure frequency was reduced by half. Side effects occurred in 48% of patients (most commonly dizziness, fatigue, and irritability) and caused 16% to withdraw from the study. Overall, perampanel was a suitable as only add-on medication for patients with epilepsy.

3.
Hum Mol Genet ; 32(3): 431-449, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35997788

ABSTRACT

Usher syndrome (USH) is the most common form of hereditary deaf-blindness in humans. USH is a complex genetic disorder, assigned to three clinical subtypes differing in onset, course and severity, with USH1 being the most severe. Rodent USH1 models do not reflect the ocular phenotype observed in human patients to date; hence, little is known about the pathophysiology of USH1 in the human eye. One of the USH1 genes, USH1C, exhibits extensive alternative splicing and encodes numerous harmonin protein isoforms that function as scaffolds for organizing the USH interactome. RNA-seq analysis of human retinae uncovered harmonin_a1 as the most abundant transcript of USH1C. Bulk RNA-seq analysis and immunoblotting showed abundant expression of harmonin in Müller glia cells (MGCs) and retinal neurons. Furthermore, harmonin was localized in the terminal endfeet and apical microvilli of MGCs, presynaptic region (pedicle) of cones and outer segments (OS) of rods as well as at adhesive junctions between MGCs and photoreceptor cells (PRCs) in the outer limiting membrane (OLM). Our data provide evidence for the interaction of harmonin with OLM molecules in PRCs and MGCs and rhodopsin in PRCs. Subcellular expression and colocalization of harmonin correlate with the clinical phenotype observed in USH1C patients. We also demonstrate that primary cilia defects in USH1C patient-derived fibroblasts could be reverted by the delivery of harmonin_a1 transcript isoform. Our studies thus provide novel insights into PRC cell biology, USH1C pathophysiology and development of gene therapy treatment(s).


Subject(s)
Usher Syndromes , Humans , Usher Syndromes/genetics , Usher Syndromes/therapy , Usher Syndromes/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Retina/metabolism , Photoreceptor Cells/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
4.
Transl Stroke Res ; 13(3): 462-482, 2022 06.
Article in English | MEDLINE | ID: mdl-34628598

ABSTRACT

Inflammasomes are known to contribute to brain damage after acute ischemic stroke (AIS). TAK1 is predominantly expressed in microglial cells and can regulate the NLRP3 inflammasome, but its impact on other inflammasomes including NLRC4 and AIM2 after AIS remains elusive. EPO has been shown to reduce NLRP3 protein levels in different disease models. Whether EPO-mediated neuroprotection after AIS is conveyed via an EPO/TAK1/inflammasome axis in microglia remains to be clarified. Subjecting mice deficient for TAK1 in microglia/macrophages (Mi/MΦ) to AIS revealed a significant reduction in infarct sizes and neurological impairments compared to the corresponding controls. Post-ischemic increased activation of TAK1, NLRP3, NLRC4, and AIM2 inflammasomes including their associated downstream cascades were markedly reduced upon deletion of Mi/MΦ TAK1. EPO administration improved clinical outcomes and dampened stroke-induced activation of TAK1 and inflammasome cascades, which was not evident after the deletion of Mi/MΦ TAK1. Pharmacological inhibition of NLRP3 in microglial BV-2 cells did not influence post-OGD IL-1ß levels, but increased NLRC4 and AIM2 protein levels, suggesting compensatory activities among inflammasomes. Overall, we provide evidence that Mi/MΦ TAK1 regulates the expression and activation of the NLRP3, NLRC4, AIM2 inflammasomes. Furthermore, EPO mitigated stroke-induced activation of TAK1 and inflammasomes, indicating that EPO conveyed neuroprotection might be mediated via an EPO/TAK1/inflammasome axis.


Subject(s)
Erythropoietin , Ischemic Stroke , Stroke , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Erythropoietin/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Ischemic Stroke/drug therapy , MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Stroke/metabolism
6.
Ann Neurol ; 90(5): 789-807, 2021 11.
Article in English | MEDLINE | ID: mdl-34476836

ABSTRACT

OBJECTIVE: Parkinson's disease (PD) manifests in motor dysfunction, non-motor symptoms, and eventual dementia (PDD). Neuropathological hallmarks include nigrostriatal neurodegeneration, Lewy body (LB) pathology, and neuroinflammation. Alpha-synuclein (α-syn), a primary component of LBs, is implicated in PD pathogenesis, accumulating, and aggregating in both familial and sporadic PD. However, as α-syn pathology is often comorbid with amyloid-beta (Aß) plaques and phosphorylated tau (pTau) tangles in PDD, it is still unclear whether α-syn is the primary cause of neurodegeneration in sporadic PDD. We aimed to determine how the absence of α-syn would affect PDD manifestation. METHODS: IFN-ß knockout (Ifnb-/- ) mice spontaneously develop progressive behavior abnormalities and neuropathology resembling PDD, notably with α-syn+ LBs. We generated Ifnb/Snca double knockout (DKO) mice and evaluated their behavior and neuropathology compared with wild-type (Wt), Ifnb-/- , and Snca-/- mice using immunohistochemistry, electron microscopy, immunoblots, qPCR, and modification of neuronal signaling. RESULTS: Ifnb/Snca DKO mice developed all clinical PDD-like behavioral manifestations induced by IFN-ß loss. Independently of α-syn expression, lack of IFN-ß alone induced Aß plaques, pTau tangles, and LB-like Aß+ /pTau+ inclusion bodies and neuroinflammation. IFN-ß loss caused significant elevated glial and neuronal TNF-α and neuronal TNFR1, associated with neurodegeneration. Restoring neuronal IFN-ß signaling or blocking TNFR1 rescued caspase 3/t-BID-mediated neuronal-death through upregulation of c-FLIPS and lowered intraneuronal Aß and pTau accumulation. INTERPRETATION: These findings increase our understanding of PD pathology and suggest that targeting α-syn alone is not sufficient to mitigate disease. Targeting specific aspects of neuroinflammation, such as aberrant neuronal TNF-α/TNFR1 or IFN-ß/IFNAR signaling, may attenuate disease. ANN NEUROL 2021;90:789-807.


Subject(s)
Neuroinflammatory Diseases/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Progression , Lewy Bodies/pathology , Lewy Body Disease/metabolism , Mice, Knockout , Neuroglia/pathology , Parkinson Disease/genetics , Plaque, Amyloid/metabolism , Tumor Necrosis Factor-alpha/deficiency
7.
EMBO J ; 40(6): e105123, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33555074

ABSTRACT

Similar to the brain, the eye is considered an immune-privileged organ where tissue-resident macrophages provide the major immune cell constituents. However, little is known about spatially restricted macrophage subsets within different eye compartments with regard to their origin, function, and fate during health and disease. Here, we combined single-cell analysis, fate mapping, parabiosis, and computational modeling to comprehensively examine myeloid subsets in distinct parts of the eye during homeostasis. This approach allowed us to identify myeloid subsets displaying diverse transcriptional states. During choroidal neovascularization, a typical hallmark of neovascular age-related macular degeneration (AMD), we recognized disease-specific macrophage subpopulations with distinct molecular signatures. Our results highlight the heterogeneity of myeloid subsets and their dynamics in the eye that provide new insights into the innate immune system in this organ which may offer new therapeutic targets for ophthalmological diseases.


Subject(s)
Choroid/blood supply , Eye/immunology , Macrophages/immunology , Myeloid Cells/immunology , Neovascularization, Physiologic/physiology , Animals , Choroid/embryology , Computational Biology , Computer Simulation , Eye/cytology , Eye/metabolism , Female , Homeostasis/immunology , Humans , Immunity, Innate/immunology , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Myeloid Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic/genetics
8.
Curr Eye Res ; 45(9): 1124-1135, 2020 09.
Article in English | MEDLINE | ID: mdl-31935132

ABSTRACT

PURPOSE: In glaucoma, an apoptotic death of retinal ganglion cells (RGCs) has been shown. However, little is known about other cell death mechanisms, like autophagy or necrosis. Therefore, we investigated these mechanisms in addition to antibody deposits in an experimental autoimmune glaucoma model. METHODS: Rats were immunized with a retinal ganglion cell-layer homogenate (RGA), while controls received sodium chloride. Untreated rats served as natїve group. After seven weeks, retinal cross-sections were stained with antibodies against RGCs (Brn-3a), apoptosis (cleaved caspase 2, cleaved caspase 3 as well as caspase 3, 8, and 9), autophagy (LC3BII and LAMP1), and necrosis (RIPK3) followed by cell counts. Autophagy was additionally visualized via transmission electron microscopy on retinal sections. Antibody deposits were also analyzed. RESULTS: We noted a RGC loss after RGA immunization compared to both control groups. Also, significantly more cleaved caspase 2+ RGCs were observed in RGA animals. More caspase 3 and 8 signals were noted in RGA retinas compared to both controls, while no changes were seen in regard to caspase 9. Furthermore, significantly more cleaved caspase 3+ cells were detected in RGA animals. We noted an increase of LC3BII+ and LAMP1+ autophagic cells in the RGA group, while no alterations were seen regarding necrotic RIPK3+ cells. Autophagic vesicles were observed via transmission electron microscopy. IgG staining revealed significant differences between the RGA group and controls concerning IgG deposits in the ganglion cell layer. CONCLUSIONS: Due to the novel results from this study, we conclude that IgG antibodies are involved in RGC loss in this model leading to apoptotic and autophagic cell loss. These results could help to develop new therapy strategies for glaucoma patients.


Subject(s)
Apoptosis/immunology , Autoantigens/immunology , Autoimmune Diseases/pathology , Autophagy/immunology , Disease Models, Animal , Glaucoma/pathology , Retinal Ganglion Cells/pathology , Animals , Autoantibodies/blood , Autoimmune Diseases/immunology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Glaucoma/immunology , Immunoglobulin G/blood , Lysosomal Membrane Proteins/metabolism , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Rats , Rats, Inbred Lew , Retinal Ganglion Cells/immunology
9.
J Clin Invest ; 130(3): 1315-1329, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31846439

ABSTRACT

Acute graft-versus-host disease (GVHD) can affect the central nervous system (CNS). The role of microglia in CNS-GVHD remains undefined. In agreement with microglia activation, we found that profound morphological changes and MHC-II and CD80 upregulation occurred upon GVHD induction. RNA sequencing-based analysis of purified microglia obtained from mice with CNS-GVHD revealed TNF upregulation. Selective TNF gene deletion in microglia of Cx3cr1creER Tnffl/- mice reduced MHC-II expression and decreased CNS T cell infiltrates and VCAM-1+ endothelial cells. GVHD increased microglia TGF-ß-activated kinase-1 (TAK1) activation and NF-κB/p38 MAPK signaling. Selective Tak1 deletion in microglia using Cx3cr1creER Tak1fl/fl mice resulted in reduced TNF production and microglial MHC-II and improved neurocognitive activity. Pharmacological TAK1 inhibition reduced TNF production and MHC-II expression by microglia, Th1 and Th17 T cell infiltrates, and VCAM-1+ endothelial cells and improved neurocognitive activity, without blocking graft-versus-leukemia effects. Consistent with these findings in mice, we observed increased activation and TNF production of microglia in the CNS of GVHD patients. In summary, we prove a role for microglia in CNS-GVHD, identify the TAK1/TNF/MHC-II axis as a mediator of CNS-GVHD, and provide a TAK1 inhibitor-based approach against GVHD-induced neurotoxicity.


Subject(s)
Central Nervous System Diseases/immunology , Graft vs Host Disease/immunology , Microglia/immunology , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology , Acute Disease , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Microglia/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Tumor Necrosis Factor-alpha/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
10.
J Exp Med ; 214(4): 905-917, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28330904

ABSTRACT

Monocytes are circulating mononuclear phagocytes, poised to extravasate to sites of inflammation and differentiate into monocyte-derived macrophages and dendritic cells. Tumor necrosis factor (TNF) and its receptors are up-regulated during monopoiesis and expressed by circulating monocytes, as well as effector monocytes infiltrating certain sites of inflammation, such as the spinal cord, during experimental autoimmune encephalomyelitis (EAE). In this study, using competitive in vitro and in vivo assays, we show that monocytes deficient for TNF or TNF receptors are outcompeted by their wild-type counterpart. Moreover, monocyte-autonomous TNF is critical for the function of these cells, as TNF ablation in monocytes/macrophages, but not in microglia, delayed the onset of EAE in challenged animals and was associated with reduced acute spinal cord infiltration of Ly6Chi effector monocytes. Collectively, our data reveal a previously unappreciated critical cell-autonomous role of TNF on monocytes for their survival, maintenance, and function.


Subject(s)
Inflammation/immunology , Monocytes/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Survival , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Mice, Inbred C57BL , Microglia/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology
11.
Front Immunol ; 8: 1930, 2017.
Article in English | MEDLINE | ID: mdl-29354133

ABSTRACT

Retinitis pigmentosa (RP) denotes a family of inherited blinding eye diseases characterized by progressive degeneration of rod and cone photoreceptors in the retina. In most cases, a rod-specific genetic defect results in early functional loss and degeneration of rods, which is followed by degeneration of cones and loss of daylight vision at later stages. Microglial cells, the immune cells of the central nervous system, are activated in retinas of RP patients and in several RP mouse models. However, it is still a matter of debate whether activated microglial cells may be responsible for the amplification of the typical degenerative processes. Here, we used Cngb1-/- mice, which represent a slow degenerative mouse model of RP, to investigate the extent of microglia activation in retinal degeneration. With a combination of FACS analysis, immunohistochemistry and gene expression analysis we established that microglia in the Cngb1-/- retina were already activated in an early, predegenerative stage of the disease. The evidence available so far suggests that early retinal microglia activation represents a first step in RP, which might initiate or accelerate photoreceptor degeneration.

12.
J Exp Med ; 213(7): 1163-74, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27325888

ABSTRACT

Pseudo-TORCH syndrome (PTS) is characterized by microcephaly, enlarged ventricles, cerebral calcification, and, occasionally, by systemic features at birth resembling the sequelae of congenital infection but in the absence of an infectious agent. Genetic defects resulting in activation of type 1 interferon (IFN) responses have been documented to cause Aicardi-Goutières syndrome, which is a cause of PTS. Ubiquitin-specific peptidase 18 (USP18) is a key negative regulator of type I IFN signaling. In this study, we identified loss-of-function recessive mutations of USP18 in five PTS patients from two unrelated families. Ex vivo brain autopsy material demonstrated innate immune inflammation with calcification and polymicrogyria. In vitro, patient fibroblasts displayed severely enhanced IFN-induced inflammation, which was completely rescued by lentiviral transduction of USP18. These findings add USP18 deficiency to the list of genetic disorders collectively termed type I interferonopathies. Moreover, USP18 deficiency represents the first genetic disorder of PTS caused by dysregulation of the response to type I IFNs. Therapeutically, this places USP18 as a promising target not only for genetic but also acquired IFN-mediated CNS disorders.


Subject(s)
Autoimmune Diseases of the Nervous System , Brain/immunology , Calcinosis , Endopeptidases/deficiency , Immunity, Innate , Interferon Type I/immunology , Microglia/immunology , Nervous System Malformations , Signal Transduction , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , Brain/pathology , Calcinosis/genetics , Calcinosis/immunology , Calcinosis/pathology , Endopeptidases/immunology , Female , Humans , Interferon Type I/genetics , Male , Microglia/pathology , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Nervous System Malformations/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Ubiquitin Thiolesterase
13.
Nat Immunol ; 17(7): 797-805, 2016 07.
Article in English | MEDLINE | ID: mdl-27135602

ABSTRACT

Perivascular, subdural meningeal and choroid plexus macrophages are non-parenchymal macrophages that mediate immune responses at brain boundaries. Although the origin of parenchymal microglia has recently been elucidated, much less is known about the precursors, the underlying transcriptional program and the dynamics of the other macrophages in the central nervous system (CNS). It was assumed that they have a high turnover from blood-borne monocytes. However, using parabiosis and fate-mapping approaches in mice, we found that CNS macrophages arose from hematopoietic precursors during embryonic development and established stable populations, with the notable exception of choroid plexus macrophages, which had dual origins and a shorter life span. The generation of CNS macrophages relied on the transcription factor PU.1, whereas the MYB, BATF3 and NR4A1 transcription factors were not required.


Subject(s)
Central Nervous System/immunology , Hematopoietic Stem Cells/physiology , Macrophages/physiology , Microglia/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/immunology , Parabiosis , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
14.
J Immunol ; 196(6): 2733-41, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26873993

ABSTRACT

Streptococci are common human colonizers with a species-specific mucocutaneous distribution. At the same time, they are among the most important and most virulent invasive bacterial pathogens. Thus, site-specific cellular innate immunity, which is predominantly executed by resident and invading myeloid cells, has to be adapted with respect to streptococcal sensing, handling, and response. In this article, we show that TLR13 is the critical mouse macrophage (MΦ) receptor in the response to group B Streptococcus, both in bone marrow-derived MΦs and in mature tissue MΦs, such as those residing in the lamina propria of the colon and the dermis, as well as in microglia. In contrast, TLR13 and its chaperone UNC-93B are dispensable for a potent cytokine response of blood monocytes to group B Streptococcus, although monocytes serve as the key progenitors of intestinal and dermal MΦs. Furthermore, a specific role for TLR13 with respect to MΦ function is supported by the response to staphylococci, where TLR13 and UNC-93B limit the cytokine response in bone marrow-derived MΦs and microglia, but not in dermal MΦs. In summary, TLR13 is a critical and site-specific receptor in the single MΦ response to ß-hemolytic streptococci.


Subject(s)
Macrophages/physiology , Membrane Transport Proteins/metabolism , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Toll-Like Receptors/metabolism , Animals , Colon/pathology , Cytokines/metabolism , Hemolysis , Host-Pathogen Interactions , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Macrophages/microbiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/pathology , Organ Specificity , Skin/pathology , Toll-Like Receptors/genetics
15.
Curr Opin Neurobiol ; 36: 38-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26397019

ABSTRACT

Type I interferons (IFN) are pleiotropic cytokines originally described as molecules used for communication between cells to trigger the protective defenses against viral infections. Upon activation, type I IFN can be produced locally in the central nervous system (CNS) from a number of different cell types including microglia, the CNS-resident macrophages. Increased type I IFN production and signaling in microglia are critically important to limit viral infection and disease progression in multiple sclerosis. However, recent findings suggest that even baseline levels of constitutive IFN expression and secretion are important for homeostasis of the CNS. In fact, in the absence of viral particles chronic elevation of IFN I may be tremendously harmful for the CNS, as assumed for patients suffering from Aicardi-Goutières syndrome, Cree encephalitis or other type I interferonopathies. The highly diverse nature of type I IFN for brain homeostasis during health and disease will be discussed in this report.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity/immunology , Central Nervous System/immunology , Interferon Type I/immunology , Microglia/immunology , Homeostasis/immunology , Humans , Multiple Sclerosis/immunology , Myelin Sheath/immunology , Nervous System Malformations/immunology , Phagocytosis/immunology , Signal Transduction/immunology
16.
Cell ; 163(2): 324-39, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451483

ABSTRACT

Neurodegenerative diseases have been linked to inflammation, but whether altered immunomodulation plays a causative role in neurodegeneration is not clear. We show that lack of cytokine interferon-ß (IFN-ß) signaling causes spontaneous neurodegeneration in the absence of neurodegenerative disease-causing mutant proteins. Mice lacking Ifnb function exhibited motor and cognitive learning impairments with accompanying α-synuclein-containing Lewy bodies in the brain, as well as a reduction in dopaminergic neurons and defective dopamine signaling in the nigrostriatal region. Lack of IFN-ß signaling caused defects in neuronal autophagy prior to α-synucleinopathy, which was associated with accumulation of senescent mitochondria. Recombinant IFN-ß promoted neurite growth and branching, autophagy flux, and α-synuclein degradation in neurons. In addition, lentiviral IFN-ß overexpression prevented dopaminergic neuron loss in a familial Parkinson's disease model. These results indicate a protective role for IFN-ß in neuronal homeostasis and validate Ifnb mutant mice as a model for sporadic Lewy body and Parkinson's disease dementia.


Subject(s)
Interferon-beta/metabolism , Neurons/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Autophagy , Disease Models, Animal , Genetic Therapy , Interferon-beta/genetics , Interferon-beta/therapeutic use , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Mice , Mice, Inbred C57BL , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Transcriptome , alpha-Synuclein/metabolism
17.
EMBO J ; 34(12): 1612-29, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25896511

ABSTRACT

Microglia are tissue macrophages of the central nervous system (CNS) that control tissue homeostasis. Microglia dysregulation is thought to be causal for a group of neuropsychiatric, neurodegenerative and neuroinflammatory diseases, called "microgliopathies". However, how the intracellular stimulation machinery in microglia is controlled is poorly understood. Here, we identified the ubiquitin-specific protease (Usp) 18 in white matter microglia that essentially contributes to microglial quiescence. We further found that microglial Usp18 negatively regulates the activation of Stat1 and concomitant induction of interferon-induced genes, thereby terminating IFN signaling. The Usp18-mediated control was independent from its catalytic activity but instead required the interaction with Ifnar2. Additionally, the absence of Ifnar1 restored microglial activation, indicating a tonic IFN signal which needs to be negatively controlled by Usp18 under non-diseased conditions. These results identify Usp18 as a critical negative regulator of microglia activation and demonstrate a protective role of Usp18 for microglia function by regulating the Ifnar pathway. The findings establish Usp18 as a new molecule preventing destructive microgliopathy.


Subject(s)
Brain/metabolism , Endopeptidases/deficiency , Interferons/metabolism , Microglia/metabolism , Models, Neurological , Signal Transduction/physiology , Animals , Blotting, Western , Cloning, Molecular , DNA Primers/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Histological Techniques , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Statistics, Nonparametric , Ubiquitin Thiolesterase
18.
Proc Natl Acad Sci U S A ; 112(5): 1577-82, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605921

ABSTRACT

Protein modification by the ubiquitin-like protein ISG15 is an interferon (IFN) effector system, which plays a major role in antiviral defense. ISG15 modification is counteracted by the isopeptidase USP18, a major negative regulator of IFN signaling, which was also shown to exert its regulatory function in an isopeptidase-independent manner. To dissect enzymatic and nonenzymatic functions of USP18 in vivo, we generated knock-in mice (USP18(C61A/C61A)) expressing enzymatically inactive USP18. USP18(C61A/C61A) mice displayed increased levels of ISG15 conjugates, validating that USP18 is a major ISG15 isopeptidase in vivo. Unlike USP18(-/-) mice, USP18(C61A/C61A) animals did not exhibit morphological abnormalities, fatal IFN hypersensitivity, or increased lethality, clearly showing that major USP18 functions are unrelated to its protease activity. Strikingly, elevated ISGylation in USP18(C61A/C61A) mice was accompanied by increased viral resistance against vaccinia virus and influenza B virus infections. Enhanced resistance upon influenza B infection in USP18(C61A/C61A) mice was completely reversed in USP18(C61A/C61A) mice, which additionally lack ISG15, providing evidence that the observed reduction in viral titers is ISG15 dependent. These results suggest that increasing ISGylation by specific inhibition of USP18 protease activity could constitute a promising antiviral strategy with only a minimal risk of severe adverse effects.


Subject(s)
Cytokines/metabolism , Drug Resistance, Viral , Ubiquitin Thiolesterase/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Influenza B virus/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ubiquitins/metabolism
19.
Trends Immunol ; 35(4): 139-40, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24636251

ABSTRACT

Microglia, the brain resident macrophages, are first responders to any violation of central nervous system (CNS) homeostasis. A recent paper in Cell shows that the specific depletion of microglia causes deficits in learning-induced synaptic remodeling through paucity of the microglial neurotrophin brain-derived neurotrophic factor (BDNF).


Subject(s)
Brain/cytology , Brain/physiology , Microglia/cytology , Microglia/physiology , Animals , Humans
20.
Nat Neurosci ; 16(11): 1618-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077561

ABSTRACT

Microglia are brain macrophages and, as such, key immune-competent cells that can respond to environmental changes. Understanding the mechanisms of microglia-specific responses during pathologies is hence vital for reducing disease burden. The definition of microglial functions has so far been hampered by the lack of genetic in vivo approaches that allow discrimination of microglia from closely related peripheral macrophage populations in the body. Here we introduce a mouse experimental system that specifically targets microglia to examine the role of a mitogen-associated protein kinase kinase kinase (MAP3K), transforming growth factor (TGF)-ß-activated kinase 1 (TAK1), during autoimmune inflammation. Conditional depletion of TAK1 in microglia only, not in neuroectodermal cells, suppressed disease, significantly reduced CNS inflammation and diminished axonal and myelin damage by cell-autonomous inhibition of the NF-κB, JNK and ERK1/2 pathways. Thus, we found TAK1 to be pivotal in CNS autoimmunity, and we present a tool for future investigations of microglial function in the CNS.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/immunology , Gene Targeting , MAP Kinase Kinase Kinases/metabolism , Microglia/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/physiology , Brain/metabolism , CD11c Antigen/genetics , CX3C Chemokine Receptor 1 , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Estrogen Antagonists/pharmacology , Gene Expression Regulation/drug effects , Luminescent Proteins/genetics , Lymph Nodes/pathology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Nerve Tissue Proteins/metabolism , RNA, Untranslated/genetics , Receptors, Chemokine/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...