Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31915282

ABSTRACT

The virus-induced signaling adaptor (VISA) complex plays a critical role in the innate immune response to RNA viruses. However, the mechanism of VISA complex formation remains unclear. Here, we demonstrate that thioredoxin 2 (TRX2) interacts with VISA at mitochondria both in vivo and in vitro Knockdown and knockout of TRX2 enhanced the formation of the VISA-associated complex, as well as virus-triggered activation of interferon regulatory factor 3 (IRF3) and transcription of the interferon beta 1 (IFNB1) gene. TRX2 inhibits the formation of VISA aggregates by repressing reactive oxygen species (ROS) production, thereby disrupting the assembly of the VISA complex. Furthermore, our data suggest that the C93 residue of TRX2 is essential for inhibition of VISA aggregation, whereas the C283 residue of VISA is required for VISA aggregation. Collectively, these findings uncover a novel mechanism of TRX2 that negatively regulates VISA complex formation.IMPORTANCE The VISA-associated complex plays pivotal roles in inducing type I interferons (IFNs) and eliciting the innate antiviral response. Many host proteins are identified as VISA-associated-complex proteins, but how VISA complex formation is regulated by host proteins remains enigmatic. We identified the TRX2 protein as an important regulator of VISA complex formation. Knockout of TRX2 increases virus- or poly(I·C)-triggered induction of type I IFNs at the VISA level. Mechanistically, TRX2 inhibits the production of ROS at its C93 site, which impairs VISA aggregates at its C283 site, and subsequently impedes the assembly of the VISA complex. Our findings suggest that TRX2 plays an important role in the regulation of VISA complex assembly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Viral , Immunity, Innate , Mitochondrial Proteins/metabolism , Respirovirus Infections/immunology , Sendai virus/immunology , Thioredoxins/metabolism , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon beta-1a/metabolism , Poly I-C/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , THP-1 Cells
2.
CNS Drugs ; 33(12): 1187-1199, 2019 12.
Article in English | MEDLINE | ID: mdl-31686407

ABSTRACT

Vitamin D and its main active metabolite 1,25-dihydroxyvitamin D serve a crucial role in maintenance of a healthy calcium metabolism, yet have additional roles in immune and central nervous system cell homeostasis. Serum levels of 25-hydroxyvitamin D are a biomarker of future disease activity in patients with early relapsing-remitting multiple sclerosis (RRMS), and vitamin D supplementation in patients with low circulating 25-dihydroxyvitamin D levels has been anticipated as a potential efficacious treatment strategy. The results of the first large randomized clinical trials (RCTs), the SOLAR and CHOLINE studies, have now been published. The SOLAR study compared 14,000 IU of vitamin D3 (cholecalciferol) per day with placebo for 48 weeks in 232 randomized patients, whereas CHOLINE compared vitamin D3 100,000 IU every other week with placebo for 96 weeks in 129 randomized patients. All patients in both studies also used interferon-ß-1a. None of the studies met their primary endpoints, which were no evidence of disease activity (NEDA-3) at 48 weeks in SOLAR and annualized relapse rate at 96 weeks in CHOLINE. Both studies did, however, suggest modest effects on secondary endpoints. Thus, vitamin D reduced the number of new or enlarging lesions and new T2 lesions in SOLAR, and the annualized relapse rate and number of new T1 lesions, volume of hypointense T1 lesions, and disability progression in the 90 patients who completed 96 weeks' follow-up in CHOLINE. We conclude that none of the RCTs on vitamin supplementation in MS have met their primary clinical endpoint in the intention to treat cohorts. This contrasts the observation studies, where each 25 nmol/l increase in 25-hydroxyvitamin D levels were associated with 14-34% reduced relapse risk and 15-50% reduced risk of new lesions on magnetic resonnance imaging. This discrepancy may have several explanations, including confounding and reverse causality in the observational studies. The power calculations of the RCTs have been based on the observational studies, and the RCTs may have been underpowered to detect less prominent yet important effects of vitamin D supplementation. Although the effect of vitamin D supplementation is uncertain and less pronounced than suggested by observational studies, current evidence still support that people with MS should avoid vitamin D insufficiency, and preferentially aim for vitamin D levels around 100 nmol/L or somewhat higher.


Subject(s)
Multiple Sclerosis/metabolism , Vitamin D/metabolism , Animals , Dietary Supplements , Disease Progression , Humans , Interferon beta-1a/metabolism , Observational Studies as Topic , Randomized Controlled Trials as Topic , Vitamin D/analogs & derivatives
3.
J Immunol ; 199(8): 2618-2623, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904124

ABSTRACT

The transitional stage of B cell development is a formative stage in the spleen where autoreactive specificities are censored as B cells gain immune competence, but the intrinsic and extrinsic factors regulating survival of transitional stage 1 (T1) B cells are unknown. We report that B cell expression of IFN-ß is required for optimal survival and TLR7 responses of transitional B cells in the spleen and was overexpressed in T1 B cells from BXD2 lupus-prone mice. Single-cell gene expression analysis of B6 Ifnb+/+ versus B6 Ifnb-/- T1 B cells revealed heterogeneous expression of Ifnb in wild-type B cells and distinct gene expression patterns associated with endogenous IFN-ß. Single-cell analysis of BXD2 T1 B cells revealed that Ifnb is expressed in early T1 B cell development with subsequent upregulation of Tlr7 and Ifna1 Together, these data suggest that T1 B cell expression of IFN-ß plays a key role in regulating responsiveness to external factors.


Subject(s)
B-Lymphocytes/immunology , Interferon-beta/metabolism , Lupus Nephritis/immunology , Precursor Cells, B-Lymphoid/immunology , Spleen/immunology , Animals , B-Lymphocyte Subsets/immunology , Cell Differentiation , Cell Survival , Disease Susceptibility , Interferon beta-1a/genetics , Interferon beta-1a/metabolism , Interferon-alpha , Lymphocyte Activation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Single-Cell Analysis , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
4.
J Immunol ; 197(6): 2338-52, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27511736

ABSTRACT

Type I IFN (IFN-α/ß)-driven immune responses to acute viral infection are critical to counter replication and prevent dissemination. However, the mechanisms underlying host resistance to HSV type 1 (HSV-1) are incompletely understood. In this study, we show that mice with deficiencies in IFN-α/ß signaling or stimulator of IFN genes (STING) exhibit exacerbated neurovirulence and atypical lymphotropic dissemination of HSV-1 following ocular infection. Synergy between IFN-α/ß signaling and efficacy of early adaptive immune responses to HSV-1 were dissected using bone marrow chimeras and adoptive cell transfer approaches to profile clonal expansion, effector function, and recruitment of HSV-specific CD8(+) T cells. Lymphotropic viral dissemination was commensurate with abrogated CD8(+) T cell responses and pathological alterations of fibroblastic reticular cell networks in the draining lymph nodes. Our results show that resistance to HSV-1 in the trigeminal ganglia during acute infection is conferred in part by STING and IFN-α/ß signaling in both bone marrow-derived and -resident cells, which coalesce to support a robust HSV-1-specific CD8(+) T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Interferon beta-1a/metabolism , Interferon-alpha/metabolism , Lymphadenitis/immunology , Lymphadenitis/virology , Membrane Proteins/metabolism , Adaptive Immunity , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/pathology , Eye/virology , Herpes Simplex/immunology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Interferon beta-1a/genetics , Interferon beta-1a/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Lymphadenitis/physiopathology , Membrane Proteins/deficiency , Mice , Signal Transduction , Trigeminal Ganglion/immunology , Trigeminal Ganglion/physiopathology , Trigeminal Ganglion/virology , Virus Replication
5.
J Interferon Cytokine Res ; 36(9): 534-41, 2016 09.
Article in English | MEDLINE | ID: mdl-27348209

ABSTRACT

Human type I Interferons (IFN-ß, IFN-ɛ, IFN-κ, IFN-ω, and 12 subtypes of IFN-α) are a family of pleiotropic cytokines with antiviral, antiproliferative, and immunomodulatory activities. They signal through the same cell surface receptors, IFNAR1 and IFNAR2, yet evoking markedly differential potency. One differentiating factor of IFN-ß from other type I interferons is the presence of a consensus sequence (NG) for deamidation. Comparing almost completely deamidated IFN-ß-1a with untreated IFN-ß-1a, this present study reports the increased activities in 3 in-vitro bioassays testing the antiviral, antiproliferative, and immunomodulatory properties, respectively, of the molecule. Deamidated IFN-ß-1a has the potential to improve current therapies in multiple sclerosis, and its ability to potentiate the MHC-Class I expression suggests a clinical benefit in diseases where the downmodulation of the MHC-class I expression plays a role (eg, in immuno-oncology combination therapies or antiviral agents). The present study on IFN-ß deamidation adds a new prospective on deamidation as part of a posttranslational modification code that allows the modulation of the biological properties of proteins. Moreover, it underlines the unique IFN-ß-1a properties that differentiate this molecule from other members of the type I interferon family.


Subject(s)
Interferon beta-1a/metabolism , Interferon beta-1a/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , CHO Cells , Circular Dichroism , Cricetulus , Humans , Immunologic Factors/chemistry , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Interferon beta-1a/chemistry , Oxidation-Reduction , Peptide Fragments , Protein Denaturation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
6.
Biomed Res Int ; 2016: 4570351, 2016.
Article in English | MEDLINE | ID: mdl-28078290

ABSTRACT

The role of nitric oxide and its reactive derivatives (NO x ) is well known in the pathogenesis of multiple sclerosis, which is an inflammatory disease while NO x seems to be important in coordinating inflammatory response. The purpose of the present study was to assess serum NO x as one of the nitrogen species and inflammatory parameters in relapsing-remitting multiple sclerosis patients and to compare the effectiveness of various types of disease-modifying therapies that reduce nitric oxide and inflammatory biomarkers. Elevated NO x level was observed in patients who received the first-line disease-modifying therapy (interferons beta-1a and beta-1b) in comparison with the subjects treated with the second-line disease-modifying therapy (natalizumab; fingolimod) and healthy controls without significant differences in C-reactive protein and interleukin-1 beta. A negative correlation was observed between serum NO x level and the duration of multiple sclerosis confirmed in the whole study population and in subjects treated with the first-line agents. Only serum NO x , concentration could reveal a potential efficacy of disease-modifying therapy with a better reduction in NO x level due to the second-line agents of disease-modifying therapy.


Subject(s)
Inflammation/blood , Inflammation/drug therapy , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Reactive Nitrogen Species/blood , Adolescent , Adult , Biomarkers/blood , Fingolimod Hydrochloride/administration & dosage , Humans , Inflammation/pathology , Interferon beta-1a/administration & dosage , Interferon beta-1a/metabolism , Interferon beta-1b/administration & dosage , Interferon beta-1b/metabolism , Middle Aged , Multiple Sclerosis/pathology , Natalizumab/administration & dosage , Nitric Oxide/blood
7.
BMC Pharmacol Toxicol ; 16: 25, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26392348

ABSTRACT

BACKGROUND: The innate immune receptor RIG-I detects viral RNA within the cytosol of infected cells. Activation of RIG-I leads to the induction of antiviral cytokines, in particular type I interferon, the inhibition of a T(H)17 response as well as to the suppression of tumor growth. Therefore, RIG-I is a promising drug target for the treatment of cancer as well as multiple sclerosis. A specific ligand for RIG-I is currently in preclinical testing. The first-in-human trial will need to be carefully designed to avoid an overshooting cytokine response. Therefore, the ResI study was set up to analyze the human immune response to standard treatment with recombinant interferon-beta to establish biomarkers for safety and efficacy of the upcoming first-in-human trial investigating the RIG-I ligand. METHODS/DESIGN: ResI is a single center, prospective, open label, non-randomized phase I clinical trial. Three different cohorts (20 healthy volunteers, 20 patients with RRMS and ongoing interferon-beta treatment and 10 patients starting on interferon-beta) will receive standard interferon-beta-1a therapy for nine days. The study will be conducted according to the principles of the german medicinal products act, ICH-GCP, and the Declaration of Helsinki on the phase I unit of the Institute of Clinical Chemistry and Clinical Pharmacology and in the Department of Neurology, both University Hospital Bonn. Interferon-beta-induced cytokine levels, surface marker on immune cells, mRNA- and miRNA-expression as well as psychometric response will be investigated as target variables. DISCUSSION: The ResI study will assess biomarkers in response to interferon-ß treatment to guide the dose steps within the first-in-human trial with a newly developed RIG-I ligand. Thus, ResI is a biomarker study to enhance the safety of the clinical development of a first-in-class compound. The data can additionally be used for the development of other therapies based on type I interferon induction such as TLR ligands. Moreover, it will help to understand the interferon-beta induced immune response in a controlled in vivo setting in the human system. TRIAL REGISTRATION: clinicaltrials.gov ID NCT02364986.


Subject(s)
Interferon Type I/metabolism , Interferon beta-1a/therapeutic use , MicroRNAs/genetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Aged , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression/drug effects , Humans , Interferon beta-1a/genetics , Interferon beta-1a/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/metabolism , Outcome Assessment, Health Care , Prospective Studies , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic , Recombinant Proteins/therapeutic use , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...