Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
Cell ; 181(5): 1080-1096.e19, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32380006

ABSTRACT

Environmental signals shape host physiology and fitness. Microbiota-derived cues are required to program conventional dendritic cells (cDCs) during the steady state so that they can promptly respond and initiate adaptive immune responses when encountering pathogens. However, the molecular underpinnings of microbiota-guided instructive programs are not well understood. Here, we report that the indigenous microbiota controls constitutive production of type I interferons (IFN-I) by plasmacytoid DCs. Using genome-wide analysis of transcriptional and epigenetic regulomes of cDCs from germ-free and IFN-I receptor (IFNAR)-deficient mice, we found that tonic IFNAR signaling instructs a specific epigenomic and metabolic basal state that poises cDCs for future pathogen combat. However, such beneficial biological function comes with a trade-off. Instructed cDCs can prime T cell responses against harmless peripheral antigens when removing roadblocks of peripheral tolerance. Our data provide fresh insights into the evolutionary trade-offs that come with successful adaptation of vertebrates to their microbial environment.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Microbiota/immunology , Adaptive Immunity/immunology , Adaptive Immunity/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/microbiology , Female , Male , Mice , Mice, Inbred C57BL , Microbiota/physiology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
2.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096319

ABSTRACT

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Subject(s)
Brain/cytology , Chemokine CXCL10/immunology , Cognition Disorders/genetics , Endothelial Cells/immunology , Epithelial Cells/immunology , Illness Behavior/physiology , Receptor, Interferon alpha-beta/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/immunology , Cell Communication/immunology , Cells, Cultured , Cognition Disorders/psychology , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Endothelium/cytology , Endothelium/immunology , Epithelium/immunology , Interferon Type I/therapeutic use , Interferon-Induced Helicase, IFIH1 , Male , Mice , RNA, Double-Stranded/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, CXCR3/immunology , Signal Transduction/immunology , Virus Diseases/immunology
3.
Small ; 20(10): e2306892, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37867244

ABSTRACT

Poly(I:C) is a synthetic analogue of dsRNA capable of activating both TLR3 and RLRs, such as MDA-5 and RIG-I, as pathogen recognition receptors. While poly(I:C) is known to provoke a robust type I IFN, type III IFN, and Th1 cytokine response, its therapeutic use as a vaccine adjuvant is limited due to its vulnerability to nucleases and poor uptake by immune cells. is encapsulated poly(I:C) into lipid nanoparticles (LNPs) containing an ionizable cationic lipid that can electrostatically interact with poly(I:C). LNP-formulated poly(I:C) triggered both lysosomal TLR3 and cytoplasmic RLRs, in vitro and in vivo, whereas poly(I:C) in an unformulated soluble form only triggered endosomal-localized TLR3. Administration of LNP-formulated poly(I:C) in mouse models led to efficient translocation to lymphoid tissue and concurrent innate immune activation following intramuscular (IM) administration, resulting in a significant increase in innate immune activation compared to unformulated soluble poly(I:C). When used as an adjuvant for recombinant full-length SARS-CoV-2 spike protein, LNP-formulated poly(I:C) elicited potent anti-spike antibody titers, surpassing those of unformulated soluble poly(I:C) by orders of magnitude and offered complete protection against a SARS-CoV-2 viral challenge in vivo, and serum from these mice are capable of significantly reducing viral infection in vitro.


Subject(s)
Liposomes , Nanoparticles , Poly I-C , Spike Glycoprotein, Coronavirus , Toll-Like Receptor 3 , Animals , Mice , Humans , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Adjuvants, Immunologic/pharmacology
4.
Immunity ; 42(2): 332-343, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692705

ABSTRACT

Dysfunction in Ataxia-telangiectasia mutated (ATM), a central component of the DNA repair machinery, results in Ataxia Telangiectasia (AT), a cancer-prone disease with a variety of inflammatory manifestations. By analyzing AT patient samples and Atm(-/-) mice, we found that unrepaired DNA lesions induce type I interferons (IFNs), resulting in enhanced anti-viral and anti-bacterial responses in Atm(-/-) mice. Priming of the type I interferon system by DNA damage involved release of DNA into the cytoplasm where it activated the cytosolic DNA sensing STING-mediated pathway, which in turn enhanced responses to innate stimuli by activating the expression of Toll-like receptors, RIG-I-like receptors, cytoplasmic DNA sensors, and their downstream signaling partners. This study provides a potential explanation for the inflammatory phenotype of AT patients and establishes damaged DNA as a cell intrinsic danger signal that primes the innate immune system for a rapid and amplified response to microbial and environmental threats.


Subject(s)
Ataxia Telangiectasia/immunology , DNA Damage , DNA/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Membrane Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Bone Marrow Cells/immunology , Cell Line , Cytosol/immunology , Cytosol/microbiology , DNA Repair/genetics , Enzyme Activation/immunology , HEK293 Cells , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Interferon-gamma/biosynthesis , Macrophages/immunology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Neuroinflammation ; 20(1): 213, 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37737190

ABSTRACT

BACKGROUND: Type I interferons (IFN-I) are fundamental in controlling viral infections but fatal interferonopathy is restricted in the immune-privileged central nervous system (CNS). In contrast to the well-established role of Interferon Regulatory Factor 7 (IRF7) in the regulation of IFN-I response in the periphery, little is known about the specific function in the CNS. METHODS: To investigate the role for IRF7 in antiviral response during neurotropic virus infection, mice deficient for IRF3 and IRF7 were infected systemically with Langat virus (LGTV). Viral burden and IFN-I response was analyzed in the periphery and the CNS by focus formation assay, RT-PCR, immunohistochemistry and in vivo imaging. Microglia and infiltration of CNS-infiltration of immune cells were characterized by flow cytometry. RESULTS: Here, we demonstrate that during infection with the neurotropic Langat virus (LGTV), an attenuated member of the tick-borne encephalitis virus (TBEV) subgroup, neurons do not rely on IRF7 for cell-intrinsic antiviral resistance and IFN-I induction. An increased viral replication in IRF7-deficient mice suggests an indirect antiviral mechanism. Astrocytes rely on IRF7 to establish a cell-autonomous antiviral response. Notably, the loss of IRF7 particularly in astrocytes resulted in a high IFN-I production. Sustained production of IFN-I in astrocytes is independent of an IRF7-mediated positive feedback loop. CONCLUSION: IFN-I induction in the CNS is profoundly regulated in a cell type-specific fashion.


Subject(s)
Encephalitis, Tick-Borne , Interferon Regulatory Factor-7 , Interferon Type I , Animals , Mice , Antibodies , Astrocytes , Central Nervous System , Interferon Regulatory Factor-7/genetics , Encephalitis, Tick-Borne/immunology
6.
Hum Mol Genet ; 29(16): 2662-2673, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32644125

ABSTRACT

Spinal muscular atrophy (SMA) is a fatal neurodegenerative disease of newborns and children caused by mutations or deletions of the survival of motoneuron gene 1 resulting in low levels of the SMN protein. While neuromuscular degeneration is the cardinal symptom of the disease, the reduction of the ubiquitously expressed SMN additionally elicits non-motoneuron symptoms. Impaired bone development is a key feature of SMA, but it is yet unknown whether this is an indirect functional consequence of muscle weakness or caused by bone-intrinsic mechanisms. Therefore, we radiologically examined SMA patients in a prospective, non-randomized cohort study characterizing bone size and bone mineral density (BMD) and performed equivalent measurements in pre-symptomatic SMA mice. BMD as well as lumbar vertebral body size were significantly reduced in SMA patients. This growth defect but not BMD reduction was confirmed in SMA mice by µCT before the onset of neuromuscular symptoms indicating that it is at least partially independent of neuromuscular degeneration. Interestingly, the number of chondroblasts in the hypertrophic zone of the growth plate was significantly reduced. This was underlined by RNAseq and expression data from developing SMA mice vertebral bodies, which revealed molecular changes related to cell division and cartilage remodeling. Together, these findings suggest a bone intrinsic defect in SMA. This phenotype may not be rescued by novel drugs that enhance SMN levels in the central nervous system only.


Subject(s)
Bone Development/genetics , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Neurodegenerative Diseases/genetics , Survival of Motor Neuron 1 Protein/genetics , Adolescent , Animals , Bone Density/genetics , Cartilage/growth & development , Cartilage/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Child , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/diagnostic imaging , Muscular Atrophy, Spinal/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/pathology , Phenotype
7.
Mol Ther ; 29(4): 1370-1381, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33484964

ABSTRACT

Synthetic mRNAs are an appealing platform with multiple biomedical applications ranging from protein replacement therapy to vaccination. In comparison with conventional mRNA, synthetic self-amplifying mRNAs (sa-mRNAs) are gaining interest because of their higher and longer-lasting expression. However, sa-mRNAs also elicit an innate immune response, which may complicate their clinical application. Approaches to reduce the innate immunity of sa-mRNAs have not been studied in detail. Here we investigated, in vivo, the effect of several innate immune inhibitors and a novel cellulose-based mRNA purification approach on the type I interferon (IFN) response and the translation and vaccination efficacy of our formerly developed sa-mRNA vaccine against Zika virus. Among the investigated inhibitors, we found that corticosteroids and especially topical application of clobetasol at the sa-mRNA injection site was the most efficient in suppressing the type I IFN response and increasing the translation of sa-mRNA. However, clobetasol prevented formation of antibodies against sa-mRNA-encoded antigens and should therefore be avoided in a vaccination context. Residual dsRNA by-products of the in vitro transcription reaction are known inducers of immediate type I IFN responses. We additionally demonstrate a drastic reduction of these dsRNA by-products upon cellulose-based purification, reducing the innate immune response and improving sa-mRNA vaccination efficacy.


Subject(s)
Immunity, Innate/genetics , RNA, Messenger/genetics , Vaccination , Zika Virus Infection/drug therapy , Adrenal Cortex Hormones/chemistry , Cellulose/chemistry , Clobetasol/pharmacology , Gene Expression Regulation/genetics , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Protein Biosynthesis/drug effects , Protein Biosynthesis/immunology , RNA, Messenger/chemical synthesis , RNA, Messenger/chemistry , RNA, Messenger/pharmacology , Zika Virus/drug effects , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/virology
8.
J Am Chem Soc ; 143(26): 9872-9883, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34166595

ABSTRACT

Small-molecular Toll-like receptor 7/8 (TLR7/8) agonists hold promise as immune modulators for a variety of immune therapeutic purposes including cancer therapy or vaccination. However, due to their rapid systemic distribution causing difficult-to-control inflammatory off-target effects, their application is still problematic, in particular systemically. To address this problem, we designed and robustly fabricated pH-responsive nanogels serving as versatile immunodrug nanocarriers for safe delivery of TLR7/8-stimulating imidazoquinolines after intravenous administration. To this aim, a primary amine-reactive methacrylamide monomer bearing a pendant squaric ester amide is introduced, which is polymerized under controlled RAFT polymerization conditions. Corresponding PEG-derived squaric ester amide block copolymers self-assemble into precursor micelles in polar protic solvents. Their cores are amine-reactive and can sequentially be transformed by acid-sensitive cross-linkers, dyes, and imidazoquinolines. Remaining squaric ester amides are hydrophilized affording fully hydrophilic nanogels with profound stability in human plasma but stimuli-responsive degradation upon exposure to endolysosomal pH conditions. The immunomodulatory behavior of the imidazoquinolines alone or conjugated to the nanogels was demonstrated by macrophages in vitro. In vivo, however, we observed a remarkable impact of the nanogel: After intravenous injection, a spatially controlled immunostimulatory activity was evident in the spleen, whereas systemic off-target inflammatory responses triggered by the small-molecular imidazoquinoline analogue were absent. These findings underline the potential of squaric ester-based, pH-degradable nanogels as a promising platform to permit intravenous administration routes of small-molecular TLR7/8 agonists and, thus, the opportunity to explore their adjuvant potency for systemic vaccination or cancer immunotherapy purposes.


Subject(s)
Adjuvants, Immunologic/chemistry , Esters/chemistry , Nanogels/chemistry , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , Drug Carriers/chemistry , Drug Liberation , Humans , Hydrogen-Ion Concentration , Immunotherapy , Mice, Inbred BALB C , Micelles , Optical Imaging , Polymerization , Polymers/chemistry
9.
Immunity ; 37(1): 171-86, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22749822

ABSTRACT

Mononuclear phagocytes are an important component of an innate immune system perceived as a system ready to react upon encounter of pathogens. Here, we show that in response to microbial stimulation, mononuclear phagocytes residing in nonmucosal lymphoid organs of germ-free mice failed to induce expression of a set of inflammatory response genes, including those encoding the various type I interferons (IFN-I). Consequently, NK cell priming and antiviral immunity were severely compromised. Whereas pattern recognition receptor signaling and nuclear translocation of the transcription factors NF-κB and IRF3 were normal in mononuclear phagocytes of germ-free mice, binding to their respective cytokine promoters was impaired, which correlated with the absence of activating histone marks. Our data reveal a previously unrecognized role for postnatally colonizing microbiota in the introduction of chromatin level changes in the mononuclear phagocyte system, thereby poising expression of central inflammatory genes to initiate a powerful systemic immune response during viral infection.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Metagenome/immunology , Phagocytes/immunology , Animals , Cytokines/biosynthesis , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Phagocytes/metabolism , Virus Diseases/immunology
10.
Angew Chem Int Ed Engl ; 60(17): 9467-9473, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33464672

ABSTRACT

The search for vaccines that protect from severe morbidity and mortality because of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19) is a race against the clock and the virus. Here we describe an amphiphilic imidazoquinoline (IMDQ-PEG-CHOL) TLR7/8 adjuvant, consisting of an imidazoquinoline conjugated to the chain end of a cholesterol-poly(ethylene glycol) macromolecular amphiphile. It is water-soluble and exhibits massive translocation to lymph nodes upon local administration through binding to albumin, affording localized innate immune activation and reduction in systemic inflammation. The adjuvanticity of IMDQ-PEG-CHOL was validated in a licensed vaccine setting (quadrivalent influenza vaccine) and an experimental trimeric recombinant SARS-CoV-2 spike protein vaccine, showing robust IgG2a and IgG1 antibody titers in mice that could neutralize viral infection in vitro and in vivo in a mouse model.


Subject(s)
Adjuvants, Immunologic/therapeutic use , COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , Imidazoles/therapeutic use , Immunity, Innate/drug effects , Quinolines/therapeutic use , Animals , COVID-19 Vaccines/immunology , Cholesterol/analogs & derivatives , Cholesterol/immunology , Cholesterol/therapeutic use , Female , Humans , Imidazoles/immunology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Membrane Glycoproteins/agonists , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Polyethylene Glycols/therapeutic use , Quinolines/immunology , Recombinant Proteins/immunology , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/immunology , Surface-Active Agents/therapeutic use , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists
11.
J Am Chem Soc ; 142(28): 12133-12139, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32524819

ABSTRACT

Synthetic immune-stimulatory drugs such as agonists of the Toll-like receptors (TLR) 7/8 are potent activators of antigen-presenting cells (APCs), however, they also induce severe side effects due to leakage from the site of injection into systemic circulation. Here, we report on the design and synthesis of an amphiphilic polymer-prodrug conjugate of an imidazoquinoline TLR7/8 agonist that in aqueous medium forms vesicular structures of 200 nm. The conjugate contains an endosomal enzyme-responsive linker enabling degradation of the vesicles and release of the TLR7/8 agonist in native form after endocytosis, which results in high in vitro TLR agonist activity. In a mouse model, locally administered vesicles provoke significantly more potent and long-lasting immune stimulation in terms of interferon expression at the injection site and in draining lymphoid tissue compared to a nonamphiphilic control and the native TLR agonist. Moreover, the vesicles induce robust activation of dendritic cells in the draining lymph node in vivo.


Subject(s)
Imidazoles/pharmacology , Membrane Glycoproteins/agonists , Prodrugs/pharmacology , Quinolines/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , beta-Galactosidase/immunology , Animals , Imidazoles/chemistry , Imidazoles/metabolism , Immunity, Innate/drug effects , Immunity, Innate/immunology , Membrane Glycoproteins/immunology , Mice , Molecular Structure , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacology , Prodrugs/chemistry , Prodrugs/metabolism , Quinolines/chemistry , Quinolines/metabolism , Surface Properties , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism
12.
PLoS Pathog ; 14(8): e1007235, 2018 08.
Article in English | MEDLINE | ID: mdl-30075026

ABSTRACT

During Coxsackievirus B3 (CVB3) infection hepatitis is a potentially life threatening complication, particularly in newborns. Studies with type I interferon (IFN-I) receptor (IFNAR)-deficient mice revealed a key role of the IFN-I axis in the protection against CVB3 infection, whereas the source of IFN-I and cell types that have to be IFNAR triggered in order to promote survival are still unknown. We found that CVB3 infected IFN-ß reporter mice showed effective reporter induction, especially in hepatocytes and only to a minor extent in liver-resident macrophages. Accordingly, upon in vitro CVB3 infection of primary hepatocytes from murine or human origin abundant IFN-ß responses were induced. To identify sites of IFNAR-triggering we performed experiments with Mx reporter mice, which upon CVB3 infection showed massive luciferase induction in the liver. Immunohistological studies revealed that during CVB3 infection MX1 expression of hepatocytes was induced primarily by IFNAR-, and not by IFN-III receptor (IFNLR)-triggering. CVB3 infection studies with primary human hepatocytes, in which either the IFN-I or the IFN-III axis was inhibited, also indicated that primarily IFNAR-, and to a lesser extent IFNLR-triggering was needed for ISG induction. Interestingly, CVB3 infected mice with a hepatocyte-specific IFNAR ablation showed severe liver cell necrosis and ubiquitous viral dissemination that resulted in lethal disease, as similarly detected in classical IFNAR-/- mice. In conclusion, we found that during CVB3 infection hepatocytes are major IFN-I producers and that the liver is also the organ that shows strong IFNAR-triggering. Importantly, hepatocytes need to be IFNAR-triggered in order to prevent virus dissemination and to assure survival. These data are compatible with the hypothesis that during CVB3 infection hepatocytes serve as important IFN-I producers and sensors not only in the murine, but also in the human system.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human/immunology , Hepatocytes/metabolism , Interferon-beta/genetics , Liver/pathology , Receptor, Interferon alpha-beta/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops , Coxsackievirus Infections/complications , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Enterovirus B, Human/growth & development , Humans , Interferon-beta/metabolism , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/virology , Receptor, Interferon alpha-beta/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Vero Cells , Viral Load/genetics , Viral Load/immunology
13.
Am J Pathol ; 189(3): 521-539, 2019 03.
Article in English | MEDLINE | ID: mdl-30593822

ABSTRACT

Humanized mice developing functional human T cells endogenously and capable of recognizing cognate human leukocyte antigen-matched tumors are emerging as relevant models for studying human immuno-oncology in vivo. Herein, mice transplanted with human CD34+ stem cells and bearing endogenously developed human T cells for >15 weeks were infected with an oncogenic recombinant Epstein-Barr virus (EBV), encoding enhanced firefly luciferase and green fluorescent protein. EBV-firefly luciferase was detectable 1 week after infection by noninvasive optical imaging in the spleen, from where it spread rapidly and systemically. EBV infection resulted into a pronounced immunologic skewing regarding the expansion of CD8+ T cells in the blood outnumbering the CD4+ T and CD19+ B cells. Furthermore, within 10 weeks of infections, mice developing EBV-induced tumors had significantly higher absolute numbers of CD8+ T cells in lymphatic tissues than mice controlling tumor development. Tumor outgrowth was paralleled by an up-regulation of the programmed cell death receptor 1 on CD8+ and CD4+ T cells, indicative for T-cell dysfunction. Histopathological examinations and in situ hybridizations for EBV in tumors, spleen, liver, and kidney revealed foci of EBV-infected cells in perivascular regions in close association with programmed cell death receptor 1-positive infiltrating lymphocytes. The strong spatiotemporal correlation between tumor development and the T-cell dysfunctional status seen in this viral oncogenesis humanized model replicates observations obtained in the clinical setting.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Epstein-Barr Virus Infections/pathology , Humans , Lymphocyte Activation , Mice , Mice, Mutant Strains , Neoplasms/pathology , Neoplasms/virology
14.
PLoS Pathog ; 13(5): e1006382, 2017 May.
Article in English | MEDLINE | ID: mdl-28542326

ABSTRACT

The type I interferon (IFN) response is imperative for the establishment of the early antiviral immune response. Here we report the identification of the first type I IFN antagonist encoded by murine cytomegalovirus (MCMV) that shuts down signaling following pattern recognition receptor (PRR) sensing. Screening of an MCMV open reading frame (ORF) library identified M35 as a novel and strong negative modulator of IFNß promoter induction following activation of both RNA and DNA cytoplasmic PRR. Additionally, M35 inhibits the proinflammatory cytokine response downstream of Toll-like receptors (TLR). Using a series of luciferase-based reporters with specific transcription factor binding sites, we determined that M35 targets NF-κB-, but not IRF-mediated, transcription. Expression of M35 upon retroviral transduction of immortalized bone marrow-derived macrophages (iBMDM) led to reduced IFNß transcription and secretion upon activation of stimulator of IFN genes (STING)-dependent signaling. On the other hand, M35 does not antagonize interferon-stimulated gene (ISG) 56 promoter induction or ISG transcription upon exogenous stimulation of the type I IFN receptor (IFNAR). M35 is present in the viral particle and, upon MCMV infection of fibroblasts, is immediately shuttled to the nucleus where it exerts its immunomodulatory effects. Deletion of M35 from the MCMV genome and hence from the viral particle resulted in elevated type I IFN transcription and secretion in vitro and in vivo. In the absence of M35, lower viral titers are observed during acute infection of the host, and productive infection in the salivary glands was not detected. In conclusion, the M35 protein is released by MCMV immediately upon infection in order to deftly inhibit the antiviral type I IFN response by targeting NF-κB-mediated transcription. The identification of this novel viral protein reinforces the importance of timely countermeasures in the complex relationship between virus and host.


Subject(s)
Cytomegalovirus Infections/immunology , Interferon Type I/antagonists & inhibitors , Muromegalovirus/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction , Viral Proteins/metabolism , Animals , Cytomegalovirus Infections/virology , Interferon Type I/genetics , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Macrophages/immunology , Macrophages/virology , Mice , Muromegalovirus/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding , Receptors, Pattern Recognition/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Viral Proteins/genetics
15.
PLoS Pathog ; 13(11): e1006696, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29112952

ABSTRACT

Klebsiella pneumoniae is a significant cause of nosocomial pneumonia and an alarming pathogen owing to the recent isolation of multidrug resistant strains. Understanding of immune responses orchestrating K. pneumoniae clearance by the host is of utmost importance. Here we show that type I interferon (IFN) signaling protects against lung infection with K. pneumoniae by launching bacterial growth-controlling interactions between alveolar macrophages and natural killer (NK) cells. Type I IFNs are important but disparate and incompletely understood regulators of defense against bacterial infections. Type I IFN receptor 1 (Ifnar1)-deficient mice infected with K. pneumoniae failed to activate NK cell-derived IFN-γ production. IFN-γ was required for bactericidal action and the production of the NK cell response-amplifying IL-12 and CXCL10 by alveolar macrophages. Bacterial clearance and NK cell IFN-γ were rescued in Ifnar1-deficient hosts by Ifnar1-proficient NK cells. Consistently, type I IFN signaling in myeloid cells including alveolar macrophages, monocytes and neutrophils was dispensable for host defense and IFN-γ activation. The failure of Ifnar1-deficient hosts to initiate a defense-promoting crosstalk between alveolar macrophages and NK cell was circumvented by administration of exogenous IFN-γ which restored endogenous IFN-γ production and restricted bacterial growth. These data identify NK cell-intrinsic type I IFN signaling as essential driver of K. pneumoniae clearance, and reveal specific targets for future therapeutic exploitations.


Subject(s)
Interferon Type I/immunology , Killer Cells, Natural/immunology , Klebsiella Infections/immunology , Macrophages, Alveolar/immunology , Signal Transduction/immunology , Animals , Drug Resistance, Multiple/immunology , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/immunology , Respiratory Tract Infections/immunology
16.
J Immunol ; 198(4): 1595-1605, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28077601

ABSTRACT

Among innovative adjuvants conferring a Th1-shift, RNAdjuvant is a promising candidate. This adjuvant consists of a 547-nt uncapped noncoding ssRNA containing polyU repeats that is stabilized by a cationic carrier peptide. Whereas vaccination of mice with an influenza subunit vaccine induced moderate virus-specific IgG1, vaccination together with RNAdjuvant significantly enhanced this IgG1 and additionally promoted the formation of IgG2b/c, which is indicative of Th1 responses. Furthermore, such sera neutralized influenza virus, whereas this effect was not detected upon vaccination with the subunit vaccine alone. Similarly, upon vaccination with virus-like particles displaying vesicular stomatitis virus G protein, RNAdjuvant promoted the formation of virus-specific IgG2b/c and enhanced neutralizing IgG responses to an extent that mice were protected against lethal virus infection. RNAdjuvant induced dendritic cells to upregulate activation markers and produce IFN-I. Although these effects were strictly TLR7 dependent, RNAdjuvant-mediated augmentation of vaccine responses needed concurrent TLR and RIG-I-like helicase signaling. This was indicated by the absence of the adjuvant effect in vaccinated MyD88-/-Cardif-/- mice, which are devoid of TLR (with the exception of TLR3) and RIG-I-like helicase signaling, whereas in vaccinated MyD88-/- mice the adjuvant effect was reduced. Notably, i.m. RNAdjuvant injection induced local IFN-I responses and did not induce systemic effects, implying good tolerability and a favorable safety profile for RNAdjuvant.


Subject(s)
Adjuvants, Immunologic , Immunoglobulin G/blood , Influenza Vaccines/immunology , Membrane Glycoproteins/immunology , RNA, Untranslated/immunology , Toll-Like Receptor 7/immunology , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/adverse effects , Animals , Antibodies, Viral/blood , DEAD Box Protein 58/immunology , DEAD Box Protein 58/metabolism , Immunoglobulin G/immunology , Influenza Vaccines/administration & dosage , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Th1 Cells/immunology , Toll-Like Receptor 7/metabolism , Vaccination , Vaccines, Subunit/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/immunology
17.
Proc Natl Acad Sci U S A ; 113(29): 8098-103, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27382168

ABSTRACT

Agonists of Toll-like receptors (TLRs) are potent activators of the innate immune system and hold promise as vaccine adjuvant and for anticancer immunotherapy. Unfortunately, in soluble form they readily enter systemic circulation and cause systemic inflammatory toxicity. Here we demonstrate that by covalent ligation of a small-molecule imidazoquinoline-based TLR7/8 agonist to 50-nm-sized degradable polymeric nanogels the potency of the agonist to activate TLR7/8 in in vitro cultured dendritic cells is largely retained. Importantly, imidazoquinoline-ligated nanogels focused the in vivo immune activation on the draining lymph nodes while dramatically reducing systemic inflammation. Mechanistic studies revealed a prevalent passive diffusion of the nanogels to the draining lymph node. Moreover, immunization studies in mice have shown that relative to soluble TLR7/8 agonist, imidazoquinoline-ligated nanogels induce superior antibody and T-cell responses against a tuberculosis antigen. This approach opens possibilities to enhance the therapeutic benefit of small-molecule TLR agonist for a variety of applications.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Imidazoles/administration & dosage , Nanostructures/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Antigens, Bacterial/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gels , Hydrogen-Ion Concentration , Imidazoles/chemistry , Imidazoles/pharmacology , Immunization , Lymph Nodes/drug effects , Lymph Nodes/immunology , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Nanostructures/chemistry , T-Lymphocytes/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists
18.
Angew Chem Int Ed Engl ; 58(43): 15390-15395, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31397948

ABSTRACT

Uncontrolled systemic inflammatory immune triggering has hampered the clinical translation of several classes of small-molecule immunomodulators, such as imidazoquinoline TLR7/8 agonists for vaccine design and cancer immunotherapy. By taking advantage of the inherent serum-protein-binding property of lipid motifs and their tendency to accumulate in lymphoid tissue, we designed amphiphilic lipid-polymer conjugates that suppress systemic inflammation but provoke potent lymph-node immune activation. This work provides a rational basis for the design of lipid-polymer amphiphiles for optimized lymphoid targeting.


Subject(s)
Immunity, Innate , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , Cholesterol/chemistry , Imidazoles/chemistry , Immunity, Innate/drug effects , Immunologic Factors/chemistry , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Lipids/chemistry , Lymph Nodes/drug effects , Lymph Nodes/immunology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Polymers/chemistry , Quinolines/chemistry , Quinolines/pharmacology , RAW 264.7 Cells , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism
19.
J Am Chem Soc ; 140(43): 14300-14307, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30277761

ABSTRACT

Small molecule immuno-modulators such as agonists of Toll-like receptors (TLRs) are attractive compounds to stimulate innate immune cells toward potent antiviral and antitumor responses. However, small molecules rapidly enter the systemic circulation and cause "wasted inflammation". Hence, synthetic strategies to confine their radius of action to lymphoid tissue are of great relevance, to both enhance their efficacy and concomitantly limit toxicity. Here, we demonstrate that covalent conjugation of a small molecule TLR7/8 agonist immunomodulatory to a micelle-forming amphiphilic block copolymer greatly alters the pharmacokinetic profile, resulting in highly efficient lymphatic delivery. Moreover, we designed amphiphilic block copolymers in such a way to form thermodynamically stable micelles through π-π stacking between aromatic moieties, and we engineered the block copolymers to undergo an irreversible amphiphilic to hydrophilic transition in response to the acidic endosomal pH.


Subject(s)
Lymph Nodes/drug effects , Polymers/pharmacology , Surface-Active Agents/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Humans , Hydrogen-Ion Concentration , Lymph Nodes/immunology , Micelles , Models, Molecular , Molecular Structure , Polymers/chemistry , Surface-Active Agents/chemistry , Thermodynamics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology
20.
J Hepatol ; 68(4): 682-690, 2018 04.
Article in English | MEDLINE | ID: mdl-29274730

ABSTRACT

BACKGROUND & AIM: Virus-induced fulminant hepatitis is a major cause of acute liver failure. During acute viral hepatitis the impact of type I interferon (IFN-I) on myeloid cells, including liver-resident Kupffer cells (KC), is only partially understood. Herein, we dissected the impact of locally induced IFN-I responses on myeloid cell function and hepatocytes during acute liver inflammation. METHODS: Two different DNA-encoded viruses, vaccinia virus (VACV) and murine cytomegalovirus (MCMV), were studied. In vivo imaging was applied to visualize local IFN-ß induction and IFN-I receptor (IFNAR) triggering in VACV-infected reporter mice. Furthermore, mice with a cell type-selective IFNAR ablation were analyzed to dissect the role of IFNAR signaling in myeloid cells and hepatocytes. Experiments with Cx3cr1+/gfp mice revealed the origin of reconstituted KC. Finally, mixed bone marrow chimeric mice were studied to specifically analyze the effect of IFNAR triggering on liver infiltrating monocytes. RESULTS: VACV infection induced local IFN-ß responses, which lead to IFNAR signaling primarily within the liver. IFNAR triggering was needed to control the infection and prevent fulminant hepatitis. The severity of liver inflammation was independent of IFNAR triggering of hepatocytes, whereas IFNAR triggering of myeloid cells protected from excessive inflammation. Upon VACV or MCMV infection KC disappeared, whereas infiltrating monocytes differentiated to KC afterwards. During IFNAR triggering such replenished monocyte-derived KC comprised more IFNAR-deficient than -competent cells in mixed bone marrow chimeric mice, whereas after the decline of IFNAR triggering both subsets showed an even distribution. CONCLUSION: Upon VACV infection IFNAR triggering of myeloid cells, but not of hepatocytes, critically modulates acute viral hepatitis. During infection with DNA-encoded viruses IFNAR triggering of liver-infiltrating blood monocytes delays the development of monocyte-derived KC, pointing towards new therapeutic strategies for acute viral hepatitis. LAY SUMMARY: Viral infection can cause fulminant hepatitis, which in turn is a major cause of acute liver failure. Herein, we aimed to study the role of type 1 interferon responses in acute viral hepatitis. We identified that during infection with DNA-encoded viruses, type 1 interferon receptor triggering of blood monocytes delays the development of monocyte-derived Kupffer cells. This points to new therapeutic strategies for acute viral hepatitis.


Subject(s)
Hepatitis, Viral, Animal/physiopathology , Kupffer Cells/physiology , Receptor, Interferon alpha-beta/physiology , Signal Transduction/physiology , Acute Disease , Animals , Hepatitis, Viral, Animal/etiology , Mice , Mice, Inbred C57BL , Vaccinia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL