Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 925
Filter
2.
J Immunol ; 212(11): 1680-1692, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38607278

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are strongly implicated as a major source of IFN-I in systemic lupus erythematosus (SLE), triggered through TLR-mediated recognition of nucleic acids released from dying cells. However, relatively little is known about how TLR signaling and IFN-I production are regulated in pDCs. In this article, we describe a role for integrin αvß3 in regulating TLR responses and IFN-I production by pDCs in mouse models. We show that αv and ß3-knockout pDCs produce more IFN-I and inflammatory cytokines than controls when stimulated through TLR7 and TLR9 in vitro and in vivo. Increased cytokine production was associated with delayed acidification of endosomes containing TLR ligands, reduced LC3 conjugation, and increased TLR signaling. This dysregulated TLR signaling results in activation of B cells and promotes germinal center (GC) B cell and plasma cell expansion. Furthermore, in a mouse model of TLR7-driven lupus-like disease, deletion of αvß3 from pDCs causes accelerated autoantibody production and pathology. We therefore identify a pDC-intrinsic role for αvß3 in regulating TLR signaling and preventing activation of autoreactive B cells. Because αvß3 serves as a receptor for apoptotic cells and cell debris, we hypothesize that this regulatory mechanism provides important contextual cues to pDCs and functions to limit responses to self-derived nucleic acids.


Subject(s)
Autoimmunity , Dendritic Cells , Integrin alphaVbeta3 , Lupus Erythematosus, Systemic , Mice, Knockout , Signal Transduction , Toll-Like Receptor 7 , Animals , Mice , Dendritic Cells/immunology , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Autoimmunity/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Lupus Erythematosus, Systemic/immunology , Signal Transduction/immunology , Mice, Inbred C57BL , Cytokines/metabolism , Cytokines/immunology , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , B-Lymphocytes/immunology , Autoantibodies/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Lymphocyte Activation/immunology , Disease Models, Animal
4.
Cell Immunol ; 399-400: 104823, 2024.
Article in English | MEDLINE | ID: mdl-38520831

ABSTRACT

AAV-mediated gene transfer is a promising platform still plagued by potential host-derived, antagonistic immune responses to therapeutic components. CpG-mediated TLR9 stimulation activates innate immune cells and leads to cognate T cell activation and suppression of transgene expression. Here, we demonstrate that CpG depletion increased expression of an antibody transgene product by 2-3-fold as early as 24 h post-vector administration in mice. No significant differences were noted in anti-transgene product/ anti-AAV capsid antibody production or cytotoxic gene induction. Instead, CpG depletion significantly reduced the presence of a pDC-like myeloid cell population, which was able to directly bind the antibody transgene product via Fc-FcγR interactions. Thus, we extend the mechanisms of TLR9-mediated antagonism of transgene expression in AAV gene therapy to include the actions of a previously unreported pDC-like cell population.


Subject(s)
Dendritic Cells , Dependovirus , Genetic Therapy , Genetic Vectors , Mice, Inbred C57BL , Toll-Like Receptor 9 , Transgenes , Animals , Dendritic Cells/immunology , Dependovirus/genetics , Mice , Genetic Therapy/methods , Toll-Like Receptor 9/immunology , CpG Islands/genetics , CpG Islands/immunology , Receptors, IgG/immunology , Receptors, IgG/genetics , Receptors, IgG/metabolism
5.
Nature ; 628(8006): 145-153, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538785

ABSTRACT

As hippocampal neurons respond to diverse types of information1, a subset assembles into microcircuits representing a memory2. Those neurons typically undergo energy-intensive molecular adaptations, occasionally resulting in transient DNA damage3-5. Here we found discrete clusters of excitatory hippocampal CA1 neurons with persistent double-stranded DNA (dsDNA) breaks, nuclear envelope ruptures and perinuclear release of histone and dsDNA fragments hours after learning. Following these early events, some neurons acquired an inflammatory phenotype involving activation of TLR9 signalling and accumulation of centrosomal DNA damage repair complexes6. Neuron-specific knockdown of Tlr9 impaired memory while blunting contextual fear conditioning-induced changes of gene expression in specific clusters of excitatory CA1 neurons. Notably, TLR9 had an essential role in centrosome function, including DNA damage repair, ciliogenesis and build-up of perineuronal nets. We demonstrate a novel cascade of learning-induced molecular events in discrete neuronal clusters undergoing dsDNA damage and TLR9-mediated repair, resulting in their recruitment to memory circuits. With compromised TLR9 function, this fundamental memory mechanism becomes a gateway to genomic instability and cognitive impairments implicated in accelerated senescence, psychiatric disorders and neurodegenerative disorders. Maintaining the integrity of TLR9 inflammatory signalling thus emerges as a promising preventive strategy for neurocognitive deficits.


Subject(s)
CA1 Region, Hippocampal , DNA Breaks, Double-Stranded , DNA Repair , Inflammation , Memory , Toll-Like Receptor 9 , Animals , Female , Male , Mice , Aging/genetics , Aging/pathology , CA1 Region, Hippocampal/physiology , Centrosome/metabolism , Cognitive Dysfunction/genetics , Conditioning, Classical , Extracellular Matrix/metabolism , Fear , Genomic Instability/genetics , Histones/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Memory/physiology , Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Neuroinflammatory Diseases/genetics , Neurons/metabolism , Neurons/pathology , Nuclear Envelope/pathology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
6.
Nat Med ; 29(10): 2547-2558, 2023 10.
Article in English | MEDLINE | ID: mdl-37696935

ABSTRACT

Inducing antiretroviral therapy (ART)-free virological control is a critical step toward a human immunodeficiency virus type 1 (HIV-1) cure. In this phase 2a, placebo-controlled, double-blinded trial, 43 people (85% males) with HIV-1 on ART were randomized to (1) placebo/placebo, (2) lefitolimod (TLR9 agonist)/placebo, (3) placebo/broadly neutralizing anti-HIV-1 antibodies (bNAbs) or (4) lefitolimod/bNAb. ART interruption (ATI) started at week 3. Lefitolimod was administered once weekly for the first 8 weeks, and bNAbs were administered twice, 1 d before and 3 weeks after ATI. The primary endpoint was time to loss of virologic control after ATI. The median delay in time to loss of virologic control compared to the placebo/placebo group was 0.5 weeks (P = 0.49), 12.5 weeks (P = 0.003) and 9.5 weeks (P = 0.004) in the lefitolimod/placebo, placebo/bNAb and lefitolimod/bNAb groups, respectively. Among secondary endpoints, viral doubling time was slower for bNAb groups compared to non-bNAb groups, and the interventions were overall safe. We observed no added benefit of lefitolimod. Despite subtherapeutic plasma bNAb levels, 36% (4/11) in the placebo/bNAb group compared to 0% (0/10) in the placebo/placebo group maintained virologic control after the 25-week ATI. Although immunotherapy with lefitolimod did not lead to ART-free HIV-1 control, bNAbs may be important components in future HIV-1 curative strategies. ClinicalTrials.gov identifier: NCT03837756 .


Subject(s)
HIV Infections , HIV-1 , Toll-Like Receptor 9 , Female , Humans , Male , Adjuvants, Immunologic , Antibodies, Neutralizing , Broadly Neutralizing Antibodies/therapeutic use , HIV Antibodies/therapeutic use , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/immunology
7.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35878041

ABSTRACT

Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-ß (IFN-ß) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli-evoked lung injury, and suppresses production of IFN-ß and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-ß delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-ß accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-ß-mediated resolution. These findings point to a pivotal role of IFN-ß in orchestrating timely resolution of neutrophil and TLR9 activation-driven airway inflammation and uncover an IFN-ß-initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.


Subject(s)
Interferon-beta , Lipoxins , Respiratory Distress Syndrome , Animals , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli , Escherichia coli Infections/immunology , Humans , Inflammation/drug therapy , Interferon-beta/immunology , Interferon-beta/pharmacology , Lipoxins/pharmacology , Mice , Receptors, Formyl Peptide/antagonists & inhibitors , Respiratory Distress Syndrome/drug therapy , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transcriptional Activation/drug effects
8.
Comput Math Methods Med ; 2022: 8660752, 2022.
Article in English | MEDLINE | ID: mdl-35132333

ABSTRACT

Swine pneumonia commonly known as swine pasteurellosis is an infectious disease of swine caused by Pasteurella multocida infection. It has been reported that Toll-like receptors (TLRs) play a vital role in swine pneumonia progression. However, the underlying mechanism has not been elucidated. This research was aimed at investigating the molecular mechanism by which TLR9 regulates swine pneumonia progression. Our findings illustrated that the HD-13 strain of Pasteurella multocida D (HD-13) accelerated TLR9 expression in porcine alveolar macrophage 3D4/21 cells; HD-13 activated the inflammatory response via accelerating TLR9 expression. Mechanistically, HD-13 activated mitogen-activated protein kinase (MAPK) and nuclear factor kB (NF-κB) signals. In conclusion, HD-13 may activate MAPK and NF-κB pathways via accelerating TLR9 expression, thereby accelerating the inflammatory response in the progression of swine pneumonia. TLR9 may serve as a novel therapeutic target for swine pneumonia. Our research may provide a theoretical basis for the prevention and treatment of swine pneumonia.


Subject(s)
Pasteurella Infections/veterinary , Pasteurella multocida/pathogenicity , Pneumonia/veterinary , Swine Diseases/immunology , Swine Diseases/microbiology , Toll-Like Receptor 9/immunology , Animals , Cells, Cultured , Computational Biology , Cytokines/genetics , Cytokines/immunology , Disease Progression , MAP Kinase Signaling System/immunology , NF-kappa B/immunology , Pasteurella Infections/immunology , Pasteurella Infections/microbiology , Pasteurella multocida/classification , Pasteurella multocida/immunology , Pneumonia/immunology , Pneumonia/microbiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/immunology , Sus scrofa , Swine , Swine Diseases/genetics , Toll-Like Receptor 9/genetics , Up-Regulation
9.
Cell Prolif ; 55(3): e13192, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35084069

ABSTRACT

OBJECTIVES: Synthetic oligodeoxynucleotides (ODNs) that contain unmethylated cytosine-phosphate-guanine (CpG) motifs serve as immune adjuvants in disease treatment. However, the poor cell permeability and safety concerns limit their medical applications, and biocompatible strategies for efficient delivery of functional CpG ODNs are highly desirable. MATERIALS AND METHODS: Self-assembled, cell membrane-coated CpG nanoparticles (NP) are prepared, and their physicochemical properties are characterized. The uncoated and membrane-coated CpG NP are compared for their biocompatibility, cellular uptake kinetics, endocytic pathways, subcellular localization, and immunostimulatory activities in macrophages and microglia. RESULTS: Macrophage- or microglia-derived cell membrane camouflaging alters the endocytic pathways of CpG NP, promotes their targeted delivery to the cells with homologous membrane, ensures their endosomal localization, and enhances their immunomodulatory effects. CONCLUSIONS: We design a type of biomimetic NP consisting of self-assembled CpG NP core and cell membrane shell, and demonstrate its advantages in the modulation of peripheral and central immune cells. Our study provides a new strategy for the application of CpG ODNs.


Subject(s)
Immunomodulation/immunology , Macrophages/immunology , Nanoparticles/metabolism , Oligodeoxyribonucleotides/immunology , Animals , Cytosine/metabolism , Macrophages/metabolism , Oligodeoxyribonucleotides/metabolism , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
10.
Eur J Immunol ; 52(2): 270-284, 2022 02.
Article in English | MEDLINE | ID: mdl-34773640

ABSTRACT

Recognition of pathogen-associated molecular patterns (PAMPs) through Toll-like receptors (TLRs) plays a pivotal role in first-line pathogen defense. TLRs are also likely triggered during a Plasmodium infection in vivo by parasite-derived components. However, the contribution of innate responses to liver infection and to the subsequent clinical outcome of a blood infection is not well understood. To assess the potential effects of enhanced TLR-signalling on Plasmodium infection, we systematically examined the effect of agonist-primed immune responses to sporozoite inoculation in the P. berghei/C57Bl/6 murine malaria model. We could identify distinct stage-specific effects on the course of infection after stimulation with two out of four TLR-ligands tested. Priming with a TLR9 agonist induced killing of pre-erythrocytic stages in the liver that depended on macrophages and the expression of inducible nitric oxide synthase (iNOS). These factors have previously not been recognized as antigen-independent effector mechanisms against Plasmodium liver stages. Priming with TLR4 and -9 agonists also translated into blood stage-specific protection against experimental cerebral malaria (ECM). These insights are relevant to the activation of TLR signalling pathways by adjuvant systems of antimalaria vaccine strategies. The protective role of TLR4-activation against ECM might also explain some unexpected clinical effects observed with pre-erythrocytic vaccine approaches.


Subject(s)
Liver Diseases , Liver , Macrophage Activation , Macrophages/immunology , Malaria , Plasmodium berghei/immunology , Signal Transduction , Toll-Like Receptor 9/immunology , Animals , Female , Liver/immunology , Liver/parasitology , Liver Diseases/genetics , Liver Diseases/immunology , Liver Diseases/parasitology , Malaria/genetics , Malaria/immunology , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 9/genetics
11.
Virology ; 566: 98-105, 2022 01.
Article in English | MEDLINE | ID: mdl-34896902

ABSTRACT

The innate and acquired immune response induced by a commercial inactivated vaccine against Bovine Herpesvirus-1 (BoHV-1) and protection conferred against the virus were analyzed in cattle. Vaccination induced high levels of BoHV-1 antibodies at 30, 60, and 90 days post-vaccination (dpv). IgG1 and IgG2 isotypes were detected at 90 dpv, as well as virus-neutralizing antibodies. An increase of anti-BoHV-1 IgG1 in nasal swabs was detected 6 days post-challenge in vaccinated animals. After viral challenge, lower virus excretion and lower clinical score were observed in vaccinated as compared to unvaccinated animals, as well as BoHV-1-specific proliferation of lymphocytes and production of IFNγ, TNFα, and IL-4. Downregulation of the expression of endosome Toll-like receptors 8-9 was detected after booster vaccination. This is the first thorough study of the immunity generated by a commercial vaccine against BoHV-1 in cattle.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Herpesvirus 1, Bovine/immunology , Herpesvirus Vaccines/administration & dosage , Immunoglobulin G/biosynthesis , Infectious Bovine Rhinotracheitis/prevention & control , Toll-Like Receptor 8/immunology , Toll-Like Receptor 9/immunology , Adaptive Immunity/drug effects , Animals , Antibodies, Viral , Cattle , Cell Proliferation , Endosomes/immunology , Endosomes/metabolism , Gene Expression , Herpesvirus 1, Bovine/pathogenicity , Immunity, Innate/drug effects , Immunization, Secondary/methods , Infectious Bovine Rhinotracheitis/genetics , Infectious Bovine Rhinotracheitis/immunology , Infectious Bovine Rhinotracheitis/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/immunology , Lymphocytes/virology , Male , Nasal Cavity/immunology , Nasal Cavity/virology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/genetics , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccination/methods , Vaccines, Inactivated
13.
Front Immunol ; 12: 698420, 2021.
Article in English | MEDLINE | ID: mdl-34497606

ABSTRACT

Siglec-H is a DAP12-associated receptor on plasmacytoid dendritic cells (pDCs) and microglia. Siglec-H inhibits TLR9-induced IFN-α production by pDCs. Previously, it was found that Siglec-H-deficient mice develop a lupus-like severe autoimmune disease after persistent murine cytomegalovirus (mCMV) infection. This was due to enhanced type I interferon responses, including IFN-α. Here we examined, whether other virus infections can also induce autoimmunity in Siglec-H-deficient mice. To this end we infected Siglec-H-deficient mice with influenza virus or with Lymphocytic Choriomeningitis virus (LCMV) clone 13. With both types of viruses we did not observe induction of autoimmune disease in Siglec-H-deficient mice. This can be explained by the fact that both types of viruses are ssRNA viruses that engage TLR7, rather than TLR9. Also, Influenza causes an acute infection that is rapidly cleared and the chronicity of LCMV clone 13 may not be sufficient and may rather suppress pDC functions. Siglec-H inhibited exclusively TLR-9 driven type I interferon responses, but did not affect type II or type III interferon production by pDCs. Siglec-H-deficient pDCs showed impaired Hck expression, which is a Src-family kinase expressed in myeloid cells, and downmodulation of the chemokine receptor CCR9, that has important functions for pDCs. Accordingly, Siglec-H-deficient pDCs showed impaired migration towards the CCR9 ligand CCL25. Furthermore, autoimmune-related genes such as Klk1 and DNase1l3 are downregulated in Siglec-H-deficient pDCs as well. From these findings we conclude that Siglec-H controls TLR-9-dependent, but not TLR-7 dependent inflammatory responses after virus infections and regulates chemokine responsiveness of pDCs.


Subject(s)
Arenaviridae Infections/immunology , Autoimmune Diseases/immunology , Interferon Type I/immunology , Lectins/immunology , Orthomyxoviridae Infections/immunology , Receptors, Cell Surface/immunology , Animals , Autoimmune Diseases/virology , Autoimmunity/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Influenza A Virus, H3N2 Subtype , Lectins/deficiency , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/deficiency , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
14.
Aging (Albany NY) ; 13(18): 21962-21974, 2021 09 19.
Article in English | MEDLINE | ID: mdl-34537761

ABSTRACT

Dulaglutide, a glucagon-like peptide-1 receptor (GLP-1R) agonist, is widely used to treat diabetes. However, its effects on muscle wasting due to aging are poorly understood. In the current study, we investigated the therapeutic potential and underlying mechanism of dulaglutide in muscle wasting in aged mice. Dulaglutide improved muscle mass and strength in aged mice. Histological analysis revealed that the cross-sectional area of the tibialis anterior (TA) in the dulaglutide-treated group was thicker than that in the vehicle group. Moreover, dulaglutide increased the shift toward middle and large-sized fibers in both young and aged mice compared to the vehicle. Dulaglutide increased myofiber type I and type IIa in young (18.5% and 8.2%) and aged (1.8% and 19.7%) mice, respectively, compared to the vehicle group. Peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, decreased but increased by dulaglutide in aged mice. The expression of atrophic factors such as myostatin, atrogin-1, and muscle RING-finger protein-1 was decreased in aged mice, whereas that of the myogenic factor, MyoD, was increased in both young and aged mice following dulaglutide treatment. In aged mice, optic atrophy-1 (OPA-1) protein was decreased, whereas Toll-like receptor-9 (TLR-9) and its targeting inflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor-α [TNF-α]) were elevated in the TA and quadriceps (QD) muscles. In contrast, dulaglutide administration reversed this expression pattern, thereby significantly attenuating the expression of inflammatory cytokines in aged mice. These data suggest that dulaglutide may exert beneficial effects in the treatment of muscle wasting due to aging.


Subject(s)
Aging/metabolism , Glucagon-Like Peptides/analogs & derivatives , Immunoglobulin Fc Fragments/administration & dosage , Muscle, Skeletal/physiopathology , Recombinant Fusion Proteins/administration & dosage , Sarcopenia/drug therapy , Sarcopenia/immunology , Toll-Like Receptor 9/immunology , Aging/drug effects , Aging/genetics , Animals , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Glucagon-Like Peptides/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Muscle Proteins/genetics , Muscle Proteins/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/immunology , Sarcopenia/etiology , Sarcopenia/genetics , Signal Transduction/drug effects , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
BMC Genom Data ; 22(1): 37, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34544361

ABSTRACT

BACKGROUND: Transcription factors (TFs) control gene expression by direct binding to regulatory regions of target genes but also by impacting chromatin landscapes and modulating DNA accessibility for other TFs. In recent years several TFs have been defined that control cell fate decisions and effector functions in the immune system. Plasmacytoid dendritic cells (pDCs) are an immune cell type with the unique capacity to produce high amounts of type I interferons quickly in response to contact with viral components. Hereby, this cell type is involved in anti-infectious immune responses but also in the development of inflammatory and autoimmune diseases. To date, the global TF reservoir in pDCs early after activation remains to be fully characterized. RESULTS: To fill this gap, we have performed a comprehensive analysis in naïve versus TLR9-activated murine pDCs in a time course study covering early timepoints after stimulation (2 h, 6 h, 12 h) integrating gene expression (RNA-Seq) and chromatin landscape (ATAC-Seq) studies. To unravel the biological processes underlying the changes in TF expression on a global scale gene ontology (GO) analyses were performed. We found that 70% of all genes annotated as TFs in the mouse genome (1014 out of 1636) are expressed in pDCs for at least one stimulation time point and are covering a wide range of TF classes defined by their specific DNA binding mechanisms. GO analysis revealed involvement of TLR9-induced TFs in epigenetic modulation, NFκB and JAK-STAT signaling, and protein production in the endoplasmic reticulum. pDC activation predominantly "turned on" the chromatin regions associated with TF genes. Our in silico analyses pointed at the AP-1 family of TFs as less noticed but possibly important players in these cells after activation. AP-1 family members exhibit (1) increased gene expression, (2) enhanced chromatin accessibility in their promoter region, and (3) a TF DNA binding motif that is globally enriched in genomic regions that were found more accessible in pDCs after TLR9 activation. CONCLUSIONS: In this study we define the complete set of TLR9-regulated TFs in pDCs. Further, this study identifies the AP-1 family of TFs as potentially important but so far less well characterized regulators of pDC function.


Subject(s)
Chromatin/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Transcription Factors/metabolism , Animals , Dendritic Cells/cytology , Female , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 9/immunology , Transcription Factor AP-1/metabolism
16.
Virulence ; 12(1): 1997-2012, 2021 12.
Article in English | MEDLINE | ID: mdl-34339354

ABSTRACT

Little is known about how Talaromyces marneffei, a thermally dimorphic fungus that causes substantial morbidity and mortality in Southeast Asia, evades the human immune system. Polarization of macrophages into fungal-inhibiting M1-like and fungal-promoting M2-like types has been shown to play an important role in the innate immune response against fungal pathogens. This mechanism has not been defined for T. marneffei. Here, we demonstrated that T. marneffei promotes its survival in human macrophages by inducing them toward M2-like polarization. Our investigations of the mechanism revealed that T. marneffei infection led to SOCS3 protein degradation by inducing tyrosine phosphorylation, thereby relieving the inhibitory effect of SOCS3 on p-STAT6, a key factor for M2-like polarization. Our SOCS3-overexpression experiments showed that SOCS3 is a positive regulator of M1-like polarization and plays an important role in limiting M2-like polarization. Furthermore, we found that inhibition of the TLR9 pathway partially blocked T. marneffei-induced M2-like polarization and significantly enhanced the killing activity of macrophages against T. marneffei. Collectively, these results reveal a novel mechanism by which T. marneffei evades the immune response of human macrophages.


Subject(s)
Immune Evasion , Macrophages/microbiology , Suppressor of Cytokine Signaling 3 Protein/immunology , Talaromyces , Toll-Like Receptor 9/immunology , Cell Polarity , Humans , Immunity, Innate , Macrophages/immunology , Mycoses/immunology , Suppressor of Cytokine Signaling 3 Protein/genetics , Talaromyces/genetics , Talaromyces/pathogenicity
17.
Adv Sci (Weinh) ; 8(19): e2101031, 2021 10.
Article in English | MEDLINE | ID: mdl-34365741

ABSTRACT

IRF8 is a key regulator of innate immunity receptor signaling and plays diverse functions in the development of hematopoietic cells. The effects of IRF8 on hematopoietic stem cells (HSCs) are still unknown. Here, it is demonstrated that IRF8 deficiency results in a decreased number of long-term HSCs (LT-HSCs) in mice. However, the repopulation capacity of individual HSCs is significantly increased. Transcriptomic analysis shows that IFN-γ and IFN-α signaling is downregulated in IRF8-deficient HSCs, while their response to proinflammatory cytokines is unchanged ex vivo. Further tests show that Irf8-/- HSCs can not respond to CpG, an agonist of Toll-like receptor 9 (TLR9) in mice, while long-term CpG stimulation increases wild-type HSC abundance and decreases their bone marrow colony-forming capacity. Mechanistically, as the primary producer of proinflammatory cytokines in response to CpG stimulation, dendritic cells has a blocked TLR9 signaling due to developmental defect in Irf8-/- mice. Macrophages remain functionally intact but severely reduce in Irf8-/- mice. In NK cells, IRF8 directly regulates the expression of Tlr9 and its deficiency leads to no increased IFNγ production upon CpG stimulation. These results indicate that IRF8 regulates HSCs, at least in part, through controlling TLR9 signaling in diverse innate immune cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Immunity, Innate/immunology , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Animals , Gene Expression Profiling/methods , Hematopoietic Stem Cells/immunology , Immunity, Innate/genetics , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Models, Animal , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 9/genetics
18.
Front Immunol ; 12: 672143, 2021.
Article in English | MEDLINE | ID: mdl-34093572

ABSTRACT

Mice reconstituted with a human immune system (humanized mice) provide a robust model to study human immunology, vaccinology, and human infectious diseases. However, the development and function of B cells in humanized mice is impaired. B cells from humanized mice are immature and are impaired in IgM to IgG isotype switch in response to infection or vaccination. In the present study we report that Toll-like receptor 9 (TLR9) agonist CpG-B combined with CD40-targeting vaccination triggered human B cell immunoglobin class-switch from IgM+ to IgG+ B cells in humanized mice. Human B cells from mice vaccinated with CpG-B as adjuvant were more mature in phenotype and produced significant levels of both total IgG and antigen-specific IgG. We found that CpG-B treatment activated human pDCs (plasmacytoid dendritic cells) in vivo to induce interferon-alpha (IFN-α)expression in humanized mice. Pre-depletion of human pDC in vivo abrogated the adjuvant effect of CpG-B. Our results indicate that TLR9 and CD40-targeting vaccination triggers human B cell maturation and immunoglobulin class-switch in a pDC-dependent manner in humanized mice. The findings also shed light on induction of human IgG antibodies in humanized mouse models.


Subject(s)
CD40 Antigens/immunology , Dendritic Cells/immunology , Toll-Like Receptor 9/immunology , Vaccination/methods , Adjuvants, Immunologic/pharmacology , Animals , B-Lymphocytes , Dendritic Cells/drug effects , HIV-1 , Humans , Immunoglobulin Class Switching/drug effects , Immunoglobulin Class Switching/immunology , Immunoglobulin G , Mice , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology
19.
Mucosal Immunol ; 14(5): 1100-1112, 2021 09.
Article in English | MEDLINE | ID: mdl-34103660

ABSTRACT

Major histocompatibility complex class II (MHCII) is dynamically expressed on intestinal epithelial cells (IECs) throughout the intestine, but its regulation remains poorly understood. We observed that spontaneous upregulation of IEC MHCII in locally bred Rag1-/- mice correlated with serum Interleukin (IL)-18, was transferrable via co-housing to commercially bred immunodeficient mice and could be inhibited by both IL-12 and IL-18 blockade. Overproduction of intestinal IL-18 due to an activating Nlrc4 mutation upregulated IEC MHCII via classical inflammasome machinery independently of immunodeficiency or dysbiosis. Immunodeficient dysbiosis increased Il-18 transcription, which synergized with NLRC4 inflammasome activity to drive elevations in serum IL-18. This IL-18-MHCII axis was confirmed in several other models of intestinal and systemic inflammation. Elevated IL-18 reliably preceded MHCII upregulation, suggesting an indirect effect on IECs, and mice with IL-18 overproduction showed activation or expansion of type 1 lymphocytes. Interferon gamma (IFNg) was uniquely able to upregulate IEC MHCII in enteroid cultures and was required for MHCII upregulation in several in vivo systems. Thus, we have linked intestinal dysbiosis, systemic inflammation, and inflammasome activity to IEC MHCII upregulation via an intestinal IL-18-IFNg axis. Understanding this process may be crucial for determining the contribution of IEC MHCII to intestinal homeostasis, host defense, and tolerance.


Subject(s)
Gastrointestinal Microbiome/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Interferon-gamma/metabolism , Interleukin-18/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Biomarkers , Cytokines , Dysbiosis/immunology , Enterocytes/metabolism , Gene Expression , Homeodomain Proteins/genetics , Immunity, Mucosal , Immunophenotyping , Inflammasomes/metabolism , Mice , Mice, Knockout , Models, Biological , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
20.
Macromol Biosci ; 21(6): e2100083, 2021 06.
Article in English | MEDLINE | ID: mdl-33896107

ABSTRACT

Cancer immunotherapy has risen as a promising method in clinical practice for cancer treatment and DNA-based immune intervention materials, along with DNA nanotechnology, have obtained increasing importance in this field. In this review, various immunostimulating DNA materials are introduced and the mechanisms via which they exerted an immune effect are explained. Then, representative examples in which DNA is used as the leading component for anticancer applications through immune stimulation are provided and their efficacy is evaluated. Finally, the challenges for those materials in clinical applications are discussed and suggestions for possible further research directions are also put forward.


Subject(s)
Aptamers, Nucleotide/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Toll-Like Receptor 9/antagonists & inhibitors , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Dinucleotide Repeats/genetics , Dinucleotide Repeats/immunology , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/chemical synthesis , Immune Checkpoint Inhibitors/metabolism , Immunization/methods , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...