Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 851
Filter
1.
Nat Immunol ; 18(6): 622-632, 2017 06.
Article in English | MEDLINE | ID: mdl-28459433

ABSTRACT

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Immunity, Innate/immunology , Monocytes/immunology , Neonatal Sepsis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Calgranulin A/drug effects , Calgranulin B/drug effects , Epigenesis, Genetic , Fetal Blood , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Immunoblotting , Infant, Newborn , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Monocytes/drug effects , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neonatal Sepsis/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/immunology
2.
Immunity ; 52(5): 782-793.e5, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32272082

ABSTRACT

Splenic red pulp macrophages (RPMs) contribute to erythrocyte homeostasis and are required for iron recycling. Heme induces the expression of SPIC transcription factor in monocyte-derived macrophages and promotes their differentiation into RPM precursors, pre-RPMs. However, the requirements for differentiation into mature RPMs remain unknown. Here, we have demonstrated that interleukin (IL)-33 associated with erythrocytes and co-cooperated with heme to promote the generation of mature RPMs through activation of the MyD88 adaptor protein and ERK1/2 kinases downstream of the IL-33 receptor, IL1RL1. IL-33- and IL1RL1-deficient mice showed defective iron recycling and increased splenic iron deposition. Gene expression and chromatin accessibility studies revealed a role for GATA transcription factors downstream of IL-33 signaling during the development of pre-RPMs that retained full potential to differentiate into RPMs. Thus, IL-33 instructs the development of RPMs as a response to physiological erythrocyte damage with important implications to iron recycling and iron homeostasis.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/immunology , Iron/metabolism , Macrophages/immunology , Signal Transduction/immunology , Spleen/metabolism , Animals , Erythrocytes/immunology , Erythrocytes/metabolism , Heme/immunology , Heme/metabolism , Homeostasis/immunology , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Macrophages/metabolism , Mice, Knockout , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Spleen/cytology
3.
Immunity ; 46(4): 635-648, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410990

ABSTRACT

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Subject(s)
Inflammation/immunology , Interleukin-1alpha/immunology , Myeloid Differentiation Factor 88/immunology , Skin Diseases/immunology , Syk Kinase/immunology , Animals , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout , Models, Immunological , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Skin Diseases/genetics , Skin Diseases/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism
4.
J Immunol ; 213(8): 1255-1263, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39240186

ABSTRACT

Dendritic cell (DC)-based vaccines are promising immunotherapies for cancer. Although DC-based therapies are known to activate tumor-specific T cells, the interplay between DCs and NK cells in this setting is not fully understood. In this study, we demonstrated a novel TLR7/ mitochondrial reactive oxygen species (mROS)/IL-12 axis that drives potent NK cell responses against tumors. We showed that TLR7 activation by imiquimod in peripheral blood monocyte-derived CD11c+ DCs triggered mROS production, leading to enhanced IL-12 secretion and subsequent NK cell activation, as evidenced by increased IFN-γ production and tumor cell cytotoxicity. Notably, mROS neutralization abrogates NK cell-mediated tumor cell lysis, and TLR7-mediated DC activation of NK cells occurs independently of MyD88, suggesting involvement of the noncanonical NF-κB pathway. Our findings provide a rationale for targeting the TLR7/mROS/IL-12 axis to enhance the efficacy of DC-based cancer immunotherapy.


Subject(s)
Dendritic Cells , Interleukin-12 , Killer Cells, Natural , Lymphocyte Activation , Mitochondria , Reactive Oxygen Species , Toll-Like Receptor 7 , Dendritic Cells/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Humans , Reactive Oxygen Species/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Killer Cells, Natural/immunology , Mitochondria/metabolism , Mitochondria/immunology , Lymphocyte Activation/immunology , Imiquimod , Monocytes/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/immunology , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor , Aminoquinolines/pharmacology , Animals
5.
Nat Immunol ; 13(6): 612-20, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22581261

ABSTRACT

The adaptors DOCK8 and MyD88 have been linked to serological memory. Here we report that DOCK8-deficient patients had impaired antibody responses and considerably fewer CD27(+) memory B cells. B cell proliferation and immunoglobulin production driven by Toll-like receptor 9 (TLR9) were considerably lower in DOCK8-deficient B cells, but those driven by the costimulatory molecule CD40 were not. In contrast, TLR9-driven expression of AICDA (which encodes the cytidine deaminase AID), the immunoglobulin receptor CD23 and the costimulatory molecule CD86 and activation of the transcription factor NF-κB, the kinase p38 and the GTPase Rac1 were intact. DOCK8 associated constitutively with MyD88 and the tyrosine kinase Pyk2 in normal B cells. After ligation of TLR9, DOCK8 became tyrosine-phosphorylated by Pyk2, bound the Src-family kinase Lyn and linked TLR9 to a Src-kinase Syk-transcription factor STAT3 cascade essential for TLR9-driven B cell proliferation and differentiation. Thus, DOCK8 functions as an adaptor in a TLR9-MyD88 signaling pathway in B cells.


Subject(s)
B-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/immunology , Immunologic Memory/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 9/immunology , Adolescent , Animals , Cell Differentiation/immunology , Child , Child, Preschool , Flow Cytometry , Focal Adhesion Kinase 2/immunology , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phosphorylation , STAT3 Transcription Factor/immunology , src-Family Kinases/immunology
6.
Immunity ; 43(6): 1148-59, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682986

ABSTRACT

Toll-like receptor 9 (TLR9), its adaptor MyD88, the downstream transcription factor interferon regulatory factor 7 (IRF7), and type I interferons (IFN-I) are all required for resistance to infection with ectromelia virus (ECTV). However, it is not known how or in which cells these effectors function to promote survival. Here, we showed that after infection with ECTV, the TLR9-MyD88-IRF7 pathway was necessary in CD11c(+) cells for the expression of proinflammatory cytokines and the recruitment of inflammatory monocytes (iMos) to the draining lymph node (dLN). In the dLN, the major producers of IFN-I were infected iMos, which used the DNA sensor-adaptor STING to activate IRF7 and nuclear factor κB (NF-κB) signaling to induce the expression of IFN-α and IFN-ß, respectively. Thus, in vivo, two pathways of DNA pathogen sensing act sequentially in two distinct cell types to orchestrate resistance to a viral disease.


Subject(s)
Interferon Type I/immunology , Monocytes/immunology , Signal Transduction/immunology , Animals , DNA Virus Infections/immunology , Ectromelia virus , Ectromelia, Infectious/immunology , Flow Cytometry , Interferon Regulatory Factor-7/immunology , Interferon Type I/biosynthesis , Lymph Nodes/immunology , Membrane Proteins/immunology , Mice , Mice, Knockout , Mice, Mutant Strains , Myeloid Differentiation Factor 88/immunology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9/immunology
7.
Fish Shellfish Immunol ; 151: 109724, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942251

ABSTRACT

Takifugu rubripes is a highly valued cultured fish in Asia, while pathogen infections can result in severe diseases and lead to substantial economic losses. Toll-like receptors (TLRs), as pattern recognition receptors, play a crucial role on recognition pathogens and initiation innate immune response. However, the immunological properties of teleost-specific TLR23 remain largely unknown. In this study, we investigated the biological functions of TLR23 (TrTLR23) from T. rubripes, found that TrTLR23 existed in various organs. Following bacterial pathogen challenge, the expression levels of TrTLR23 were significantly increased in immune related organs. TrTLR23 located on the cellular membrane and specifically recognized pathogenic microorganism. Co-immunoprecipitation and antibody blocking analysis revealed that TrTLR23 recruited myeloid differentiation primary response protein (MyD88), thereby mediating the activation of the ERK signaling pathway. Furthermore, in vivo showed that, when TrTLR23 is overexpressed in T. rubripes, bacterial replication in fish tissues is significantly inhibited. Consistently, when TrTLR23 expression in T. rubripes is knocked down, bacterial replication is significantly enhanced. In conclusion, these findings suggested that TrTLR23 played a critical role on mediation TLR23-MyD88-ERK axis against bacterial infection. This study revealed that TLR23 involved in the innate immune mechanism, and provided the foundation for development disease control strategies in teleost.


Subject(s)
Fish Diseases , Fish Proteins , Immunity, Innate , Myeloid Differentiation Factor 88 , Takifugu , Toll-Like Receptors , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Takifugu/immunology , Takifugu/genetics , Fish Diseases/immunology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/immunology , Immunity, Innate/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , MAP Kinase Signaling System/immunology , Gene Expression Regulation/immunology , Edwardsiella/physiology , Edwardsiella/immunology , Vibrio/physiology
8.
Fish Shellfish Immunol ; 151: 109697, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38871139

ABSTRACT

Myeloid differentiation factor-88 (MyD88) is a key adaptor of the toll-like receptor (TLR) signaling pathway and plays a crucial role in innate immune signal transduction in animals. However, the MyD88-mediated signal transduction mechanism in shellfish has not been well studied. In this study, a new MyD88 gene, CfMyD88-2, was identified in the Zhikong scallop, Chlamys farreri. The 1779 bp long open reading frame encodes 592 amino acids. The N-terminus of CfMyD88-2 contains a conserved death domain (DD), followed by a TIR (TLR/Interleukin-1 Receptor) domain. The results of the multi-sequence comparison showed that the TIR domain sequences were highly conserved. Phylogenetic analysis revealed that CfMyD88-2 was first associated with Mizuhopecten yessoensis MyD88-4 and Argopecten irradians MyD88-4. CfMyD88-2 mRNA was expressed in all scallop tissues, as detected by qRT-PCR, and the expression level was the highest in the mantle and hepatopancreas. In addition, CfMyD88-2 mRNA expression significantly increased after pathogen-associated molecular patterns (PAMPs, such as lipopolysaccharide, peptidoglycan, or polyinosinic-polycytidylic acid) stimulation. The results of the co-immunoprecipitation experiments in HEK293T cells showed that both CfMyD88-1 and CfMyD88-2 interacted with the TLR protein of scallops, suggesting the existence of more than one functional TLR-MyD88 signaling axis in scallops. Dual luciferase reporter gene assays indicated that the overexpressed CfMyD88-2 in HEK293T cells activated interferon (IFN) α, IFN-ß, IFN-γ, and NF-κB reporter genes, indicating that the protein has multiple functions. The results of the subcellular localization experiment uncovered that CfMyD88-2 was mainly localized in the cytoplasm of human cells. In summary, the novel identified CfMyD88-2 can respond to the challenge of PAMPs, participate in TLR immune signaling, and may activate downstream effector genes such as NF-κB gene. These research results will be useful in advancing the theory of innate immunity in invertebrates and provide a reference for the selection of disease-resistant scallops in the future.


Subject(s)
Amino Acid Sequence , Gene Expression Regulation , Immunity, Innate , Myeloid Differentiation Factor 88 , Pectinidae , Phylogeny , Sequence Alignment , Toll-Like Receptors , Animals , Immunity, Innate/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Pectinidae/immunology , Pectinidae/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/chemistry , Sequence Alignment/veterinary , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Signal Transduction/immunology , Humans , HEK293 Cells , Base Sequence
9.
Fish Shellfish Immunol ; 149: 109550, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593891

ABSTRACT

Signal transducing adapter molecule 2 (STAM2), a member of the Signal Transducing Adapter Molecule (STAM) family, is a protein with significant implications in diverse signaling pathways and endocytic membrane trafficking. However, the role of the STAM2, especially in fish, remains largely unknown. In this study, we discovered that STAM2 negatively regulates the NF-κB signaling pathway, and its inhibitory effect is enhanced upon LPS induction. Our study confirmed that STAM2 can enhance the degradation of myeloid differentiation primary-response protein 88 (MyD88), an upstream regulator of NF-κB pathway. Furthermore, the UIM domain of STAM2 is important for the inhibition of MyD88. Mechanistically, STAM2 inhibits the NF-κB signaling pathway by targeting the MyD88 autophagy pathway. In addition, we showed that STAM2 promotes the proliferation of Vibrio harveyi. In summary, our study reveals that STAM2 inhibits NF-κB signaling activation and mediates innate immunity in teleost via the autophagy pathway.


Subject(s)
Fish Diseases , Fish Proteins , Immunity, Innate , Myeloid Differentiation Factor 88 , NF-kappa B , Perciformes , Vibrio Infections , Vibrio , Animals , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/immunology , Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Lipopolysaccharides/pharmacology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/immunology , NF-kappa B/metabolism , NF-kappa B/immunology , NF-kappa B/genetics , Perciformes/immunology , Perciformes/genetics , Signal Transduction/immunology , Vibrio/physiology , Vibrio Infections/immunology , Vibrio Infections/veterinary
10.
Fish Shellfish Immunol ; 151: 109734, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950759

ABSTRACT

Toll-like receptors (TLRs) are pattern recognition receptors that trigger host immune responses against various pathogens by detecting evolutionarily conserved pathogen-associated molecular patterns (PAMPs). TLR21 is a member of the Toll-like receptor family, and emerging data suggest that it recognises unmethylated CpG DNA and is considered a functional homologue of mammalian TLR9. However, little is known regarding the role of TLR21 in the fish immune response. In the present study, we isolated the cDNA sequence of TLR21 from the largemouth bass (Micropterus salmoides) and termed it MsTLR21. The MsTLR21 gene contained an open reading frame (ORF) of 2931 bp and encodes a polypeptide of 976 amino acids. The predicted MsTLR21 protein has two conserved domains, a conserved leucine-rich repeats (LRR) domain and a C-terminal Toll-interleukin (IL) receptor (TIR) domain, similar to those of other fish and mammals. In healthy largemouth bass, the TLR21 transcript was broadly expressed in all the examined tissues, with the highest expression levels in the gills. After challenge with Nocardia seriolae and polyinosinic polycytidylic acid (Poly[I:C]), the expression of TLR21 mRNA was upregulated or downregulated in all tissues tested. Overexpression of TLR21 in 293T cells showed that it has a positive regulatory effect on nuclear factor-kappaB (NF-κB) and interferons-ß (IFN-ß) activity. Subcellular localisation analysis showed that TLR21 was expressed in the cytoplasm. We performed pull-down assays and determined that TLR21 did not interact with myeloid differentiation primary response gene 88 (Myd88); however, it interacted with TIR domain-containing adaptor inducing interferon-ß (TRIF). Taken together, these findings suggest that MsTLR21 plays important roles in TLR/IL-1R signalling pathways and the immune response to pathogen invasion.


Subject(s)
Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Bass , Fish Diseases , Fish Proteins , NF-kappa B , Phylogeny , Animals , Bass/immunology , Bass/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/immunology , Fish Diseases/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Signal Transduction/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Sequence Alignment/veterinary , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/chemistry , Gene Expression Profiling/veterinary , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism , Base Sequence
11.
PLoS Pathog ; 17(10): e1009970, 2021 10.
Article in English | MEDLINE | ID: mdl-34597344

ABSTRACT

Toxoplasma gondii is an orally acquired pathogen that induces strong IFN-γ based immunity conferring protection but that can also be the cause of immunopathology. The response in mice is driven in part by well-characterized MyD88-dependent signaling pathways. Here we focus on induction of less well understood immune responses that do not involve this Toll-like receptor (TLR)/IL-1 family receptor adaptor molecule, in particular as they occur in the intestinal mucosa. Using eYFP-IL-12p40 reporter mice on an MyD88-/- background, we identified dendritic cells, macrophages, and neutrophils as cellular sources of MyD88-independent IL-12 after peroral T. gondii infection. Infection-induced IL-12 was lower in the absence of MyD88, but was still clearly above noninfected levels. Overall, this carried through to the IFN-γ response, which while generally decreased was still remarkably robust in the absence of MyD88. In the latter mice, IL-12 was strictly required to induce type I immunity. Type 1 and type 3 innate lymphoid cells (ILC), CD4+ T cells, and CD8+ T cells each contributed to the IFN-γ pool. We report that ILC3 were expanded in infected MyD88-/- mice relative to their MyD88+/+ counterparts, suggesting a compensatory response triggered by loss of MyD88. Furthermore, bacterial flagellin and Toxoplasma specific CD4+ T cell populations in the lamina propria expanded in response to infection in both WT and KO mice. Finally, we show that My88-independent IL-12 and T cell mediated IFN-γ production require the presence of the intestinal microbiota. Our results identify MyD88-independent intestinal immune pathways induced by T. gondii including myeloid cell derived IL-12 production, downstream type I immunity and IFN-γ production by ILC1, ILC3, and T lymphocytes. Collectively, our data reveal an underlying network of immune responses that do not involve signaling through MyD88.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal/immunology , Interleukin-12 Subunit p40/immunology , Toxoplasmosis, Animal/immunology , Animals , Intestinal Mucosa/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology , Toll-Like Receptors/deficiency , Toll-Like Receptors/immunology , Toxoplasma/immunology
12.
Nat Immunol ; 12(5): 416-24, 2011 May.
Article in English | MEDLINE | ID: mdl-21441935

ABSTRACT

The molecular mechanisms involved in the full activation of innate immunity achieved through Toll-like receptors (TLRs) remain to be fully elucidated. In addition to their classical antigen-presenting function, major histocompatibility complex (MHC) class II molecules might mediate reverse signaling. Here we report that deficiency in MHC class II attenuated the TLR-triggered production of proinflammatory cytokines and type I interferon in macrophages and dendritic cells, which protected mice from endotoxin shock. Intracellular MHC class II molecules interacted with the tyrosine kinase Btk via the costimulatory molecule CD40 and maintained Btk activation, but cell surface MHC class II molecules did not. Then, Btk interacted with the adaptor molecules MyD88 and TRIF and thereby promoted TLR signaling. Therefore, intracellular MHC class II molecules can act as adaptors, promoting full activation of TLR-triggered innate immune responses.


Subject(s)
Histocompatibility Antigens Class II/immunology , Immunity, Innate/immunology , Protein-Tyrosine Kinases/metabolism , Toll-Like Receptors/immunology , Adaptor Proteins, Vesicular Transport/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , CD40 Antigens/immunology , Cell Line , Cytokines/blood , Cytokines/immunology , Enzyme Activation , Immunoblotting , Interferon-gamma/blood , Interferon-gamma/immunology , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Protein-Tyrosine Kinases/immunology , Sepsis/immunology , Specific Pathogen-Free Organisms
13.
Nat Immunol ; 12(4): 344-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21358639

ABSTRACT

Sepsis is one of the most challenging health problems worldwide. Here we found that phagocytes from patients with sepsis had considerable upregulation of Toll-like receptor 4 (TLR4) and TLR2; however, shock-inducing inflammatory responses mediated by these TLRs were inhibited by ES-62, an immunomodulator secreted by the filarial nematode Acanthocheilonema viteae. ES-62 subverted TLR4 signaling to block TLR2- and TLR4-driven inflammatory responses via autophagosome-mediated downregulation of the TLR adaptor-transducer MyD88. In vivo, ES-62 protected mice against endotoxic and polymicrobial septic shock by TLR4-mediated induction of autophagy and was protective even when administered after the induction of sepsis. Given that the treatments for septic shock at present are inadequate, the autophagy-dependent mechanism of action by ES-62 might form the basis for urgently needed therapeutic intervention against this life-threatening condition.


Subject(s)
Helminth Proteins/pharmacology , Myeloid Differentiation Factor 88/metabolism , Phagosomes/drug effects , Shock, Septic/prevention & control , Toll-Like Receptor 4/metabolism , Animals , Autophagy/drug effects , Autophagy/immunology , Cells, Cultured , Female , Humans , Immunoblotting , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Myeloid Differentiation Factor 88/immunology , Phagosomes/immunology , Phagosomes/metabolism , Shock, Septic/genetics , Shock, Septic/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
14.
Immunity ; 41(2): 311-24, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25088769

ABSTRACT

Nod2 has been extensively characterized as a bacterial sensor that induces an antimicrobial and inflammatory gene expression program. Therefore, it is unclear why Nod2 mutations that disrupt bacterial recognition are paradoxically among the highest risk factors for Crohn's disease, which involves an exaggerated immune response directed at intestinal bacteria. Here, we identified several abnormalities in the small-intestinal epithelium of Nod2(-/-) mice including inflammatory gene expression and goblet cell dysfunction, which were associated with excess interferon-γ production by intraepithelial lymphocytes and Myd88 activity. Remarkably, these abnormalities were dependent on the expansion of a common member of the intestinal microbiota Bacteroides vulgatus, which also mediated exacerbated inflammation in Nod2(-/-) mice upon small-intestinal injury. These results indicate that Nod2 prevents inflammatory pathologies by controlling the microbiota and support a multihit disease model involving specific gene-microbe interactions.


Subject(s)
Bacteroides/immunology , Disease Susceptibility/immunology , Enteritis/immunology , Intestine, Small/immunology , Nod2 Signaling Adaptor Protein/genetics , Animals , Bacterial Typing Techniques , Crohn Disease/immunology , Enteritis/genetics , Goblet Cells/pathology , Inflammation/genetics , Inflammation/immunology , Interferon-gamma/biosynthesis , Intestinal Mucosa/immunology , Intestine, Small/microbiology , Lymphocytes/immunology , Mice , Mice, Knockout , Microbiota/immunology , Myeloid Differentiation Factor 88/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/immunology
15.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30131424

ABSTRACT

Plasmacytoid dendritic cells (pDC) are the major source of type I interferons (IFN-I) during viral infections, in response to triggering of endosomal Toll-like receptors (TLRs) 7 or 9 by viral single-stranded RNA or unmethylated CpG DNA, respectively. Synthetic ligands have been used to disentangle the underlying signaling pathways. The adaptor protein AP3 is necessary to transport molecular complexes of TLRs, synthetic CpG DNA, and MyD88 into endosomal compartments allowing interferon regulatory factor 7 (IRF7) recruitment whose phosphorylation then initiates IFN-I production. High basal expression of IRF7 by pDC and its further enhancement by positive IFN-I feedback signaling appear to be necessary for robust cytokine production. In contrast, we show here that in vivo during mouse cytomegalovirus (MCMV) infection pDC produce high amounts of IFN-I downstream of the TLR9-to-MyD88-to-IRF7 signaling pathway without requiring IFN-I positive feedback, high IRF7 expression, or AP3-driven endosomal routing of TLRs. Hence, the current model of the molecular requirements for professional IFN-I production by pDC, established by using synthetic TLR ligands, does not strictly apply to a physiological viral infection.


Subject(s)
Dendritic Cells/immunology , Herpesviridae Infections/immunology , Interferon Type I/immunology , Muromegalovirus/immunology , Signal Transduction/immunology , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Animals , Dendritic Cells/pathology , Endosomes/genetics , Endosomes/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
16.
PLoS Pathog ; 16(1): e1008292, 2020 01.
Article in English | MEDLINE | ID: mdl-31999809

ABSTRACT

Humoral immune responses initiate in the lymph node draining the site of viral infection (dLN). Some viruses subvert LN B cell activation; however, our knowledge of viral hindrance of B cell responses of important human pathogens is lacking. Here, we define mechanisms whereby chikungunya virus (CHIKV), a mosquito-transmitted RNA virus that causes outbreaks of acute and chronic arthritis in humans, hinders dLN antiviral B cell responses. Infection of WT mice with pathogenic, but not acutely cleared CHIKV, induced MyD88-dependent recruitment of monocytes and neutrophils to the dLN. Blocking this influx improved lymphocyte accumulation, dLN organization, and CHIKV-specific B cell responses. Both inducible nitric oxide synthase (iNOS) and the phagocyte NADPH oxidase (Nox2) contributed to impaired dLN organization and function. Infiltrating monocytes expressed iNOS through a local IRF5- and IFNAR1-dependent pathway that was partially TLR7-dependent. Together, our data suggest that pathogenic CHIKV triggers the influx and activation of monocytes and neutrophils in the dLN that impairs virus-specific B cell responses.


Subject(s)
B-Lymphocytes/immunology , Chikungunya Fever/immunology , Interferon Regulatory Factors/immunology , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , NADPH Oxidase 2/immunology , Neutrophils/immunology , Nitric Oxide Synthase Type II/immunology , Animals , Chikungunya Fever/virology , Chikungunya virus/physiology , Humans , Interferon Regulatory Factors/genetics , Lymph Nodes/immunology , Lymph Nodes/virology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NADPH Oxidase 2/genetics , Nitric Oxide Synthase Type II/genetics
17.
PLoS Pathog ; 16(5): e1008572, 2020 05.
Article in English | MEDLINE | ID: mdl-32413093

ABSTRACT

The apicomplexan Toxoplasma gondii induces strong protective immunity dependent upon recognition by Toll-like receptors (TLR)11 and 12 operating in conjunction with MyD88 in the murine host. However, TLR11 and 12 proteins are not present in humans, inspiring us to investigate MyD88-independent pathways of resistance. Using bicistronic IL-12-YFP reporter mice on MyD88+/+ and MyD88-/- genetic backgrounds, we show that CD11c+MHCII+F4/80- dendritic cells, F4/80+ macrophages, and Ly6G+ neutrophils were the dominant cellular sources of IL-12 in both wild type and MyD88 deficient mice after parasite challenge. Parasite dense granule protein GRA24 induces p38 MAPK activation and subsequent IL-12 production in host macrophages. We show that Toxoplasma triggers an early and late p38 MAPK phosphorylation response in MyD88+/+ and MyD88-/- bone marrow-derived macrophages. Using the uracil auxotrophic Type I T. gondii strain cps1-1, we demonstrate that the late response does not require active parasite proliferation, but strictly depends upon GRA24. By i. p. inoculation with cps1-1 and cps1-1:Δgra24, we identified unique subsets of chemokines and cytokines that were up and downregulated by GRA24. Finally, we demonstrate that cps1-1 triggers a strong host-protective GRA24-dependent Th1 response in the absence of MyD88. Our data identify GRA24 as a major mediator of p38 MAPK activation, IL-12 induction and protective immunity that operates independently of the TLR/MyD88 cascade.


Subject(s)
Interleukin-12/immunology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Myeloid Differentiation Factor 88/immunology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Enzyme Activation/genetics , Enzyme Activation/immunology , Interleukin-12/genetics , MAP Kinase Signaling System/genetics , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/pathology , p38 Mitogen-Activated Protein Kinases/genetics
18.
Nat Immunol ; 11(9): 836-45, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20676093

ABSTRACT

BAFF and APRIL are innate immune mediators that trigger immunoglobulin G (IgG) and IgA class-switch recombination (CSR) in B cells by engaging the receptor TACI. The mechanism that underlies CSR signaling by TACI remains unknown. Here we found that the cytoplasmic domain of TACI encompasses a conserved motif that bound MyD88, an adaptor that activates transcription factor NF-kappaB signaling pathways via a Toll-interleukin 1 (IL-1) receptor (TIR) domain. TACI lacks a TIR domain, yet triggered CSR via the DNA-editing enzyme AID by activating NF-kappaB through a Toll-like receptor (TLR)-like MyD88-IRAK1-IRAK4-TRAF6-TAK1 pathway. TACI-induced CSR was impaired in mice and humans lacking MyD88 or the kinase IRAK4, which indicates that MyD88 controls a B cell-intrinsic, TIR-independent, TACI-dependent pathway for immunoglobulin diversification.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Myeloid Differentiation Factor 88/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Animals , Cells, Cultured , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction
19.
Nat Immunol ; 11(8): 734-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20639876

ABSTRACT

Integrins are critical for the migration and function of leukocytes in inflammation. However, the interaction between integrin alpha(M) (CD11b), which has high expression in monocytes and macrophages, and Toll-like receptor (TLR)-triggered innate immunity remains unclear. Here we report that CD11b deficiency enhanced TLR-mediated responses in macrophages, rendering mice more susceptible to endotoxin shock and Escherichia coli-caused sepsis. CD11b was activated by TLR-triggered phosphatidylinositol 3-OH kinase (PI(3)K) and the effector RapL and fed back to inhibit TLR signaling by activating the tyrosine kinases Src and Syk. Syk interacted with and induced tyrosine phosphorylation of MyD88 and TRIF, which led to degradation of these adaptor molecules by the E3 ubiquitin ligase Cbl-b. Thus, TLR-triggered, active CD11b integrin engages in crosstalk with the MyD88 and TRIF pathways and subsequently inhibits TLR signaling in innate immune responses.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD11b Antigen/immunology , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins c-cbl/immunology , Toll-Like Receptors/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Enzyme Activation , Escherichia coli/immunology , Escherichia coli Infections/immunology , Gene Expression Regulation, Enzymologic , Immunity, Innate/immunology , Inflammation/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction , Syk Kinase , src-Family Kinases/immunology
20.
Immunity ; 38(3): 489-501, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23521885

ABSTRACT

The motheaten mouse has long served as a paradigm for complex autoimmune and inflammatory disease. Null mutations in Ptpn6, which encodes the nonreceptor protein-tyrosine phosphatase Shp1, cause the motheaten phenotype. However, Shp1 regulates multiple signaling pathways in different hematopoietic cell types, so the cellular and molecular mechanism of autoimmunity and inflammation in the motheaten mouse has remained unclear. By using floxed Ptpn6 mice, we dissected the contribution of innate immune cells to the motheaten phenotype. Ptpn6 deletion in neutrophils resulted in cutaneous inflammation, but not autoimmunity, providing an animal model of human neutrophilic dermatoses. By contrast, dendritic cell deletion caused severe autoimmunity, without inflammation. Genetic and biochemical analysis showed that inflammation was caused by enhanced neutrophil integrin signaling through Src-family and Syk kinases, whereas autoimmunity resulted from exaggerated MyD88-dependent signaling in dendritic cells. Our data demonstrate that disruption of distinct Shp1-regulated pathways in different cell types combine to cause motheaten disease.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Inflammation/immunology , Neutrophils/immunology , Animals , Autoimmunity/genetics , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , Calgranulin A/genetics , Calgranulin A/immunology , Calgranulin A/metabolism , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/metabolism , Dermis/immunology , Dermis/metabolism , Dermis/pathology , Female , Flow Cytometry , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Syk Kinase , src-Family Kinases/immunology , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL