Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 411
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 17(8): e1009905, 2021 08.
Article in English | MEDLINE | ID: mdl-34449811

ABSTRACT

B1 cells, a subset of B lymphocytes whose developmental origin, phenotype, and function differ from that of conventional B2 cells, are the main source of "natural" IgM but can also respond to infection by rapidly producing pathogen-specific IgM directed against T-independent antigens. Francisella tularensis (Ft) is a Gram-negative bacterium that causes tularemia. Infection with Ft Live Vaccine Strain activates B1 cells for production of IgM directed against the bacterial LPS in a process incompletely understood. Here we show that immunization with purified Ft LPS elicits production of LPS-specific IgM and IgG3 by B1 cells independently of TLR2 or MyD88. Immunization, but not infection, generated peritoneum-resident memory B1 cells that differentiated into LPS-specific antibody secreting cells (ASC) upon secondary challenge. IL-5 was rapidly induced by immunization with Ft LPS and was required for production of LPS-specific IgM. Antibody-mediated depletion of ILC2 indicated that these cells were the source of IL-5 and were required for IgM production. IL-25, an alarmin that strongly activates ILC2, was rapidly secreted in response to immunization or infection and its administration to mice significantly increased IgM production and B1 cell differentiation to ASC. Conversely, mice lacking IL-17RB, the IL-25 receptor, showed impaired IL-5 induction, IgM production, and B1 ASC differentiation in response to immunization. Administration of IL-5 to Il17rb-/- mice rescued these B1 cells-mediated responses. Il17rb-/- mice were more susceptible to infection with Ft LVS and failed to develop immunity upon secondary challenge suggesting that LPS-specific IgM is one of the protective adaptive immune mechanisms against tularemia. Our results indicated that immunization with Ft LPS triggers production of IL-25 that, through stimulation of IL-5 release by ILC2, promotes B1 cells activation and differentiation into IgM secreting cells. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against T-independent bacterial antigens.


Subject(s)
Antibodies, Bacterial/immunology , B-Lymphocyte Subsets/immunology , Francisella tularensis/immunology , Immunoglobulin M/immunology , Interleukin-5/metabolism , Interleukins/metabolism , Lipopolysaccharides/pharmacology , Animals , Antibodies, Bacterial/metabolism , B-Lymphocyte Subsets/metabolism , Immunity, Innate , Immunoglobulin M/metabolism , Interleukin-5/genetics , Interleukins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Receptors, Interleukin-17/physiology , Toll-Like Receptor 2/physiology , Tularemia/immunology , Tularemia/microbiology , Tularemia/pathology
2.
PLoS Pathog ; 16(5): e1008188, 2020 05.
Article in English | MEDLINE | ID: mdl-32365080

ABSTRACT

As a canonical adaptor for the Toll-like receptor (TLR) family, myeloid differentiation primary response protein 88 (MyD88) has crucial roles in host defense against infection by microbial pathogens, and its dysregulation might induce autoimmune diseases. Here, we demonstrate that the chicken Cullin 3-based ubiquitin ligase adaptor Speckle-type BTB-POZ protein (chSPOP) recognizes the intermediate domain of chicken MyD88 (chMyD88) and degrades it through the proteasome pathway. Knockdown or genetic ablation of chSPOP leads to aberrant elevation of chMyD88 protein. Through this interaction, chSPOP negatively regulates NF-κB pathway activity and thus the production of IL-1ß upon LPS challenge in chicken macrophages. Furthermore, Spop-deficient mice are more susceptible to infection with Salmonella typhimurium. Collectively, these findings demonstrate MyD88 as a bona fide substrate of SPOP and uncover a mechanism by which SPOP regulates MyD88 abundance and disease susceptibility.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , CHO Cells , Chickens/metabolism , Cricetulus , Cullin Proteins/metabolism , HeLa Cells , Humans , Immunity, Innate/physiology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Nuclear Proteins/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proteostasis/physiology , Repressor Proteins/physiology , Signal Transduction , Ubiquitin/metabolism , Ubiquitination
3.
PLoS Pathog ; 16(8): e1008639, 2020 08.
Article in English | MEDLINE | ID: mdl-32790743

ABSTRACT

Leptospirosis is a worldwide re-emerging zoonosis caused by pathogenic Leptospira spp. All vertebrate species can be infected; humans are sensitive hosts whereas other species, such as rodents, may become long-term renal carrier reservoirs. Upon infection, innate immune responses are initiated by recognition of Microbial Associated Molecular Patterns (MAMPs) by Pattern Recognition Receptors (PRRs). Among MAMPs, the lipopolysaccharide (LPS) is recognized by the Toll-Like-Receptor 4 (TLR4) and activates both the MyD88-dependent pathway at the plasma membrane and the TRIF-dependent pathway after TLR4 internalization. We previously showed that leptospiral LPS is not recognized by the human-TLR4, whereas it signals through mouse-TLR4 (mTLR4), which mediates mouse resistance to acute leptospirosis. However, although resistant, mice are known to be chronically infected by leptospires. Interestingly, the leptospiral LPS has low endotoxicity in mouse cells and is an agonist of TLR2, the sensor for bacterial lipoproteins. Here, we investigated the signaling properties of the leptospiral LPS in mouse macrophages. Using confocal microscopy and flow cytometry, we showed that the LPS of L. interrogans did not induce internalization of mTLR4, unlike the LPS of Escherichia coli. Consequently, the LPS failed to induce the production of the TRIF-dependent nitric oxide and RANTES, both important antimicrobial responses. Using shorter LPS and LPS devoid of TLR2 activity, we further found this mTLR4-TRIF escape to be dependent on both the co-purifying lipoproteins and the full-length O antigen. Furthermore, our data suggest that the O antigen could alter the binding of the leptospiral LPS to the co-receptor CD14 that is essential for TLR4-TRIF activation. Overall, we describe here a novel leptospiral immune escape mechanism from mouse macrophages and hypothesize that the LPS altered signaling could contribute to the stealthiness and chronicity of the leptospires in mice.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Leptospira/immunology , Leptospirosis/immunology , Lipopolysaccharides/metabolism , Lipoproteins/metabolism , O Antigens/metabolism , Toll-Like Receptor 4/physiology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cytokines/metabolism , Female , Leptospirosis/metabolism , Leptospirosis/microbiology , Leptospirosis/pathology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lipoproteins/genetics , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , O Antigens/genetics , Signal Transduction , Toll-Like Receptor 2/physiology
4.
Blood ; 136(6): 684-697, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32325488

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) is a key regulator of NF-κB signaling. Activating single-nucleotide polymorphisms of HOIP, the catalytic subunit of LUBAC, are enriched in patients with activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL), and expression of HOIP, which parallels LUBAC activity, is elevated in ABC-DLBCL samples. Thus, to clarify the precise roles of LUBAC in lymphomagenesis, we generated a mouse model with augmented expression of HOIP in B cells. Interestingly, augmented HOIP expression facilitated DLBCL-like B-cell lymphomagenesis driven by MYD88-activating mutation. The developed lymphoma cells partly shared somatic gene mutations with human DLBCLs, with increased frequency of a typical AID mutation pattern. In vitro analysis revealed that HOIP overexpression protected B cells from DNA damage-induced cell death through NF-κB activation, and analysis of the human DLBCL database showed that expression of HOIP positively correlated with gene signatures representing regulation of apoptosis signaling, as well as NF-κB signaling. These results indicate that HOIP facilitates lymphomagenesis by preventing cell death and augmenting NF-κB signaling, leading to accumulation of AID-mediated mutations. Furthermore, a natural compound that specifically inhibits LUBAC was shown to suppress the tumor growth in a mouse transplantation model. Collectively, our data indicate that LUBAC is crucially involved in B-cell lymphomagenesis through protection against DNA damage-induced cell death and is a suitable therapeutic target for B-cell lymphomas.


Subject(s)
Apoptosis/genetics , B-Lymphocytes/enzymology , Cell Transformation, Neoplastic/genetics , Lymphoma, Large B-Cell, Diffuse/etiology , Multiprotein Complexes/physiology , Ubiquitin-Protein Ligases/genetics , Animals , B-Lymphocytes/pathology , Carrier Proteins/physiology , DNA Damage , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/physiology , Lymphoma, Large B-Cell, Diffuse/enzymology , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Mice, Transgenic , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , NF-kappa B/metabolism , Neoplasm Transplantation , Polymorphism, Single Nucleotide , Polyubiquitin/biosynthesis , Protein Processing, Post-Translational , Transcription Factors/physiology , Transcriptome , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Ubiquitins/physiology
5.
FASEB J ; 35(12): e22019, 2021 12.
Article in English | MEDLINE | ID: mdl-34792819

ABSTRACT

Exogenously applied mature naïve B220+ /CD19+ /IgM+ /IgD+ B cells are strongly protective in the context of tissue injury. However, the mechanisms by which B cells detect tissue injury and aid repair remain elusive. Here, we show in distinct models of skin and brain injury that MyD88-dependent toll-like receptor (TLR) signaling through TLR2/6 and TLR4 is essential for the protective benefit of B cells in vivo, while B cell-specific deletion of MyD88 abrogated this effect. The B cell response to injury was multi-modal with simultaneous production of both regulatory cytokines, such as IL-10, IL-35, and transforming growth factor beta (TGFß), and inflammatory cytokines, such as tumor necrosis factor alpha (TNFα), IL-6, and interferon gamma. Cytometry analysis showed that this response was time and environment-dependent in vivo, with 20%-30% of applied B cells adopting an immune modulatory phenotype with high co-expression of anti- and pro-inflammatory cytokines after 18-48 h at the injury site. B cell treatment reduced the expression of TNFα and increased IL-10 and TGFß in infiltrating immune cells and fibroblasts at the injury site. Proteomic analysis further showed that B cells have a complex time-dependent homeostatic effect on the injured microenvironment, reducing the expression of inflammation-associated proteins, and increasing proteins associated with proliferation, tissue remodeling, and protection from oxidative stress. These findings chart and validate a first mechanistic understanding of the effects of B cells as an immunomodulatory cell therapy in the context of tissue injury.


Subject(s)
B-Lymphocytes/physiology , Brain Injuries/prevention & control , Cytokines/metabolism , Myeloid Differentiation Factor 88/physiology , Skin/immunology , Wound Healing , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Interleukin-10/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Skin/injuries , Skin/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Immunity ; 39(5): 976-85, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24210353

ABSTRACT

Venoms consist of toxic components that are delivered to their victims via bites or stings. Venoms also represent a major class of allergens in humans. Phospholipase A2 (PLA2) is a conserved component of venoms from multiple species and is the major allergen in bee venom. Here we examined how bee venom PLA2 is sensed by the innate immune system and induces a type 2 immune response in mice. We found that bee venom PLA2 induced a T helper type 2 (Th2) cell-type response and group 2 innate lymphoid cell activation via the enzymatic cleavage of membrane phospholipids and release of interleukin-33. Furthermore, we showed that the IgE response to PLA2 could protect mice from future challenge with a near-lethal dose of PLA2. These data suggest that the innate immune system can detect the activity of a conserved component of venoms and induce a protective immune response against a venom toxin.


Subject(s)
Bee Venoms/enzymology , Immunity, Innate/immunology , Immunoglobulin E/biosynthesis , Insect Proteins/immunology , Lysophospholipids/immunology , Phospholipases A2/immunology , Receptors, Interleukin/immunology , Th2 Cells/immunology , Anaphylaxis/etiology , Anaphylaxis/immunology , Anaphylaxis/prevention & control , Animals , Bee Venoms/toxicity , Crotalid Venoms/immunology , Genes, Reporter , Immunoglobulin E/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukins/immunology , Lymphocyte Activation , Melitten/immunology , Membrane Lipids/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/physiology , Ovalbumin/immunology , Phospholipids/metabolism , Receptors, IgE/immunology
7.
Am J Respir Crit Care Med ; 203(9): 1099-1111, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33166473

ABSTRACT

Rationale: Cross-sectional human data suggest that enrichment of oral anaerobic bacteria in the lung is associated with an increased T-helper cell type 17 (Th17) inflammatory phenotype.Objectives: In this study, we evaluated the microbial and host immune-response dynamics after aspiration with oral commensals using a preclinical mouse model.Methods: Aspiration with a mixture of human oral commensals (MOC; Prevotella melaninogenica, Veillonella parvula, and Streptococcus mitis) was modeled in mice followed by variable time of killing. The genetic backgrounds of mice included wild-type, MyD88-knockout, and STAT3C backgrounds.Measurements and Main Results: 16S-rRNA gene sequencing characterized changes in microbiota. Flow cytometry, cytokine measurement via Luminex and RNA host-transcriptome sequencing was used to characterize the host immune phenotype. Although MOC aspiration correlated with lower-airway dysbiosis that resolved within 5 days, it induced an extended inflammatory response associated with IL-17-producing T cells lasting at least 14 days. MyD88 expression was required for the IL-17 response to MOC aspiration, but not for T-cell activation or IFN-γ expression. MOC aspiration before a respiratory challenge with S. pneumoniae led to a decrease in hosts' susceptibility to this pathogen.Conclusions: Thus, in otherwise healthy mice, a single aspiration event with oral commensals is rapidly cleared from the lower airways but induces a prolonged Th17 response that secondarily decreases susceptibility to S. pneumoniae. Translationally, these data implicate an immunoprotective role of episodic microaspiration of oral microbes in the regulation of the lung immune phenotype and mitigation of host susceptibility to infection with lower-airway pathogens.


Subject(s)
Pneumococcal Infections/prevention & control , Streptococcus pneumoniae , Th17 Cells/physiology , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Pneumococcal Infections/etiology , Prevotella melaninogenica , Streptococcus mitis , Veillonella
8.
Infect Immun ; 89(11): e0040721, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34370509

ABSTRACT

During chronic infection with Helicobacter pylori, Schlafen 4-expressing myeloid-derived suppressor cells (SLFN4+ MDSCs) create a microenvironment favoring intestinal metaplasia and neoplastic transformation. SLFN4 can be induced by alpha interferon (IFN-α), which is mainly secreted from plasmacytoid dendritic cells (pDCs). This study tested the hypothesis that Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing pDCs to secrete IFN-α. C57BL/6 mice were gavaged with H. pylori, and infection lasted 2, 4, or 6 months. Mouse pDCs were isolated from bone marrow of wild-type C57BL/6J mice. The results showed that H. pylori infection increased the number of SLFN4+ MDSCs by inducing IFN-α expression in mice. Further mechanistic experiments unraveled that IFN-α induced SLFN4 transcription by binding to the Slfn4 promoter. Furthermore, H. pylori infection stimulated pDCs to secrete IFN-α by activating the TLR9-MyD88-IRF7 pathway. Collectively, Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing secretion of IFN-α from pDCs.


Subject(s)
Carrier Proteins/genetics , Dendritic Cells/immunology , Helicobacter Infections/immunology , Helicobacter pylori , Interferon Type I/biosynthesis , Myeloid-Derived Suppressor Cells/cytology , Animals , Cell Differentiation , Interferon Regulatory Factor-7/physiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Promoter Regions, Genetic , Toll-Like Receptor 9/physiology
9.
PLoS Pathog ; 15(4): e1007744, 2019 04.
Article in English | MEDLINE | ID: mdl-30978245

ABSTRACT

Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.


Subject(s)
Bone Resorption/immunology , Myeloid Differentiation Factor 88/physiology , Osteoclasts/immunology , Osteomyelitis/immunology , Receptors, Interleukin-1 Type I/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bone Resorption/metabolism , Bone Resorption/microbiology , Cell Differentiation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteomyelitis/metabolism , Osteomyelitis/microbiology , Signal Transduction , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
10.
Am J Pathol ; 190(1): 176-189, 2020 01.
Article in English | MEDLINE | ID: mdl-31676329

ABSTRACT

Nephronophthisis (NPHP), the leading genetic cause of end-stage renal failure in children and young adults, is a group of autosomal recessive diseases characterized by kidney-cyst degeneration and fibrosis for which no therapy is currently available. To date, mutations in >25 genes have been identified as causes of this disease that, in several cases, result in chronic DNA damage in kidney tubular cells. Among such mutations, those in the transcription factor-encoding GLIS2 cause NPHP type 7. Loss of function of mouse Glis2 causes senescence of kidney tubular cells. Senescent cells secrete proinflammatory molecules that induce progressive organ damage through several pathways, among which NF-κB signaling is prevalent. Herein, we show that the NF-κB signaling is active in Glis2 knockout kidney epithelial cells and that genetic inactivation of the toll-like receptor (TLR)/IL-1 receptor or pharmacologic elimination of senescent cells (senolytic therapy) reduces tubule damage, fibrosis, and apoptosis in the Glis2 mouse model of NPHP. Notably, in Glis2, Tlr2 double knockouts, senescence was also reduced and proliferation was increased, suggesting that loss of TLR2 activity improves the regenerative potential of tubular cells in Glis2 knockout kidneys. Our results further suggest that a combination of TLR/IL-1 receptor inhibition and senolytic therapy may delay the progression of kidney disease in NPHP type 7 and other forms of this disease.


Subject(s)
Cellular Senescence/immunology , Disease Models, Animal , Immunity, Innate/immunology , Kidney Diseases, Cystic/pathology , Kidney Tubules/pathology , Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Animals , Apoptosis , Kidney Diseases, Cystic/immunology , Kidney Diseases, Cystic/metabolism , Kidney Tubules/immunology , Kidney Tubules/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Toll-Like Receptor 2/physiology
11.
Nat Immunol ; 10(9): 965-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668221

ABSTRACT

The intensity and duration of immune responses are controlled by many proteins that modulate Toll-like receptor (TLR) signaling. TANK has been linked to positive regulation of the transcription factors IRF3 and NF-kappaB. Here we demonstrate that TANK is not involved in interferon responses and is a negative regulator of proinflammatory cytokine production induced by TLR signaling. TLR-induced polyubiquitination of the ubiquitin ligase TRAF6 was upregulated in Tank(-/-) macrophages. Notably, Tank(-/-) mice spontaneously developed fatal glomerulonephritis owing to deposition of immune complexes. Autoantibody production in Tank(-/-) mice was abrogated by antibiotic treatment or the absence of interleukin 6 (IL-6) or the adaptor MyD88. Our results demonstrate that constitutive TLR signaling by intestinal commensal microflora is suppressed by TANK.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autoimmune Diseases/prevention & control , Glomerulonephritis/prevention & control , Signal Transduction/physiology , Toll-Like Receptors/physiology , Animals , Autoimmunity , CD40 Antigens/physiology , Female , Intestines/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Receptors, Antigen, B-Cell/physiology , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin/metabolism
12.
FASEB J ; 34(9): 12324-12337, 2020 09.
Article in English | MEDLINE | ID: mdl-32662527

ABSTRACT

Autophagy is an important mechanism for cellular homeostasis and survival during pathologic stress conditions in the kidney, such as ischemia-reperfusion (IR) injury. In this study, renal IR was induced in female C57BL/6 mice after melatonin administration. Renal function, histological damage, inflammatory infiltration, cytokine production, oxidative stress, antioxidant capacity, autophagy changing, apoptosis levels, and autophagy-associated intracellular signaling pathway were assessed to evaluate the impact of antecedent melatonin treatment on IR-induced renal injury. The administration of melatonin resulted in significantly preserved renal function, and the protective effect was associated with ameliorated oxidative stress, limited pro-inflammatory cytokine production, and neutrophil and macrophage infiltration. Moreover, autophagic flux was increased after melatonin administration while the apoptosis levels were decreased in the melatonin-pretreated mice. Using TAK-242 and CRX-527, we confirmed that MyD88-dependent TLR4 and MEK/ERK/mTORC1 signaling participated in melatonin-induced autophagy in IR mice. Collectively, our results provide novel evidence that antecedent melatonin treatment provides protection for the kidney against IR injury by enhancing autophagy, as regulated by the TLR4/MyD88/MEK/ERK/mTORC1 signaling pathway. Therefore, melatonin preconditioning offers a potential therapeutic approach to prevent renal IR injury related to various clinical conditions.


Subject(s)
Autophagy/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Kidney/blood supply , Mechanistic Target of Rapamycin Complex 1/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Myeloid Differentiation Factor 88/physiology , Reperfusion Injury/prevention & control , Toll-Like Receptor 4/physiology , Animals , Autophagy/physiology , Female , Inflammation/prevention & control , Melatonin/pharmacology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects
13.
Immunity ; 36(6): 1047-59, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749354

ABSTRACT

Dendritic cells (DCs), monocytes, and/or macrophages initiate host-protective immune responses to intracellular pathogens in part through interleukin-12 (IL-12) production, although the relative contribution of tissue resident versus recruited cells has been unclear. Here, we showed that after intraperitoneal infection with Toxoplasma gondii cysts, resident mononuclear phagocytes are replaced by circulating monocytes that differentiate in situ into inflammatory DCs (moDCs) and F4/80(+) macrophages. Importantly, NK cell-derived interferon-γ (IFN-γ) was required for both the loss of resident mononuclear phagocytes and the local differentiation of monocytes into macrophages and moDCs. This newly generated moDC population and not the resident DCs (or macrophages) served as the major source of IL-12 at the site of infection. Thus, NK cell-derived IFN-γ is important in both regulating inflammatory cell dynamics and in driving the local differentiation of monocytes into the cells required for initiating the immune response to an important intracellular pathogen.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Monocytes/immunology , Adoptive Transfer , Animals , Antigens, Ly/analysis , Cell Differentiation , Chemotaxis, Leukocyte , Dendritic Cells/pathology , Dendritic Cells/transplantation , Genes, Reporter , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/genetics , Killer Cells, Natural/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/chemistry , Monocytes/pathology , Monocytes/transplantation , Myeloid Differentiation Factor 88/physiology , Neutrophils/immunology , Peritonitis/immunology , Peritonitis/parasitology , Phagocytes/classification , Phagocytes/immunology , Phagocytes/pathology , Receptors, Interferon/deficiency , Receptors, Interferon/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology , Toxoplasmosis, Animal/immunology , Interferon gamma Receptor
14.
J Cell Mol Med ; 24(18): 10677-10692, 2020 09.
Article in English | MEDLINE | ID: mdl-32757377

ABSTRACT

Heart failure (HF) represents a major public health burden. Inflammation has been shown to be a critical factor in the progression of HF, regardless of the aetiology. Disappointingly, the majority of clinical trials targeting aspects of inflammation in patients with HF have been largely negative. Many clinical researches demonstrate that danshen has a good efficacy on HF, and however, whether danshen exerts anti-inflammatory effects against HF remains unclear. In our study, the employment of a water extracted and alcohol precipitated of danshen extract attenuated cardiac dysfunction and inflammation response in acute myocardial infarction-induced HF rats. Transcriptome technique and validation results revealed that TLR4 signalling pathway was involved in the anti-inflammation effects of danshen. In vitro, danshen reduced the release of inflammatory mediators in LPS-stimulated RAW264.7 macrophage cells. Besides, the LPS-stimulated macrophage conditioned media was applied to induce cardiac H9C2 cells injury, which could be attenuated by danshen. Furtherly, knock-down and overexpression of TLR4 were utilized to confirm that danshen ameliorated inflammatory injury via MyD88-dependent TLR4-TRAF6-NF-κB signalling pathway in cardiomyocytes. Furthermore, by utilizing co-immunoprecipitation, danshen was proved to suppress MD2/TLR4 complex formation and MyD88 recruitment. In conclusion, our results demonstrated that danshen ameliorates inflammatory injury by controlling MD2/TLR4-MyD88 complex formation and TLR4-TRAF6-NF-κB signalling pathway in acute myocardial infarction-induced HF.


Subject(s)
Heart Failure/drug therapy , Lymphocyte Antigen 96/antagonists & inhibitors , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myocardial Infarction/complications , Phytotherapy , Plant Extracts/therapeutic use , Salvia miltiorrhiza/chemistry , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Biomarkers , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical , Heart Failure/genetics , Heart Failure/prevention & control , Lymphocyte Antigen 96/physiology , Macrophages/metabolism , Mice , Multiprotein Complexes/drug effects , Myeloid Differentiation Factor 88/physiology , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Plant Extracts/isolation & purification , RAW 264.7 Cells , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Signal Transduction/genetics , Specific Pathogen-Free Organisms , Toll-Like Receptor 4/physiology , Transcriptome/drug effects , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/prevention & control
15.
Cancer Immunol Immunother ; 69(10): 2113-2124, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32448982

ABSTRACT

Metastatic cancer involving spread to the peritoneal cavity is referred to as peritoneal carcinomatosis and has a very poor prognosis. Our previous studies demonstrated a toll-like receptor 4 (TLR4) and C-type lectin receptor (CLR; Mincle/MCL) agonist pairing of monophosphoryl lipid A (MPL) and trehalose-6,6'-dicorynomycolate (TDCM) effectively inhibits peritoneal tumor growth and ascites development through a mechanism dependent upon B1a cell-produced natural IgM, complement, and phagocytes. In the current study, we investigated the requirement for TLR4 and Fc receptor common γ chain (FcRγ), required for Mincle/MCL signaling, in the MPL/TDCM-elicited response. MPL/TDCM significantly increased macrophages and Ly6Chi monocytes in the peritoneal cavity of both TLR4-/- and FcRγ-/- mice, suggesting redundancy in the signals required for monocyte/macrophage recruitment. However, B1 cell activation, antibody secreting cell differentiation, and tumor-reactive IgM production were defective in TLR4-/-, but not FcRγ-/- mice. TRIF was required for production of IgM reactive against tumor- and mucin-related antigens, but not phosphorylcholine, whereas TLR4 was required for production of both types of reactivities. Consistent with this, B1 cells lacking TLR4 or TRIF did not proliferate or differentiate into tumor-reactive IgM-producing cells in vitro and did not reconstitute MPL/TDCM-dependent protection against peritoneal carcinomatosis in CD19-/- mice. Our results indicate a TLR4/TRIF-dependent pathway is required by B1 cells for MPL/TDCM-elicited production of protective tumor-reactive natural IgM. The dependency on TRIF signaling for tumor-reactive, but not phosphorylcholine-reactive, IgM production reveals unexpected heterogeneity in TLR4-dependent regulation of natural IgM production, thereby highlighting important differences to consider when designing vaccines or therapies targeting these specificities.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , B-Lymphocyte Subsets/immunology , Cord Factors/administration & dosage , Immunoglobulin M/immunology , Lipid A/analogs & derivatives , Peritoneal Neoplasms/immunology , Toll-Like Receptor 4/physiology , Adjuvants, Immunologic/administration & dosage , Animals , B-Lymphocyte Subsets/drug effects , Lipid A/administration & dosage , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology
16.
PLoS Pathog ; 14(3): e1006925, 2018 03.
Article in English | MEDLINE | ID: mdl-29522566

ABSTRACT

Effector molecules translocated by the Salmonella pathogenicity island (SPI)1-encoded type 3 secretion system (T3SS) critically contribute to the pathogenesis of human Salmonella infection. They facilitate internalization by non-phagocytic enterocytes rendering the intestinal epithelium an entry site for infection. Their function in vivo has remained ill-defined due to the lack of a suitable animal model that allows visualization of intraepithelial Salmonella. Here, we took advantage of our novel neonatal mouse model and analyzed various bacterial mutants and reporter strains as well as gene deficient mice. Our results demonstrate the critical but redundant role of SopE2 and SipA for enterocyte invasion, prerequisite for transcriptional stimulation and mucosal translocation in vivo. In contrast, the generation of a replicative intraepithelial endosomal compartment required the cooperative action of SipA and SopE2 or SipA and SopB but was independent of SopA or host MyD88 signaling. Intraepithelial growth had no critical influence on systemic spread. Our results define the role of SPI1-T3SS effector molecules during enterocyte invasion and intraepithelial proliferation in vivo providing novel insight in the early course of Salmonella infection.


Subject(s)
Bacterial Proteins/metabolism , Enterocytes/microbiology , Intestinal Mucosa/microbiology , Myeloid Differentiation Factor 88/physiology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Type III Secretion Systems/metabolism , Animals , Bacterial Proteins/genetics , Cell Proliferation , Enterocytes/metabolism , Enterocytes/pathology , Genetic Complementation Test , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/metabolism , Signal Transduction , Type III Secretion Systems/genetics
17.
Nat Immunol ; 9(4): 361-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18297073

ABSTRACT

Toll-like receptor 4 (TLR4) induces two distinct signaling pathways controlled by the TIRAP-MyD88 and TRAM-TRIF pairs of adaptor proteins, which elicit the production of proinflammatory cytokines and type I interferons, respectively. How TLR4 coordinates the activation of these two pathways is unknown. Here we show that TLR4 activated these two signaling pathways sequentially in a process organized around endocytosis of the TLR4 complex. We propose that TLR4 first induces TIRAP-MyD88 signaling at the plasma membrane and is then endocytosed and activates TRAM-TRIF signaling from early endosomes. Our data emphasize a unifying theme in innate immune recognition whereby all type I interferon-inducing receptors signal from an intracellular location.


Subject(s)
Endocytosis/physiology , Interferon-beta/biosynthesis , Membrane Glycoproteins/physiology , Membrane Transport Proteins/physiology , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/physiology , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Cell Line , Down-Regulation/genetics , Down-Regulation/immunology , Endocytosis/genetics , Humans , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/physiology , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/physiology , Signal Transduction/genetics , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
18.
Blood ; 132(6): 559-564, 2018 08 09.
Article in English | MEDLINE | ID: mdl-29853538

ABSTRACT

Recent studies have revealed that the intestinal bacterial microbiome plays an important role in the regulation of hematopoiesis. A correlation between adverse hematologic effects and imbalance of the intestinal microbiome, or dysbiosis, is evident in several human conditions, such as inflammatory bowel disease, obesity, and, critically, in the setting of antibiotic exposure. Here we review the effects of gut dysbiosis on the hematological compartment and our current understanding of the mechanisms through which changes in the bacterial microbiome affect hematopoiesis.


Subject(s)
Dysbiosis/complications , Gastrointestinal Microbiome , Hematopoiesis , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Bone Marrow/physiology , Dysbiosis/microbiology , Dysbiosis/physiopathology , Gastrointestinal Microbiome/drug effects , Graft Survival , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/microbiology , Mice , Models, Immunological , Myeloid Differentiation Factor 88/physiology , Neutropenia/chemically induced , Nod1 Signaling Adaptor Protein/physiology , Nutrition Disorders/complications , Nutrition Disorders/microbiology , Signal Transduction , Specific Pathogen-Free Organisms , Toll-Like Receptors/physiology
19.
Gastroenterology ; 155(5): 1539-1552.e12, 2018 11.
Article in English | MEDLINE | ID: mdl-30063920

ABSTRACT

BACKGROUND & AIMS: Activating transcription factor 6 (ATF6) regulates endoplasmic reticulum stress. We studied whether ATF6 contributes to the development of colorectal cancer (CRC) using tissue from patients and transgenic mice. METHODS: We analyzed data from 541 patients with CRC in The Cancer Genome Atlas database for genetic variants and aberrant expression levels of unfolded protein response genes. Findings were validated in a cohort of 83 patients with CRC in Germany. We generated mice with intestinal epithelial cell-specific expression of the active form of Atf6 (nATF6IEC) from 2 alleles (homozygous), mice with expression of nATF6IEC from 1 allele (heterozygous), and nATF6IECfl/fl mice (controls). All nATF6IEC mice were housed under either specific-pathogen-free or germ-free conditions. Cecal microbiota from homozygous nATF6IEC mice or control mice was transferred into homozygous nATF6IEC mice or control mice. nATF6IEC mice were crossed with mice with disruptions in the myeloid differentiation primary response gene 88 and toll-like receptor adaptor molecule 1 gene (Myd88/Trif-knockout mice). Intestinal tissues were collected from mice and analyzed by histology, immunohistochemistry, immunoblots, gene expression profiling of unfolded protein response and inflammatory genes, array-based comparative genome hybridization, and 16S ribosomal RNA gene sequencing. RESULTS: Increased expression of ATF6 was associated with reduced disease-free survival times of patients with CRC. Homozygous nATF6IEC mice developed spontaneous colon adenomas at 12 weeks of age. Compared with controls, homozygous nATF6IEC mice had changes in the profile of their cecal microbiota, increased proliferation of intestinal epithelial cells, and loss of the mucus barrier-all preceding tumor formation. These mice had increased penetration of bacteria into the inner mucus layer and activation of signal transducer and activator of transcription 3, yet inflammation was not observed at the pretumor or tumor stages. Administration of antibiotics to homozygous nATF6IEC mice greatly reduced tumor incidence, and germ-free housing completely prevented tumorigenesis. Analysis of nATF6IEC MyD88/TRIF-knockout mice showed that tumor initiation and growth required MyD88/TRIF-dependent activation of signal transducer and activator of transcription 3. Transplantation of cecal microbiota from nATF6IEC mice and control mice, collected before tumor formation, caused tumor formation in ex-germ-free nATF6IEC mice. CONCLUSIONS: In patients with CRC, ATF6 was associated with reduced time of disease-free survival. In studies of nATF6IEC mice, we found sustained intestinal activation of ATF6 in the colon to promote dysbiosis and microbiota-dependent tumorigenesis.


Subject(s)
Activating Transcription Factor 6/physiology , Colorectal Neoplasms/etiology , Dysbiosis/etiology , Immunity, Innate , Intestines/microbiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Colorectal Neoplasms/mortality , Disease Progression , Humans , Mice , Myeloid Differentiation Factor 88/physiology , STAT3 Transcription Factor/physiology , Toll-Like Receptors/physiology , Unfolded Protein Response
20.
Immunity ; 33(3): 375-86, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20832340

ABSTRACT

Monocytes are effectors of the inflammatory response to microbes. Human CD14(+) monocytes specialize in phagocytosis and production of reactive oxygen species and secrete inflammatory cytokines in response to a broad range of microbial cues. Here, we have characterized the functions of human monocytes that lack CD14 (CD14(dim)) and express CD16. CD14(dim) monocytes were genetically distinct from natural killer cells. Gene expression analyses indicated similarities with murine patrolling Gr1(dim) monocytes, and they patrolled the endothelium of blood vessels after adoptive transfer, in a lymphocyte function-associated antigen-1-dependent manner. CD14(dim) monocytes were weak phagocytes and did not produce ROS or cytokines in response to cell-surface Toll-like receptors. Instead, they selectively produced TNF-α, IL-1ß, and CCL3 in response to viruses and immune complexes containing nucleic acids, via a proinflammatory TLR7-TLR 8-MyD88-MEK pathway. Thus, CD14(dim) cells are bona fide monocytes involved in the innate local surveillance of tissues and the pathogenesis of autoimmune diseases.


Subject(s)
Lipopolysaccharide Receptors/physiology , Monocytes/physiology , Nucleic Acids/physiology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/physiology , Viruses/immunology , Animals , Antigen Presentation , Cytokines/biosynthesis , GPI-Linked Proteins , HLA-DR Antigens/analysis , Humans , Lupus Erythematosus, Systemic/immunology , Mice , Myeloid Differentiation Factor 88/physiology , Reactive Oxygen Species/metabolism , Receptors, IgG/analysis
SELECTION OF CITATIONS
SEARCH DETAIL