Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(1): 54-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31819256

ABSTRACT

Ptpn6 is a cytoplasmic phosphatase that functions to prevent autoimmune and interleukin-1 (IL-1) receptor-dependent, caspase-1-independent inflammatory disease. Conditional deletion of Ptpn6 in neutrophils (Ptpn6∆PMN) is sufficient to initiate IL-1 receptor-dependent cutaneous inflammatory disease, but the source of IL-1 and the mechanisms behind IL-1 release remain unclear. Here, we investigate the mechanisms controlling IL-1α/ß release from neutrophils by inhibiting caspase-8-dependent apoptosis and Ripk1-Ripk3-Mlkl-regulated necroptosis. Loss of Ripk1 accelerated disease onset, whereas combined deletion of caspase-8 and either Ripk3 or Mlkl strongly protected Ptpn6∆PMN mice. Ptpn6∆PMN neutrophils displayed increased p38 mitogen-activated protein kinase-dependent Ripk1-independent IL-1 and tumor necrosis factor production, and were prone to cell death. Together, these data emphasize dual functions for Ptpn6 in the negative regulation of p38 mitogen-activated protein kinase activation to control tumor necrosis factor and IL-1α/ß expression, and in maintaining Ripk1 function to prevent caspase-8- and Ripk3-Mlkl-dependent cell death and concomitant IL-1α/ß release.


Subject(s)
Apoptosis/immunology , Caspase 8/immunology , Neutrophils/immunology , Protein Kinases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/genetics , Cells, Cultured , Gene Deletion , Inflammation/immunology , Interleukin-1/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Immunity ; 50(5): 1289-1304.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31079916

ABSTRACT

Pathogenic lymphocytes initiate the development of chronic inflammatory diseases. The cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) (encoded by Csf2) is a key communicator between pathogenic lymphocytes and tissue-invading inflammatory phagocytes. However, the molecular properties of GM-CSF-producing cells and the mode of Csf2 regulation in vivo remain unclear. To systematically study and manipulate GM-CSF+ cells and their progeny in vivo, we generated a fate-map and reporter of GM-CSF expression mouse strain (FROG). We mapped the phenotypic and functional profile of auto-aggressive T helper (Th) cells during neuroinflammation and identified the signature and pathogenic memory of a discrete encephalitogenic Th subset. These cells required interleukin-23 receptor (IL-23R) and IL-1R but not IL-6R signaling for their maintenance and pathogenicity. Specific ablation of this subset interrupted the inflammatory cascade, despite the unperturbed tissue accumulation of other Th subsets (e.g., Th1 and Th17), highlighting that GM-CSF expression not only marks pathogenic Th cells, but that this subset mediates immunopathology and tissue destruction.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Inflammation/genetics , Inflammation/pathology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR6/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
Immunity ; 50(2): 317-333.e6, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30683620

ABSTRACT

Interleukin-1 (IL-1) signaling is important for multiple potentially pathogenic processes in the central nervous system (CNS), but the cell-type-specific roles of IL-1 signaling are unclear. We used a genetic knockin reporter system in mice to track and reciprocally delete or express IL-1 receptor 1 (IL-1R1) in specific cell types, including endothelial cells, ventricular cells, peripheral myeloid cells, microglia, astrocytes, and neurons. We found that endothelial IL-1R1 was necessary and sufficient for mediating sickness behavior and drove leukocyte recruitment to the CNS and impaired neurogenesis, whereas ventricular IL-1R1 was critical for monocyte recruitment to the CNS. Although microglia did not express IL-1R1, IL-1 stimulation of endothelial cells led to the induction of IL-1 in microglia. Together, these findings describe the structure and functions of the brain's IL-1R1-expressing system and lay a foundation for the dissection and identification of IL-1R1 signaling pathways in the pathogenesis of CNS diseases.


Subject(s)
Brain/immunology , Neuroimmunomodulation/immunology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Animals , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Interleukin-1/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Neuroimmunomodulation/genetics , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction/genetics
4.
Immunity ; 43(6): 1125-36, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682985

ABSTRACT

The interleukin-1 receptor I (IL-1RI) is critical for host resistance to Mycobacterium tuberculosis (Mtb), yet the mechanisms of IL-1RI-mediated pathogen control remain unclear. Here, we show that without IL-1RI, Mtb-infected newly recruited Ly6G(hi) myeloid cells failed to upregulate tumor necrosis factor receptor I (TNF-RI) and to produce reactive oxygen species, resulting in compromised pathogen control. Furthermore, simultaneous ablation of IL-1RI and TNF-RI signaling on either stroma or hematopoietic cells led to early lethality, indicating non-redundant and synergistic roles of IL-1 and TNF in mediating macrophage-stroma cross-talk that was critical for optimal control of Mtb infection. Finally, we show that even in the presence of functional Mtb-specific adaptive immunity, the lack of IL-1α and not IL-1ß led to an exuberant intracellular pathogen replication and progressive non-resolving inflammation. Our study reveals functional interdependence between IL-1 and TNF in enabling Mtb control mechanisms that are critical for host survival.


Subject(s)
Interleukin-1alpha/immunology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis , Receptors, Interleukin-1 Type I/immunology
5.
Proc Natl Acad Sci U S A ; 116(26): 13067-13076, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182576

ABSTRACT

Neuroimmune interactions may contribute to severe pain and regional inflammatory and autonomic signs in complex regional pain syndrome (CRPS), a posttraumatic pain disorder. Here, we investigated peripheral and central immune mechanisms in a translational passive transfer trauma mouse model of CRPS. Small plantar skin-muscle incision was performed in female C57BL/6 mice treated daily with purified serum immunoglobulin G (IgG) from patients with longstanding CRPS or healthy volunteers followed by assessment of paw edema, hyperalgesia, inflammation, and central glial activation. CRPS IgG significantly increased and prolonged swelling and induced stable hyperalgesia of the incised paw compared with IgG from healthy controls. After a short-lasting paw inflammatory response in all groups, CRPS IgG-injected mice displayed sustained, profound microglia and astrocyte activation in the dorsal horn of the spinal cord and pain-related brain regions, indicating central sensitization. Genetic deletion of interleukin-1 (IL-1) using IL-1αß knockout (KO) mice and perioperative IL-1 receptor type 1 (IL-1R1) blockade with the drug anakinra, but not treatment with the glucocorticoid prednisolone, prevented these changes. Anakinra treatment also reversed the established sensitization phenotype when initiated 8 days after incision. Furthermore, with the generation of an IL-1ß floxed(fl/fl) mouse line, we demonstrated that CRPS IgG-induced changes are in part mediated by microglia-derived IL-1ß, suggesting that both peripheral and central inflammatory mechanisms contribute to the transferred disease phenotype. These results indicate that persistent CRPS is often contributed to by autoantibodies and highlight a potential therapeutic use for clinically licensed antagonists, such as anakinra, to prevent or treat CRPS via blocking IL-1 actions.


Subject(s)
Autoantibodies/immunology , Complex Regional Pain Syndromes/immunology , Immunoglobulin G/immunology , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Adult , Animals , Autoantibodies/administration & dosage , Autoantibodies/blood , Complex Regional Pain Syndromes/blood , Complex Regional Pain Syndromes/diagnosis , Complex Regional Pain Syndromes/drug therapy , Disease Models, Animal , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lower Extremity/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/pathology , Middle Aged , Pain Measurement , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/immunology , Receptors, Interleukin-1 Type I/metabolism , Spinal Cord Dorsal Horn/immunology , Spinal Cord Dorsal Horn/pathology
6.
Int J Mol Sci ; 23(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35163653

ABSTRACT

The interleukin-1 receptor type 1 (IL-1R1) holds pivotal roles in the immune system, as it is positioned at the "epicenter" of the inflammatory signaling networks. Increased levels of the cytokine IL-1 are a recognized feature of the immune response in the central nervous system (CNS) during injury and disease, i.e., neuroinflammation. Despite IL-1/IL-1R1 signaling within the CNS having been the subject of several studies, the roles of IL-1R1 in the CNS cellular milieu still cause controversy. Without much doubt, however, the persistent activation of the IL-1/IL-1R1 signaling pathway is intimately linked with the pathogenesis of a plethora of CNS disease states, ranging from Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS), all the way to schizophrenia and prion diseases. Importantly, a growing body of evidence is showing that blocking IL-1R1 signaling via pharmacological or genetic means in different experimental models of said CNS diseases leads to reduced neuroinflammation and delayed disease progression. The aim of this paper is to review the recent progress in the study of the biological roles of IL-1R1, as well as to highlight key aspects that render IL-1R1 a promising target for the development of novel disease-modifying treatments for multiple CNS indications.


Subject(s)
Central Nervous System Diseases/immunology , Neuroinflammatory Diseases/immunology , Receptors, Interleukin-1 Type I/immunology , Animals , Humans
7.
J Virol ; 94(12)2020 06 01.
Article in English | MEDLINE | ID: mdl-32295905

ABSTRACT

We demonstrate that female C57BL/6J mice are susceptible to a transient lower genital tract infection with MmuPV1 mouse papillomavirus and display focal histopathological abnormalities resembling those of human papillomavirus (HPV) infection. We took advantage of strains of genetically deficient mice to study in vivo the role of innate immune signaling in the control of papillomavirus. At 4 months, we sacrificed MmuPV1-infected mice and measured viral 757/3139 spliced transcripts by TaqMan reverse transcription-PCR (RT-PCR), localization of infection by RNAscope in situ hybridization, and histopathological abnormities by hematoxylin and eosin (H&E) staining. Among mice deficient in receptors for pathogen-associated molecular patterns, MyD88-/- and STING-/- mice had 1,350 and 80 copies of spliced transcripts/µg RNA, respectively, while no viral expression was detected in MAVS-/- and Ripk2-/- mice. Mice deficient in an adaptor molecule, STAT1-/-, for interferon signaling had 46,000 copies/µg RNA. Among mice with targeted deficiencies in the inflammatory response, interleukin-1 receptor knockout (IL-1R-/-) and caspase-1-/- mice had 350 and 30 copies/µg RNA, respectively. Among mice deficient in chemokine receptors, CCR6-/- mice had 120 copies/µg RNA, while CXCR2-/- and CXCR3-/- mice were negative. RNAscope confirmed focal infection in MyD88-/-, STAT1-/-, and CCR6-/- mice but was negative for other gene-deficient mice. Histological abnormalities were seen only in the latter mice. Our findings and the literature support a working model of innate immunity to papillomaviruses involving the activation of a MyD88-dependent pathway and IL-1 receptor signaling, control of viral replication by interferon-stimulated genes, and clearance of virus-transformed dysplastic cells by the action of the CCR6/CCL20 axis.IMPORTANCE Papillomaviruses infect stratified squamous epithelia, and the viral life cycle is linked to epithelial differentiation. Additionally, changes occur in viral and host gene expression, and immune cells are activated to modulate the infectious process. In vitro studies with keratinocytes cannot fully model the complex viral and host responses and do not reflect the contribution of local and migrating immune cells. We show that female C57BL/6J mice are susceptible to a transient papillomavirus cervicovaginal infection, and mice deficient in select genes involved in innate immune responses are susceptible to persistent infection with variable manifestations of histopathological abnormalities. The results of our studies support a working model of innate immunity to papillomaviruses, and the model provides a framework for more in-depth studies. A better understanding of mechanisms of early viral clearance and the development of approaches to induce clearance will be important for cancer prevention and the treatment of HPV-related diseases.


Subject(s)
Host-Pathogen Interactions/immunology , Myeloid Differentiation Factor 88/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , RNA, Messenger/immunology , RNA, Viral/immunology , Receptors, Interleukin-1 Type I/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Alternative Splicing , Animals , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/immunology , Cervix Uteri/immunology , Cervix Uteri/virology , Female , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Papillomaviridae/growth & development , Papillomaviridae/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , RNA, Messenger/genetics , RNA, Viral/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Receptors, CCR6/deficiency , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Signal Transduction , Vagina/immunology , Vagina/virology
8.
J Immunol ; 202(9): 2720-2727, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30910860

ABSTRACT

The aim of this study was to determine whether skin wounding induces monocyte (Mo) expansion in bone marrow and whether IL-1R1 signaling regulates this process. Our data show that skin wounding increases myeloid lineage-committed multipotent progenitors (MPP3 subset) and Mo in bone marrow, but this expansion is not impaired in Il1r1-/- mice. We also demonstrate that M-CSF-induced differentiation of myeloid progenitors into Mo is not impaired by the loss of IL-1R1 ex vivo, indicating that IL-R1 deficiency does not abrogate myeloid progenitor differentiation potential. In addition, we observed modestly delayed wound closure in Il1r1-/- mice associated with higher frequency of Ly6Clo Mo in the circulation at baseline and in wounds early after injury. Thus, in contrast to other models of inflammation that involve IL-1R1-dependent monopoiesis, our results demonstrate that skin wounding induces Mo progenitor and Mo expansion independently of IL-1R1 signaling.


Subject(s)
Bone Marrow/immunology , Monocytes/immunology , Receptors, Interleukin-1 Type I/deficiency , Skin/immunology , Wound Healing/immunology , Wounds and Injuries/immunology , Animals , Bone Marrow/pathology , Mice , Mice, Knockout , Monocytes/pathology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Skin/pathology , Wound Healing/genetics , Wounds and Injuries/genetics , Wounds and Injuries/pathology
9.
Infect Immun ; 88(10)2020 09 18.
Article in English | MEDLINE | ID: mdl-32719155

ABSTRACT

Group A Streptococcus (GAS) is the etiologic agent of numerous high-morbidity and high-mortality diseases. Infections are typically highly proinflammatory. During the invasive infection necrotizing fasciitis, this is in part due to the GAS protease SpeB directly activating interleukin-1ß (IL-1ß) independent of the canonical inflammasome pathway. The upper respiratory tract is the primary site for GAS colonization, infection, and transmission, but the host-pathogen interactions at this site are still largely unknown. We found that in the murine nasopharynx, SpeB enhanced IL-1ß-mediated inflammation and the chemotaxis of neutrophils. However, neutrophilic inflammation did not restrict infection and instead promoted GAS replication and disease. Inhibiting IL-1ß or depleting neutrophils, which both promote invasive infection, prevented GAS infection of the nasopharynx. Mice pretreated with penicillin became more susceptible to GAS challenge, and this reversed the attenuation from neutralization or depletion of IL-1ß, neutrophils, or SpeB. Collectively, our results suggest that SpeB is essential to activate an IL-1ß-driven neutrophil response. Unlike during invasive tissue infections, this is beneficial in the upper respiratory tract because it disrupts colonization resistance mediated by the microbiota. This provides experimental evidence that the notable inflammation of strep throat, which presents with significant swelling, pain, and neutrophil influx, is not an ineffectual immune response but rather is a GAS-directed remodeling of this niche for its pathogenic benefit.


Subject(s)
Nasopharynx/immunology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/pathogenicity , Animals , Anti-Bacterial Agents/adverse effects , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Caspase 1/genetics , Caspase 1/immunology , Chemotaxis, Leukocyte , Exotoxins/genetics , Exotoxins/immunology , Inflammation , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/immunology , Mice , Nasopharynx/microbiology , Neutrophils/immunology , Pharyngitis/genetics , Pharyngitis/immunology , Pharyngitis/microbiology , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Streptococcal Infections/genetics , Streptococcal Infections/microbiology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/genetics , Streptococcus pyogenes/growth & development , Virulence/drug effects , Virulence/genetics
10.
J Immunol ; 201(10): 2873-2878, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30305325

ABSTRACT

Sepsis disproportionately affects the very old and the very young. IL-1 signaling is important in innate host defense but may also play a deleterious role in acute inflammatory conditions (including sepsis) by promulgating life-threatening inflammation. IL-1 signaling is mediated by two distinct ligands: IL-1α and IL-1ß, both acting on a common receptor (IL-1R1). IL-1R1 targeting has not reduced adult human sepsis mortality despite biologic plausibility. Because the specific role of IL-1α or IL-1ß in sepsis survival is unknown in any age group and the role of IL-1 signaling remains unknown in neonates, we studied the role of IL-1 signaling, including the impact of IL-1α and IL-1ß, on neonatal murine sepsis survival. IL-1 signaling augments the late plasma inflammatory response to sepsis. IL-1α and not IL-1ß is the critical mediator of sepsis mortality, likely because of paracrine actions within the tissue. These data do not support targeting IL-1 signaling in neonates.


Subject(s)
Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Receptors, Interleukin-1 Type I/immunology , Sepsis/immunology , Animals , Animals, Newborn , Female , Humans , Infant, Newborn , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Sepsis/mortality , Signal Transduction/immunology
11.
J Immunol ; 201(6): 1645-1650, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30068597

ABSTRACT

IL-1R1 deficiency in mice causes severe susceptibility to Mycobacterium tuberculosis Mice and macrophage cultures lacking IL-1R1 display increased bacterial growth, suggesting that phagocytes may require IL-1R1-dependent antimicrobial signals to limit intracellular M. tuberculosis replication directly. However, the myeloid-cell-intrinsic versus -extrinsic requirements for IL-1R1 to control M. tuberculosis infection in mice have not been directly addressed. Using single-cell analysis of infected cells, competitive mixed bone marrow chimeras, and IL-1R1 conditional mutant mice, we show in this article that IL-1R1 expression by pulmonary phagocytes is uncoupled from their ability to control intracellular M. tuberculosis growth. Importantly, IL-1R1-dependent control was provided to infected cells in trans by both nonhematopoietic and hematopoietic cells. Thus, IL-1R1-mediated host resistance to M. tuberculosis infection does not involve mechanisms of cell-autonomous antimicrobicidal effector functions in phagocytes but requires the cooperation between infected cells and other cells of hematopoietic or nonhematopoietic origin to promote bacterial containment and control of infection.


Subject(s)
Immunity, Innate , Lung/immunology , Mycobacterium tuberculosis/immunology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Tuberculosis, Pulmonary/immunology , Animals , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/genetics , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/pathology
12.
J Neuroinflammation ; 16(1): 118, 2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31170999

ABSTRACT

BACKGROUND: Microglia and inflammation have context-specific impacts upon neuronal survival in different models of central nervous system (CNS) disease. Herein, we investigate how inflammatory mediators, including microglia, interleukin 1 beta (IL1ß), and signaling through interleukin 1 receptor type 1 (IL-1R1), influence the survival of retinal neurons in response to excitotoxic damage. METHODS: Excitotoxic retinal damage was induced via intraocular injections of NMDA. Microglial phenotype and neuronal survival were assessed by immunohistochemistry. Single-cell RNA sequencing was performed to obtain transcriptomic profiles. Microglia were ablated by using clodronate liposome or PLX5622. Retinas were treated with IL1ß prior to NMDA damage and cell death was assessed in wild type, IL-1R1 null mice, and mice expressing IL-1R1 only in astrocytes. RESULTS: NMDA-induced damage included neuronal cell death, microglial reactivity, upregulation of pro-inflammatory cytokines, and genes associated with IL1ß-signaling in different types of retinal neurons and glia. Expression of the IL1ß receptor, IL-1R1, was evident in astrocytes, endothelial cells, some Müller glia, and OFF bipolar cells. Ablation of microglia with clodronate liposomes or Csf1r antagonist (PLX5622) resulted in elevated cell death and diminished neuronal survival in excitotoxin-damaged retinas. Exogenous IL1ß stimulated the proliferation and reactivity of microglia in the absence of damage, reduced numbers of dying cells in damaged retinas, and increased neuronal survival following an insult. IL1ß failed to provide neuroprotection in the IL-1R1-null retina, but IL1ß-mediated neuroprotection was rescued when expression of IL-1R1 was restored in astrocytes. CONCLUSIONS: We conclude that reactive microglia provide protection to retinal neurons, since the absence of microglia is detrimental to survival. We propose that, at least in part, the survival-influencing effects of microglia may be mediated by IL1ß, IL-1R1, and interactions of microglia and other macroglia.


Subject(s)
Interleukin-1beta/metabolism , Microglia/metabolism , Neuroprotection/physiology , Receptors, Interleukin-1 Type I/metabolism , Retina/pathology , Animals , Excitatory Amino Acid Agonists/toxicity , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , N-Methylaspartate/toxicity , Neurotoxins/toxicity , Receptors, Interleukin-1 Type I/immunology , Retina/immunology
13.
J Immunol ; 199(3): 1122-1130, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28674179

ABSTRACT

CD4+ Th cells play crucial roles in orchestrating immune responses against pathogenic microbes, after differentiating into effector subsets. Recent research has revealed the importance of IFN-γ and IL-17 double-producing CD4+ Th cells, termed Th17/Th1 cells, in the induction of autoimmune and inflammatory diseases. In addition, Th17/Th1 cells are involved in the regulation of infection caused by the intracellular bacterium Mycobacterium tuberculosis in humans. However, the precise mechanism of Th17/Th1 induction during pathogen infection is unclear. In this study, we showed that the inflammasome and Fas-dependent IL-1ß induces Th17/Th1 cells in mice, in response to infection with the pathogenic intracellular bacterium Listeria monocytogenes In the spleens of infected wild-type mice, Th17/Th1 cells were induced, and expressed T-bet and Rorγt. In Pycard-/- mice, which lack the adaptor molecule of the inflammasome (apoptosis-associated speck-like protein containing a caspase recruitment domain), Th17/Th1 induction was abolished. In addition, the Fas-mediated IL-1ß production was required for Th17/Th1 induction during bacterial infection: Th17/Th1 induction was abolished in Fas-/- mice, whereas supplementation with recombinant IL-1ß restored Th17/Th1 induction via IL-1 receptor 1 (IL-1R1), and rescued the mortality of Fas-/- mice infected with Listeria IL-1R1, but not apoptosis-associated speck-like protein containing a caspase recruitment domain or Fas on T cells, was required for Th17/Th1 induction, indicating that IL-1ß stimulates IL-1R1 on T cells for Th17/Th1 induction. These results indicate that IL-1ß, produced by the inflammasome and Fas-dependent mechanisms, contributes cooperatively to the Th17/Th1 induction during bacterial infection. This study provides a deeper understanding of the molecular mechanisms underlying Th17/Th1 induction during pathogenic microbial infections in vivo.


Subject(s)
Inflammasomes/immunology , Interleukin-1beta/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , fas Receptor/metabolism , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Cell Differentiation , Interleukin-1beta/administration & dosage , Listeria monocytogenes/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology , Spleen/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , fas Receptor/deficiency , fas Receptor/genetics
14.
J Hepatol ; 68(5): 986-995, 2018 05.
Article in English | MEDLINE | ID: mdl-29366909

ABSTRACT

BACKGROUND & AIMS: Interleukin (IL)-1-type cytokines including IL-1α, IL-1ß and interleukin-1 receptor antagonist (IL-1Ra) are among the most potent molecules of the innate immune system and exert biological activities through the ubiquitously expressed interleukin-1 receptor type 1 (IL-1R1). The role of IL-1R1 in hepatocytes during acute liver failure (ALF) remains undetermined. METHODS: The role of IL-1R1 during ALF was investigated using a novel transgenic mouse model exhibiting deletion of all signaling-capable IL-1R isoforms in hepatocytes (Il1r1Hep-/-). RESULTS: ALF induced by D-galactosamine (D-GalN) and lipopolysaccharide (LPS) was significantly attenuated in Il1r1Hep-/- mice leading to reduced mortality. Conditional deletion of Il1r1 decreased activation of injurious c-Jun N-terminal kinases (JNK)/c-Jun signaling, activated nuclear factor-kappa B (NF-κB) p65, inhibited extracellular signal-regulated kinase (ERK) and prevented caspase 3-mediated apoptosis. Moreover, Il1r1Hep-/- mice exhibited reduced local and systemic inflammatory cytokine and chemokine levels, especially TNF-α, IL-1α/ß, IL-6, CC-chemokine ligand 2 (CCL2), C-X-C motif ligand 1 (CXCL-1) and CXCL-2, and a reduced neutrophil recruitment into the hepatic tissue in response to injury. NLRP3 inflammasome expression and caspase 1 activation were suppressed in the absence of the hepatocellular IL-1R1. Inhibition of IL-1R1 using IL-1ra (anakinra) attenuated the severity of liver injury, while IL-1α administration exaggerated it. These effects were lost ex vivo and at later time points, supporting a role of IL-1R1 in inflammatory signal amplification during acute liver injury. CONCLUSION: IL-1R1 in hepatocytes plays a pivotal role in an IL-1-driven auto-amplification of cell death and inflammation in the onset of ALF. LAY SUMMARY: Acute liver injury which can cause lethal liver failure is medicated by a class of proteins called cytokines. Among these, interleukin-1 (IL-1) and the corresponding receptor IL-1R1 play a prominent role in the immune system, but their role in the liver is undetermined. In the current study, a novel mouse model with defective IL-1R1 in liver cells was studied. Mice lacking this receptor in liver cells were protected from cell death to a certain extent. This protection occurred only in the presence of other, neighboring cells, arguing for the involvement of proteins derived from these cells. This effect is called paracrine signaling and the current study has for the first time shown that the IL-1R1 receptor on hepatocytes is involved in acute liver failure in this context. The approved drug anakinra - which blocks IL-1R1 - had the same effect, supporting the proposed mechanism of action. The findings of this study suggest new treatment options for patients with acute liver failure by blocking defined signals of the immune system.


Subject(s)
Hepatocytes/immunology , Interleukin-1/immunology , Liver Failure, Acute/immunology , Liver Failure, Acute/prevention & control , Receptors, Interleukin-1 Type I/deficiency , Animals , Caspases/metabolism , Chemokines/immunology , Cytokines/immunology , Disease Models, Animal , Hepatocytes/metabolism , Hepatocytes/pathology , Inflammation/immunology , Inflammation/prevention & control , Interleukin 1 Receptor Antagonist Protein/pharmacology , Liver Failure, Acute/pathology , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Transcription Factor RelA/immunology
15.
COPD ; 15(2): 99-107, 2018 04.
Article in English | MEDLINE | ID: mdl-29683730

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease associated with significant morbidity and mortality. Current diagnostic criteria based on the presence of fixed airflow obstruction and symptoms do not integrate the complex pathological changes occurring within the lung and they do not define different airway inflammatory patterns. The current management of COPD is based on 'one size fits all' approach and does not take the importance of heterogeneity in COPD population into account. The available treatments aim to alleviate symptoms and reduce exacerbation frequency but do not alter the course of the disease. Recent advances in molecular biology have furthered our understanding of inflammatory pathways in pathogenesis of COPD and have led to development of targeted therapies (biologics and small molecules) based on predefined biomarkers. Herein we shall review the trials of biologics in COPD and potential future drug developments in the field.


Subject(s)
Biological Products/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Airway Remodeling/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Eosinophils/immunology , Etanercept/therapeutic use , Humans , Infliximab/therapeutic use , Interleukin-1/antagonists & inhibitors , Interleukin-1/immunology , Interleukin-13/antagonists & inhibitors , Interleukin-13/immunology , Interleukin-5/antagonists & inhibitors , Interleukin-5/immunology , Interleukin-8/antagonists & inhibitors , Interleukin-8/immunology , Molecular Targeted Therapy , Pulmonary Disease, Chronic Obstructive/immunology , Pyrimidines/therapeutic use , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/immunology , Receptors, Interleukin-5/antagonists & inhibitors , Receptors, Interleukin-5/immunology , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/immunology , Sulfonamides/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
16.
Eur J Immunol ; 46(4): 912-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26692072

ABSTRACT

IL-1 is a key cytokine known to drive chronic inflammation and to regulate many physiological, immunological, and neuroimmunological responses via actions on diverse cell types of the body. To determine the mechanisms of IL-1 actions as part of the inflammatory response in vivo, we generated a conditional IL-1 receptor 1 (IL-1R1) mouse mutant using the Cre/LoxP system (IL-1R1(fl/fl) ). In the mutant generated, exon 5, which encodes part of the extracellular-binding region of the receptor, is flanked by LoxP sites, thereby inactivating the two previously described functional IL-1R1 gene transcripts after Cre-mediated recombination. Using keratin 14-Cre driver mice, new IL-1R1 deficient (-/-) mice were subsequently generated, in which all signaling IL-1 receptor isoforms are deleted ubiquitously. Furthermore, using vav-iCre driver mice, we deleted IL-1 receptor isoforms in the hematopoietic system. In these mice, we show that both the IL-17 and IL-22 cytokine response is reduced, when mice are challenged by the helminth Trichuris muris. We are currently crossing IL-1R1(fl/fl) mice with different Cre-expressing mice in order to study mechanisms of acute and chronic inflammatory diseases.


Subject(s)
Inflammation/immunology , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Receptors, Interleukin-1 Type I/genetics , Trichuris/immunology , Animals , Interleukin-17/immunology , Interleukins/immunology , Keratin-14/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1 Type I/immunology , Interleukin-22
17.
Eur J Immunol ; 46(11): 2531-2541, 2016 11.
Article in English | MEDLINE | ID: mdl-27569535

ABSTRACT

Allergic asthma is characterized by a strong Th2 response with inflammatory cell recruitment and structural changes in the lung. Papain is a protease allergen disrupting the airway epithelium triggering a rapid inflammation with eosinophilia mediated by innate lymphoid cell activation (ILC2) and leading to a Th2 immune response. In this study, we focused on inflammatory responses to a single exposure to papain and showed that intranasal administration of papain results in the recruitment of inflammatory cells, including neutrophils and eosinophils with a rapid production of IL-1α, IL-1ß, and IL-33. The inflammatory response is abrogated in the absence of IL-1R1 and MyD88. To decipher the cell type(s) involved in MyD88-dependent IL-1R1/MyD88 signaling, we used new cell-specific MyD88-deficient mice and found that the deletion of MyD88 signaling in single cell types such as T cells, epithelial cells, CD11c-positive or myeloid cells leads to only a partial inhibition compared to complete absence of MyD88, suggesting that several cell types contribute to the response. Importantly, the inflammatory response is largely ST2 and IL-36R independent. In conclusion, IL-1R1 signaling via MyD88 is critical for the first step of inflammatory response to papain.


Subject(s)
Allergens/immunology , Immunity, Innate , Lung/immunology , Myeloid Differentiation Factor 88/metabolism , Papain/immunology , Pneumonia/immunology , Receptors, Interleukin-1 Type I/metabolism , Allergens/administration & dosage , Animals , Eosinophils/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-33/metabolism , Lung/physiopathology , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neutrophils/immunology , Papain/administration & dosage , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1 Type I/immunology , Signal Transduction , Th2 Cells/immunology
18.
Addict Biol ; 22(5): 1279-1288, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27273552

ABSTRACT

Proinflammatory activity has been postulated to play a role in addictive processes and stress responses, but the underlying mechanisms remain largely unknown. Here, we examined the role of interleukin 1 (IL-1) and tumor necrosis factor-α (TNF-α) in regulation of voluntary alcohol consumption, alcohol reward and stress-induced drinking. Mice with a deletion of the IL-1 receptor I gene (IL-1RI KO) exhibited modestly decreased alcohol consumption. However, IL-1RI deletion affected neither the rewarding properties of alcohol, measured by conditioned place preference (CPP), nor stress-induced drinking induced by social defeat stress. TNF-α signaling can compensate for phenotypic consequences of IL1-RI deletion. We therefore hypothesized that double deletion of both IL-1RI and TNF-1 receptors (TNF-1R) may reveal the role of these pathways in regulation of alcohol intake. Double KOs consumed significantly less alcohol than control mice over a range of alcohol concentrations. The combined deletion of TNF-1R and IL-1RI did not influence alcohol reward, but did prevent increased alcohol consumption resulting from exposure to repeated bouts of social defeat stress. Taken together, these data indicate that IL-1RI and TNF-1R contribute to regulation of stress-induced, negatively reinforced drinking perhaps through overlapping signaling events downstream of these receptors, while leaving rewarding properties of alcohol largely unaffected.


Subject(s)
Alcohol Drinking/immunology , Behavior, Animal , Interleukin-1/immunology , Receptors, Interleukin-1 Type I/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Stress, Psychological/immunology , Tumor Necrosis Factor-alpha/immunology , Alcohol Drinking/genetics , Animals , Central Nervous System Depressants/administration & dosage , Conditioning, Classical , Ethanol/administration & dosage , Inflammation , Male , Mice , Mice, Knockout , Psychological Distance , Receptors, Interleukin-1 Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction , Stress, Psychological/genetics
19.
J Allergy Clin Immunol ; 138(5): 1367-1380.e5, 2016 11.
Article in English | MEDLINE | ID: mdl-27233150

ABSTRACT

BACKGROUND: Eosinophilic esophagitis (EoE) is an allergic inflammatory disorder characterized by accumulation of eosinophils in the esophagus. EoE often coexists with atopic dermatitis, a chronic inflammatory skin disease. The impaired skin barrier in patients with atopic dermatitis has been suggested as an entry point for allergic sensitization that triggers development of EoE. OBJECTIVE: We sought to define the mechanisms whereby epicutaneous sensitization through a disrupted skin barrier induces development of EoE. METHODS: To elicit experimental EoE, mice were epicutaneously sensitized with ovalbumin (OVA), followed by intranasal OVA challenge. Levels of esophageal mRNA for TH2 cytokines and the IL-33 receptor Il1rl1 (St2) were measured by using quantitative PCR. Esophageal eosinophil accumulation was assessed by using flow cytometry and hematoxylin and eosin staining. In vivo basophil depletion was achieved with diphtheria toxin treatment of Mcpt8DTR mice, and animals were repopulated with bone marrow basophils. mRNA analysis of esophageal biopsy specimens from patients with EoE was used to validate our findings in human subjects. RESULTS: Epicutaneous sensitization and intranasal challenge of wild-type mice resulted in accumulation of eosinophils and upregulation of TH2 cytokines and St2 in the esophagus. Disruption of the IL-33-ST2 axis or depletion of basophils reduced these features. Expression of ST2 on basophils was required to accumulate in the esophagus and transfer experimental EoE. Expression of IL1RL1/ST2 mRNA was increased in esophageal biopsy specimens from patients with EoE. Topical OVA application on unstripped skin induced experimental EoE in filaggrin-deficient flaky tail (ft/ft) mice but not in wild-type control or ft/ft.St2-/- mice. CONCLUSION: Epicutaneous allergic sensitization promotes EoE, and this is critically mediated through the IL-33-ST2-basophil axis.


Subject(s)
Basophils/immunology , Dermatitis, Atopic/immunology , Eosinophilic Esophagitis/immunology , Interleukin-33/immunology , Adolescent , Allergens/immunology , Animals , Child , Child, Preschool , Esophagus/immunology , Female , Filaggrin Proteins , Humans , Male , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , RNA, Messenger/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology
20.
Eur J Immunol ; 45(10): 2847-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26147228

ABSTRACT

The pathogenesis of inflammatory skin diseases such as psoriasis involves the release of numerous proinflammatory cytokines, including members of the IL-1 family. Here we report overexpression of IL-1α, IL-1ß, and IL-1 receptor antagonist mRNA, associated to expression of IL-23p19, IL-17A, and IL-22 in skin cells, upon topical application of the TLR7 agonist imiquimod (IMQ) in C57BL/6J mice. IMQ-induced skin inflammation was partially reduced in mice deficient for both IL-1α/IL-1ß or for IL-1 receptor type 1 (IL-1R1), but not in IL-1α- or IL-1ß-deficient mice, demonstrating the redundant activity of IL-1α and IL-1ß for skin inflammation. NLRP3 or apoptosis-associated Speck-like protein containing a Caspase recruitment domain-deficient mice had no significant reduction of skin inflammation in response to IMQ treatment, mainly due to the redundancy of IL-1α. However, IMQ-induced skin inflammation was abolished in the absence of MyD88, the adaptor protein shared by IL-1R and TLR signaling pathways. These results are consistent with the TLR7 dependence of IMQ-induced skin inflammation. Thus, IL-1R1 contributes to the IMQ-induced skin inflammation, and disruption of MyD88 signaling completely abrogates this response.


Subject(s)
Adjuvants, Immunologic/adverse effects , Aminoquinolines/adverse effects , Carrier Proteins/immunology , Drug Eruptions/immunology , Inflammasomes/immunology , Myeloid Differentiation Factor 88/immunology , Receptors, Interleukin-1 Type I/immunology , Signal Transduction/immunology , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Animals , Carrier Proteins/genetics , Cytokines/genetics , Cytokines/immunology , Drug Eruptions/genetics , Drug Eruptions/pathology , Imiquimod , Inflammasomes/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/immunology , Skin/pathology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL