Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 498
Filter
1.
J Interferon Cytokine Res ; 42(2): 49-61, 2022 02.
Article in English | MEDLINE | ID: mdl-35171706

ABSTRACT

The interleukin (IL)-1 superfamily of cytokines comprises 11 pro- and anti-inflammatory cytokines, which play essential roles during the immune response. Several pathogenic pathways are initiated by IL-1RL2 (interleukin 1 receptor-like 2) signaling, also known as IL-36R, in the skin, lungs, and gut. IL-36 cytokines promote the secretion of proinflammatory cytokines and chemokines, upregulation of antimicrobial peptides, proliferation mediators, and adhesion molecules on endothelial cells. In addition, the IL-36-IL-1RL2 axis has an essential role against viral infections, including a potential role in COVID-19 pathology. The evidence presented in this review highlights the importance of the axis IL-36-IL-1RL2 in the development of several inflammation-related diseases and the healing process. It suggests that IL-1RL2 ligands have specific roles depending on the tissue or cell source. However, there is still much to discover about this cytokine family, their functions in other organs, and how they accomplish a dual effect in inflammation and healing.


Subject(s)
Inflammation/physiopathology , Receptors, Interleukin-1/physiology , Animals , COVID-19/physiopathology , Cytokine Release Syndrome/physiopathology , Cytokines/physiology , Host-Pathogen Interactions , Humans , Interleukin-1/physiology , Interleukins/classification , Intestines/metabolism , Intestines/pathology , Ligands , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System , Mice , NF-kappa B/metabolism , Protein Domains , Receptors, Interleukin/classification , Receptors, Interleukin-1/agonists , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/chemistry , SARS-CoV-2 , Signal Transduction , Skin/metabolism , Skin/pathology
3.
Front Immunol ; 12: 675294, 2021.
Article in English | MEDLINE | ID: mdl-34322116

ABSTRACT

Aspergillus fumigatus airway infections are associated with increased rates of hospitalizations and declining lung function in patients with chronic lung disease. While the pathogenesis of invasive A. fumigatus infections is well studied, little is known about the development and progression of airway infections. Previous studies have demonstrated a critical role for the IL-1 cytokines, IL-1α and IL-1ß in enhancing pulmonary neutrophil recruitment during invasive aspergillosis. Here we use a mouse model of A. fumigatus airway infection to study the role of these IL-1 cytokines in immunocompetent mice. In the absence of IL-1 receptor signaling, mice exhibited reduced numbers of viable pulmonary neutrophils and increased levels of neutrophil apoptosis during fungal airway infection. Impaired neutrophil viability in these mice was associated with reduced pulmonary and systemic levels of G-CSF, and treatment with G-CSF restored both neutrophil viability and resistance to A. fumigatus airway infection. Taken together, these data demonstrate that IL-1 dependent G-CSF production plays a key role for host resistance to A. fumigatus airway infection through suppressing neutrophil apoptosis at the site of infection.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/pathogenicity , Lung/immunology , Neutrophils/physiology , Pulmonary Aspergillosis/immunology , Receptors, Interleukin-1/physiology , Animals , Apoptosis/immunology , Chemokines/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Interleukin-1alpha , Interleukin-1beta , Lung/pathology , Macrophages , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Neutrophils/immunology
4.
Invest Ophthalmol Vis Sci ; 62(6): 10, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33970198

ABSTRACT

Purpose: Interleukin (IL)-36 cytokines have been shown to play either beneficial or detrimental roles in the infection of mucosal tissues in a pathogen-dependent manner, but their involvement in fungal keratitis remains elusive. We herein investigated their expression and function in mediating corneal innate immunity against Candida albicans infection. Methods: Gene expression in mouse corneas with or without C. albicans infection was determined by regular RT- and real-time (q)-PCR, Western blot analysis, ELISA or proteome profile assay. The severity of C. albicans keratitis was assessed using clinical scoring, bacterial counting, and myeloperoxidase (MPO) activity as an indicator of neutrophil infiltration. IL36R knockout mice and IL-33-specific siRNA were used to assess the involvement IL-33 signaling in C. albicans-infected corneas. B6 CD11c-DTR mice and clodronate liposomes were used to define the involvement of dendritic cells (DCs) and macrophages in IL-36R signaling and C. albicans keratitis, respectively. Results: IL-36γ were up-regulated in C57BL6 mouse corneas in response to C. albicans infection. IL-36 receptor-deficient mice display increased severity of keratitis, with a higher fungal load, MPO, and IL-1ß levels, and lower soluble sIL-1Ra and calprotectin levels. Exogenous IL-36γ prevented fungal keratitis pathogenesis with lower fungal load and MPO activity, higher expression of sIL-1Ra and calprotectin, and lower expression of IL-1ß, at mRNA or protein levels. Protein array analysis revealed that the expression of IL-33 and REG3G were related to IL-36/IL36R signaling, and siRNA downregulation of IL-33 increased the severity of C. albicans keratitis. Depletion of dendritic cells or macrophages resulted in severe C. albicans keratitis and yet exhibited minimal effects on exogenous IL-36γ-induced protection against C. albicans infection in B6 mouse corneas. Conclusions: IL-36/IL36R signaling plays a protective role in fungal keratitis by promoting AMP expression and by suppressing fungal infection-induced expression of proinflammatory cytokines in a dendritic cell- and macrophage-independent manner.


Subject(s)
Corneal Ulcer/prevention & control , Eye Infections, Fungal/prevention & control , Immunity, Innate/physiology , Interleukin-1/physiology , Keratitis/prevention & control , Receptors, Interleukin-1/physiology , Signal Transduction/physiology , Animals , Blotting, Western , Candida albicans , Corneal Ulcer/immunology , Corneal Ulcer/microbiology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Eye Infections, Fungal/immunology , Eye Infections, Fungal/microbiology , Gene Expression Regulation/physiology , Keratitis/immunology , Keratitis/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction
5.
Arterioscler Thromb Vasc Biol ; 40(6): 1533-1542, 2020 06.
Article in English | MEDLINE | ID: mdl-32268786

ABSTRACT

OBJECTIVE: Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS: These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.


Subject(s)
Clopidogrel/pharmacokinetics , Clopidogrel/therapeutic use , Drug Resistance , Obesity/complications , Receptors, Interleukin-1/physiology , Animals , Carotid Artery Thrombosis/prevention & control , Clopidogrel/pharmacology , Cytochrome P-450 Enzyme System/genetics , Diabetes Mellitus , Diet, High-Fat , Disease Models, Animal , Fibrinolytic Agents , Gene Expression , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/enzymology , Obesity/etiology , Obesity/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Receptors, Interleukin-1/deficiency
6.
J Leukoc Biol ; 106(1): 147-160, 2019 07.
Article in English | MEDLINE | ID: mdl-30900780

ABSTRACT

TLRs are a class of pattern recognition receptors (PRRs) that detect invading microbes by recognizing pathogen-associated molecular patterns (PAMPs). Upon PAMP engagement, TLRs activate a signaling cascade that leads to the production of inflammatory mediators. The localization of TLRs, either on the plasma membrane or in the endolysosomal compartment, has been considered to be a fundamental aspect to determine to which ligands the receptors bind, and which transduction pathways are induced. However, new observations have challenged this view by identifying complex trafficking events that occur upon TLR-ligand binding. These findings have highlighted the central role that endocytosis and receptor trafficking play in the regulation of the innate immune response. Here, we review the TLR4 and TLR9 transduction pathways and the importance of their different subcellular localization during the inflammatory response. Finally, we discuss the implications of TLR9 subcellular localization in autoimmunity.


Subject(s)
Toll-Like Receptor 4/physiology , Toll-Like Receptor 9/physiology , Animals , Autoimmune Diseases/immunology , Cytokines/biosynthesis , DNA-Binding Proteins/physiology , Endocytosis , Granulins/physiology , HMGB1 Protein/physiology , Humans , Membrane Glycoproteins/physiology , Receptors, Interleukin-1/physiology , Signal Transduction , Transcription Factors/physiology
7.
Sci Rep ; 7(1): 13829, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062042

ABSTRACT

Previously we reported that corneal epithelial barrier function against Pseudomonas aeruginosa was MyD88-dependent. Here, we explored contributions of MyD88-dependent receptors using vital mouse eyes and confocal imaging. Uninjured IL-1R (-/-) or TLR4 (-/-) corneas, but not TLR2 (-/-), TLR5 (-/-), TLR7 (-/-), or TLR9 (-/-), were more susceptible to P. aeruginosa adhesion than wild-type (3.8-fold, 3.6-fold respectively). Bacteria adherent to the corneas of IL-1R (-/-) or TLR5 (-/-) mice penetrated beyond the epithelial surface only if the cornea was superficially-injured. Bone marrow chimeras showed that bone marrow-derived cells contributed to IL-1R-dependent barrier function. In vivo, but not ex vivo, stromal CD11c+ cells responded to bacterial challenge even when corneas were uninjured. These cells extended processes toward the epithelial surface, and co-localized with adherent bacteria in superficially-injured corneas. While CD11c+ cell depletion reduced IL-6, IL-1ß, CXCL1, CXCL2 and CXCL10 transcriptional responses to bacteria, and increased susceptibility to bacterial adhesion (>3-fold), the epithelium remained resistant to bacterial penetration. IL-1R (-/-) corneas also showed down-regulation of IL-6 and CXCL1 genes with and without bacterial challenge. These data show complex roles for TLR4, TLR5, IL-1R and CD11c+ cells in constitutive epithelial barrier function against P. aeruginosa, with details dependent upon in vivo conditions.


Subject(s)
CD11c Antigen/immunology , Cell Membrane Permeability , Epithelium, Corneal/immunology , Gene Expression Regulation , Myeloid Differentiation Factor 88/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Bacterial Adhesion , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/microbiology , CD11c Antigen/metabolism , Cells, Cultured , Epithelium, Corneal/metabolism , Epithelium, Corneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Receptors, Interleukin-1/physiology , Signal Transduction , Toll-Like Receptor 4/physiology , Toll-Like Receptor 5/physiology
8.
Braz. j. infect. dis ; 21(4): 418-423, July-Aug. 2017. tab
Article in English | LILACS | ID: biblio-888896

ABSTRACT

Abstract Introduction: The present study was designed to investigate the association between rs8177374 polymorphism and malaria symptoms due to exposure of Plasmodium vivax and Plasmodium falciparum. Materials and methods: A total of 454 samples were included in the study (228 malaria patients and 226 healthy individuals). Malaria patients, divided into P. vivax and P. falciparum groups on the basis of the causative species of Plasmodium, were categorized into mild and severe on the basis of clinical outcomes according to WHO criteria. Healthy individuals were used as controls. Allele specific PCR based strategy was used for the identification of rs8177374 SNP. Results: MyD88-adaptor-like gene polymorphism was associated with susceptibility to malaria (p < 0.001). C allele frequency (0.74) was higher in the population compared to T allele frequency (0.26). CT genotype increased the susceptibility of malaria (OR: 2.661; 95% CI: 1.722-4.113) and was positively associated with mild malaria (OR: 5.609; 95% CI: 3.479-9.044, p = 0.00). On the other hand, CC genotype was associated with severe malaria (OR: 3.116; 95% CI: 1.560-6.224, p = 0.00). P. vivax infection rate was higher in CT genotype carriers compared to other genotypes (OR: 3.616; 95% CI: 2.219-5.894, p < 0.001). Conclusion: MyD88-adaptor-like/TIR domain containing adaptor protein polymorphism for single nucleotide polymorphism rs8177374 is related with the susceptibility of malaria.


Subject(s)
Humans , Male , Female , Adult , Membrane Glycoproteins/physiology , Malaria, Vivax/genetics , Malaria, Falciparum/genetics , Receptors, Interleukin-1/physiology , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Pakistan , Severity of Illness Index , Membrane Glycoproteins/genetics , Case-Control Studies , Polymerase Chain Reaction , Receptors, Interleukin-1/genetics , Gene Frequency , Genotype
9.
Braz J Infect Dis ; 21(4): 418-423, 2017.
Article in English | MEDLINE | ID: mdl-28482182

ABSTRACT

INTRODUCTION: The present study was designed to investigate the association between rs8177374 polymorphism and malaria symptoms due to exposure of Plasmodium vivax and Plasmodium falciparum. MATERIALS AND METHODS: A total of 454 samples were included in the study (228 malaria patients and 226 healthy individuals). Malaria patients, divided into P. vivax and P. falciparum groups on the basis of the causative species of Plasmodium, were categorized into mild and severe on the basis of clinical outcomes according to WHO criteria. Healthy individuals were used as controls. Allele specific PCR based strategy was used for the identification of rs8177374 SNP. RESULTS: MyD88-adaptor-like gene polymorphism was associated with susceptibility to malaria (p<0.001). C allele frequency (0.74) was higher in the population compared to T allele frequency (0.26). CT genotype increased the susceptibility of malaria (OR: 2.661; 95% CI: 1.722-4.113) and was positively associated with mild malaria (OR: 5.609; 95% CI: 3.479-9.044, p=0.00). On the other hand, CC genotype was associated with severe malaria (OR: 3.116; 95% CI: 1.560-6.224, p=0.00). P. vivax infection rate was higher in CT genotype carriers compared to other genotypes (OR: 3.616; 95% CI: 2.219-5.894, p<0.001). CONCLUSION: MyD88-adaptor-like/TIR domain containing adaptor protein polymorphism for single nucleotide polymorphism rs8177374 is related with the susceptibility of malaria.


Subject(s)
Genetic Predisposition to Disease/genetics , Malaria, Falciparum/genetics , Malaria, Vivax/genetics , Membrane Glycoproteins/physiology , Polymorphism, Single Nucleotide , Receptors, Interleukin-1/physiology , Severity of Illness Index , Adult , Case-Control Studies , Female , Gene Frequency , Genotype , Humans , Male , Membrane Glycoproteins/genetics , Pakistan , Polymerase Chain Reaction , Receptors, Interleukin-1/genetics
10.
Brain Res Bull ; 130: 165-172, 2017 04.
Article in English | MEDLINE | ID: mdl-28153540

ABSTRACT

Although interleukin-1ß (IL-1ß) is a prototypical pro-inflammatory cytokine, the specific mechanisms underlying the role of its cognate receptor, the interleukin-1 receptor (IL-1R) in peripheral sensitization remain to be investigated. Since emerging evidence in the literature indicates that IL-1ß can modulate membrane-bound receptors, we decided to examine the involvement of P2Y1 receptor (P2Y1R) in IL-1ß induced pain and the potential interaction of P2Y1Rs and IL-1Rs in both naïve and carrageenan injected rats. Intraplantar (i.pl) injection of IL-1ß dose-dependently produced mechanical and thermal hypersensitivity in naïve rats. Pre-treatment with IL-1ra (i.pl, 30 and 100ng), an endogenous IL-1R antagonist, prevented the IL-1ß induced mechanical and thermal hypersensitivity. Pre-treatment with MRS2500 (i.pl, 1 and 3nmol), a specific P2Y1R antagonist, dose-dependently reduced IL-1ß induced thermal hypersensitivity, but did not affect the development of mechanical hypersensitivity. Conversely coadministration of MRS2500 (i.pl, 0.1nmol, sub-effective dose) together with IL-1ra (10nmol, sub-effective dose) significantly reduced IL-1ß induced thermal, but not mechanical hypersensitivity. We next used immunohistochemistry to demonstrate that P2Y1 and IL-1 type I receptors co-localize predominantly in small diameter neurons in the dorsal root ganglion. We also performed experiments to examine the interaction of P2Y1Rs and IL-1Rs under the inflammatory conditions induced by 2% carrageenan. Intraplantar coadministration of MRS2500 (3nmol, sub-effective dose) and IL-1ra (30ng, sub-effective dose) significantly reduced inflammatory thermal, but not mechanical, hypersensitivity. These data indicate the involvement of P2Y1Rs in IL-1ß mediated pain in both naive and carrageenan injected rats. There is a positive interaction between peripheral P2Y1Rs and IL-1Rs in both IL-1ß and carrageenan-induced thermal hypersensitivity.


Subject(s)
Hyperalgesia/physiopathology , Interleukin-1beta/physiology , Receptors, Interleukin-1/physiology , Receptors, Purinergic P2Y1/physiology , Animals , Carrageenan/administration & dosage , Hyperalgesia/chemically induced , Interleukin-1beta/administration & dosage , Male , Pain Threshold , Rats, Sprague-Dawley
11.
Sci Rep ; 6: 31663, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27526865

ABSTRACT

Acute lung injury (ALI) is a major component of multiple organ dysfunction syndrome (MODS) following pulmonary infection. Alveolar macrophages (AM) are at the center of the pathogenesis of the development of ALI. Interleukin-1ß (IL-1ß) is one of the key pro-inflammatory mediators, and its maturation is tightly controlled by the formation and activation of the inflammasome. The biological effects of IL-1ß are mediated through IL-1 receptor (IL-1R). In this study, we investigated the influence of LPS-induced IL-1ß release and IL-1RI upregulation on the development of lung inflammation. We demonstrated that in AM, LPS-TLR4 signaling not only activates Nlrp3 inflammasome activation and subsequent release of IL-1ß, but also up-regulates IL-1RI expression on AM surface through MyD88 and NF-κB dependent signaling. The upregulated IL-1RI, therefore, sensitizes AM to IL-1ß and results in pyroptosome formation, which in turn leads to AM pyroptosis, a type of caspase-1-dependent inflammatory cell death. We further showed that AM pyroptosis exaggerates lung inflammation. The present study demonstrates a novel mechanism underlying LPS-induced innate immunity; that is, a secondary upregulation of IL-1ß-IL-1RI signaling is responsible for AM pyroptosis and augmented lung injury in response to LPS.


Subject(s)
Acute Lung Injury/immunology , Interleukin-1beta/physiology , Macrophages, Alveolar/immunology , Pneumonia/immunology , Pyroptosis/immunology , Receptors, Interleukin-1/physiology , Toll-Like Receptor 4/physiology , Animals , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/administration & dosage , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Up-Regulation
12.
Proc Natl Acad Sci U S A ; 113(32): E4671-80, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27462105

ABSTRACT

Inflammasomes are critical sensors that convey cellular stress and pathogen presence to the immune system by activating inflammatory caspases and cytokines such as IL-1ß. The nature of endogenous stress signals that activate inflammasomes remains unclear. Here we show that an inhibitor of the HIV aspartyl protease, Nelfinavir, triggers inflammasome formation and elicits an IL-1R-dependent inflammation in mice. We found that Nelfinavir impaired the maturation of lamin A, a structural component of the nuclear envelope, thereby promoting the release of DNA in the cytosol. Moreover, deficiency of the cytosolic DNA-sensor AIM2 impaired Nelfinavir-mediated inflammasome activation. These findings identify a pharmacologic activator of inflammasome and demonstrate the role of AIM2 in detecting endogenous DNA release upon perturbation of nuclear envelope integrity.


Subject(s)
Inflammasomes/drug effects , Nelfinavir/pharmacology , Nuclear Envelope/drug effects , Animals , CARD Signaling Adaptor Proteins/physiology , Caspase 1/metabolism , DNA/metabolism , Inflammasomes/physiology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nuclear Envelope/physiology , Receptors, Interleukin-1/physiology
13.
Nat Immunol ; 17(8): 906-13, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27434011

ABSTRACT

Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1ß are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1ß, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.


Subject(s)
Inflammation/physiopathology , Interleukin-1alpha/physiology , Alarmins/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation , Granuloma/etiology , Granuloma/metabolism , Humans , Inflammation/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/genetics , Macrophages/physiology , Mice , Mice, Inbred BALB C , Models, Biological , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/physiopathology , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational , Receptors, Interleukin-1/physiology , Signal Transduction
14.
Biochem Biophys Res Commun ; 477(3): 509-14, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27311859

ABSTRACT

Brucella spp. are known to avoid host immune recognition and weaken the immune response to infection. Brucella like accomplish this by employing two clever strategies, called the stealth strategy and hijacking strategy. The TIR domain-containing protein (TcpB/Btp1) of Brucella melitensis is thought to be involved in inhibiting host NF-κB activation by binding to adaptors downstream of Toll-like receptors. However, of the five TIR domain-containing adaptors conserved in mammals, whether MyD88 or MAL, even other three adaptors, are specifically targeted by TcpB has not been identified. Here, we confirmed the effect of TcpB on B.melitensis virulence in mice and found that TcpB selectively targets MAL. By using siRNA against MAL, we found that TcpB from B.melitensis is involved in intracellular survival and that MAL affects intracellular replication of B.melitensis. Our results confirm that TcpB specifically targets MAL/TIRAP to disrupt downstream signaling pathways and promote intra-host survival of Brucella spp.


Subject(s)
Bacterial Proteins/physiology , Brucella/metabolism , Membrane Glycoproteins/physiology , Receptors, Interleukin-1/physiology , Virulence Factors/physiology , Humans
15.
Mediators Inflamm ; 2015: 630265, 2015.
Article in English | MEDLINE | ID: mdl-26549942

ABSTRACT

The gene encoding IL-1 was sequenced more than 30 years ago, and many related cytokines, such as IL-18, IL-33, IL-36, IL-37, IL-38, IL-1 receptor antagonist (IL-1Ra), and IL-36Ra, have since been identified. IL-1 is a potent proinflammatory cytokine and is involved in various inflammatory diseases. Other IL-1 family ligands are critical for the development of diverse diseases, including inflammatory and allergic diseases. Only IL-1Ra possesses the leader peptide required for secretion from cells, and many ligands require posttranslational processing for activation. Some require inflammasome-mediated processing for activation and release, whereas others serve as alarmins and are released following cell membrane rupture, for example, by pyroptosis or necroptosis. Thus, each ligand has the proper molecular process to exert its own biological functions. In this review, we will give a brief introduction to the IL-1 family cytokines and discuss their pivotal roles in the development of various liver diseases in association with immune responses. For example, an excess of IL-33 causes liver fibrosis in mice via activation and expansion of group 2 innate lymphoid cells to produce type 2 cytokines, resulting in cell conversion into pro-fibrotic M2 macrophages. Finally, we will discuss the importance of IL-1 family cytokine-mediated molecular and cellular networks in the development of acute and chronic liver diseases.


Subject(s)
Interleukin-1/physiology , Liver Diseases/etiology , Animals , Cytokines/biosynthesis , Hepatitis, Viral, Human/etiology , Humans , Inflammation/etiology , Interleukin-18/physiology , Interleukin-33/physiology , Macrophages/immunology , Mice , Receptors, Interleukin-1/physiology
16.
J Biol Chem ; 290(50): 30163-74, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26483549

ABSTRACT

Toll-like receptors (TLR) and the receptor for interleukin-1 (IL-1R) signaling play an important role in bacteria-mediated bone loss diseases including periodontitis, rheumatoid arthritis, and osteomyelitis. Recent studies have shown that TLR ligands inhibit the receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation from un-committed osteoclast precursors, whereas IL-1 potentiates RANKL-induced osteoclast formation. However, IL-1R and TLR belong to the same IL-1R/TLR superfamily, and activate similar intracellular signaling pathways. Here, we investigate the molecular mechanisms underlying the distinct effects of IL-1 and Porphyromonas gingivalis lipopolysaccharide (LPS-PG) on RANKL-induced osteoclast formation. Our results show that LPS-PG and IL-1 differentially regulate RANKL-induced activation of osteoclast genes encoding Car2, Ctsk, MMP9, and TRAP, as well as expression of NFATc1, a master transcription factor of osteoclastogenesis. Regulation of osteoclast genes and NFATc1 by LPS-PG and IL-1 is dependent on MyD88, an important signaling adaptor for both TLR and IL-1R family members. Furthermore, LPS-PG and IL-1 differentially regulate RANKL-costimulatory receptor OSCAR (osteoclast-associated receptor) expression and Ca(2+) oscillations induced by RANKL. Moreover, LPS-PG completely abrogates RANKL-induced gene expression of B lymphocyte-induced maturation protein-1 (Blimp1), a global transcriptional repressor of anti-osteoclastogenic genes encoding Bcl6, IRF8, and MafB. However, IL-1 enhances RANKL-induced blimp1 gene expression but suppresses the gene expression of bcl6, irf8, and mafb. Our study reveals the involvement of multiple signaling molecules in the differential regulation of RANKL-induced osteoclastogenesis by TLR2 and IL-1 signaling. Understanding the signaling cross-talk among TLR, IL-1R, and RANK is critical for identifying therapeutic strategies to control bacteria-mediated bone loss.


Subject(s)
Myeloid Differentiation Factor 88/physiology , NFATC Transcription Factors/physiology , Osteoclasts/cytology , Receptors, Interleukin-1/physiology , Toll-Like Receptor 2/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Interleukin-1/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , RANK Ligand/physiology , Signal Transduction
17.
Mol Pharmacol ; 88(5): 949-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330549

ABSTRACT

Recognition of bacterial lipopolysaccharide (LPS) by innate immune system is mediated by the cluster of differentiation 14/Toll-like receptor 4/myeloid differentiation protein 2 (MD-2) complex. In this study, we investigated the modulatory effect of gedunin, a limonoid from species of the Meliaceae family described as a heat shock protein Hsp90 inhibitor, on LPS-induced response in immortalized murine macrophages. The pretreatment of wild-type (WT) macrophages with gedunin (0.01-100 µM, noncytotoxic concentrations) inhibited LPS (50 ng/ml)-induced calcium influx, tumor necrosis factor-α, and nitric oxide production in a concentration-dependent manner. The selective effect of gedunin on MyD88-adapter-like/myeloid differentiation primary response 88- and TRIF-related adaptor molecule/TIR domain-containing adapter-inducing interferon-ß-dependent signaling pathways was further investigated. The pretreatment of WT, TIR domain-containing adapter-inducing interferon-ß knockout, and MyD88 adapter-like knockout macrophages with gedunin (10 µM) significantly inhibited LPS (50 ng/ml)-induced tumor necrosis factor-α and interleukin-6 production, at 6 hours and 24 hours, suggesting that gedunin modulates a common event between both signaling pathways. Furthermore, gedunin (10 µM) inhibited LPS-induced prostaglandin E2 production, cyclooxygenase-2 expression, and nuclear factor κB translocation into the nucleus of WT macrophages, demonstrating a wide-range effect of this chemical compound. In addition to the ability to inhibit LPS-induced proinflammatory mediators, gedunin also triggered anti-inflammatory factors interleukin-10, heme oxygenase-1, and Hsp70 in macrophages stimulated or not with LPS. In silico modeling studies revealed that gedunin efficiently docked into the MD-2 LPS binding site, a phenomenon further confirmed by surface plasmon resonance. Our results reveal that, in addition to Hsp90 modulation, gedunin acts as a competitive inhibitor of LPS, blocking the formation of the Toll-like receptor 4/MD-2/LPS complex.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Limonins/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Antigen 96/metabolism , Macrophages/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Binding Sites , Cells, Cultured , Cytokines/biosynthesis , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Limonins/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Macrophages/drug effects , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Receptors, Interleukin-1/physiology , Toll-Like Receptor 4/physiology
18.
Comput Math Methods Med ; 2015: 893507, 2015.
Article in English | MEDLINE | ID: mdl-26161132

ABSTRACT

Familial mediterranean fever (FMF) and Cryopyrin associated periodic syndromes (CAPS) are two prototypical hereditary autoinflammatory diseases, characterized by recurrent episodes of fever and inflammation as a result of mutations in MEFV and NLRP3 genes encoding Pyrin and Cryopyrin proteins, respectively. Pyrin and Cryopyrin play key roles in the multiprotein inflammasome complex assembly, which regulates activity of an enzyme, Caspase 1, and its target cytokine, IL-1ß. Overproduction of IL-1ß by Caspase 1 is the main cause of episodic fever and inflammatory findings in FMF and CAPS. We present a unifying dynamical model for FMF and CAPS in the form of coupled nonlinear ordinary differential equations. The model is composed of two subsystems, which capture the interactions and dynamics of the key molecular players and the insults on the immune system. One of the subsystems, which contains a coupled positive-negative feedback motif, captures the dynamics of inflammation formation and regulation. We perform a comprehensive bifurcation analysis of the model and show that it exhibits three modes, capturing the Healthy, FMF, and CAPS cases. The mutations in Pyrin and Cryopyrin are reflected in the values of three parameters in the model. We present extensive simulation results for the model that match clinical observations.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/diagnosis , Cryopyrin-Associated Periodic Syndromes/physiopathology , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/physiopathology , Algorithms , Carrier Proteins/genetics , Computer Simulation , Cytoskeletal Proteins/genetics , Humans , Inflammasomes , Inflammation , Interleukin 1 Receptor Antagonist Protein/physiology , Interleukin-1beta/physiology , Interleukin-6/physiology , Models, Biological , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein , Pyrin , Receptors, Interleukin-1/physiology , Receptors, Interleukin-1 Type II/physiology
19.
Inflamm Bowel Dis ; 21(3): 543-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25659087

ABSTRACT

BACKGROUND: Ileal Crohn's disease is related to NOD2 mutations and to a gut barrier dysfunction. Pseudomonas fluorescens has also been associated with ileal Crohn's disease. The aim of this study was to determine the impact of P. fluorescens on the paracellular permeability in ileum and Peyer's patches. METHODS: To explore this question, in vivo and ex vivo experiments were performed in wild-type, Nod2, Nod2, and IL-1R mice together with in vitro analyses using the Caco-2 (epithelial) and the THP-1 (monocyte) human cell lines. RESULTS: Pseudomonas fluorescens increased the paracellular permeability of the intestinal mucosa through the secretion of IL-1ß by the immune cell populations and the activation of myosin light chain kinase in the epithelial cells. Induction of the IL-1ß pathway required the expression of Nod2 in the hematopoietic compartment, and muramyl dipeptide (a Nod2 ligand) had an inhibitory effect. CONCLUSIONS: Pseudomonas fluorescens thus alters the homeostasis of the epithelial barrier function by a mechanism similar to that previously observed for Yersinia pseudotuberculosis. This work further documents a putative role of psychrotrophic bacteria in Crohn's disease.


Subject(s)
Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Nod2 Signaling Adaptor Protein/physiology , Pseudomonas fluorescens/physiology , Receptors, Interleukin-1/physiology , Animals , Blotting, Western , Caco-2 Cells , Cell Membrane Permeability , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
20.
Biochim Biophys Acta ; 1853(1): 126-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25409926

ABSTRACT

Success in developing therapeutic approaches to target brain tumor-associated inflammation in patients has been limited. Given that the inflammatory microenvironment is a hallmark signature of solid tumor development, anti-inflammatory targeting strategies have been envisioned as preventing glioblastoma initiation or progression. Consumption of foods from plant origin is associated with reduced risk of developing cancers, a chemopreventive effect that is, in part, attributed to their high content of phytochemicals with potent anti-inflammatory properties. We explored whether luteolin, a common flavonoid in many types of plants, may inhibit interleukin (IL)-1ß function induction of the inflammation biomarker cyclooxygenase (COX)-2. We found that IL-1ß triggered COX-2 expression in U-87 glioblastoma cells and synergized with luteolin to potentiate or inhibit that induction in a biphasic manner. Luteolin pretreatment of cells inhibited IL-1ß-mediated phosphorylation of inhibitor of κB, nuclear transcription factor-κB (NF-κB) p65, extracellular signal-regulated kinase-1/2, and c-Jun amino-terminal kinase in a concentration-dependent manner. Luteolin also inhibited AKT phosphorylation and survivin expression, while it triggered both caspase-3 cleavage and expression of glucose-regulated protein 78. These effects were all potentiated by IL-1ß, in part through increased nuclear translocation of NF-κB p65. Finally, luteolin was able to reduce IL-1 receptor gene expression, and treatment with IL-1 receptor antagonist or gene silencing of IL-1 receptor prevented IL-1ß/luteolin-induced COX-2 expression. Our results document a novel adaptive cellular response to luteolin, which triggers anti-survival and anti-inflammatory mechanisms that contribute to the chemopreventive properties of this diet-derived molecule.


Subject(s)
Cyclooxygenase 2/genetics , Glioblastoma/enzymology , Interleukin-1beta/pharmacology , Luteolin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , MAP Kinase Signaling System/physiology , Receptors, Interleukin-1/physiology , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...