Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-35653648

ABSTRACT

With the growing interest in developing silver-based antimicrobials, there is a need to better understand the behavior of silver within biological systems. To address this, we showed that single-photon emission computed tomography (SPECT) is a suitable method to noninvasively image 111Ag-labeled compounds in mice. Formed by neutron irradiation of palladium foil, 111Ag can be rapidly isolated with a high degree of purity and stably incorporated into antimicrobial silver nanoparticles. The imaging showed that nanoparticles are retained in the lungs for up to 48 h following intratracheal instillation, with limited uptake into the systemic circulation or organs of the reticuloendothelial system. Furthermore, in a mouse model of pulmonary Pseudomonas aeruginosa infection, the nanoparticles reduced the bacterial burden by 11.6-fold without inducing the production of pro-inflammatory mediators. Overall, SPECT imaging with 111Ag is a useful tool for noninvasively visualizing the biodistribution of silver-containing compounds in rodents. This knowledge of how silver nanoparticles distribute in vivo can be used to predict their therapeutic efficacy.

2.
Front Cell Infect Microbiol ; 11: 694789, 2021.
Article in English | MEDLINE | ID: mdl-34249781

ABSTRACT

Pseudomonas aeruginosa is a metabolically versatile opportunistic pathogen capable of infecting distinct niches of the human body, including skin wounds and the lungs of cystic fibrosis patients. Eradication of P. aeruginosa infection is becoming increasingly difficult due to the numerous resistance mechanisms it employs. Adaptive resistance is characterized by a transient state of decreased susceptibility to antibiotic therapy that is distinct from acquired or intrinsic resistance, can be triggered by various environmental stimuli and reverted by removal of the stimulus. Further, adaptive resistance is intrinsically linked to lifestyles such as swarming motility and biofilm formation, both of which are important in infections and lead to multi-drug adaptive resistance. Here, we demonstrated that NtrBC, the master of nitrogen control, had a selective role in host colonization and a substantial role in determining intrinsic resistance to ciprofloxacin. P. aeruginosa mutant strains (ΔntrB, ΔntrC and ΔntrBC) colonized the skin but not the respiratory tract of mice as well as WT and, unlike WT, could be reduced or eradicated from the skin by ciprofloxacin. We hypothesized that nutrient availability contributed to these phenomena and found that susceptibility to ciprofloxacin was impacted by nitrogen source in laboratory media. P. aeruginosa ΔntrB, ΔntrC and ΔntrBC also exhibited distinct host interactions, including modestly increased cytotoxicity toward human bronchial epithelial cells, reduced virulence factor production and 10-fold increased uptake by macrophages. These data might explain why NtrBC mutants were less adept at colonizing the upper respiratory tract of mice. Thus, NtrBC represents a link between nitrogen metabolism, adaptation and virulence of the pathogen P. aeruginosa, and could represent a target for eradication of recalcitrant infections in situ.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Ciprofloxacin/pharmacology , Host-Pathogen Interactions , Humans , Mice , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/genetics , Virulence
3.
Front Microbiol ; 12: 640787, 2021.
Article in English | MEDLINE | ID: mdl-33927701

ABSTRACT

Bacterial biofilms are complex and highly antibiotic-resistant aggregates of microbes that form on surfaces in the environment and body including medical devices. They are key contributors to the growing antibiotic resistance crisis and account for two-thirds of all infections. Thus, there is a critical need to develop anti-biofilm specific therapeutics. Here we discuss mechanisms of biofilm formation, current anti-biofilm agents, and strategies for developing, discovering, and testing new anti-biofilm agents. Biofilm formation involves many factors and is broadly regulated by the stringent response, quorum sensing, and c-di-GMP signaling, processes that have been targeted by anti-biofilm agents. Developing new anti-biofilm agents requires a comprehensive systems-level understanding of these mechanisms, as well as the discovery of new mechanisms. This can be accomplished through omics approaches such as transcriptomics, metabolomics, and proteomics, which can also be integrated to better understand biofilm biology. Guided by mechanistic understanding, in silico techniques such as virtual screening and machine learning can discover small molecules that can inhibit key biofilm regulators. To increase the likelihood that these candidate agents selected from in silico approaches are efficacious in humans, they must be tested in biologically relevant biofilm models. We discuss the benefits and drawbacks of in vitro and in vivo biofilm models and highlight organoids as a new biofilm model. This review offers a comprehensive guide of current and future biological and computational approaches of anti-biofilm therapeutic discovery for investigators to utilize to combat the antibiotic resistance crisis.

4.
Front Cell Infect Microbiol ; 11: 621081, 2021.
Article in English | MEDLINE | ID: mdl-33777834

ABSTRACT

The very common condition of sinusitis is characterized by persistent inflammation of the nasal cavity, which contributes to chronic rhinosinusitis and morbidity of cystic fibrosis patients. Colonization by opportunistic pathogens such as Staphylococcus aureus and Pseudomonas aeruginosa triggers inflammation that is exacerbated by defects in the innate immune response. Pathophysiological mechanisms underlying initial colonization of the sinuses are not well established. Despite their extensive use, current murine models of acute bacterial rhinosinusitis have not improved the understanding of early disease stages due to analytical limitations. In this study, a model is described that is technically simple, allows non-invasive tracking of bacterial infection, and screening of host-responses to infection and therapies. The model was modified to investigate longer-term infection and disease progression by using a less virulent, epidemic P. aeruginosa cystic fibrosis clinical isolate LESB65. Tracking of luminescent bacteria was possible after intranasal infections, which were sustained for up to 120 h post-infection, without compromising the overall welfare of the host. Production of reactive oxidative species was associated with neutrophil localization to the site of infection in this model. Further, host-defense peptides administered by Respimat® inhaler or intranasal instillation reduced bacterial burden and impacted disease progression as well as cytokine responses associated with rhinosinusitis. Thus, future studies using this model will improve our understanding of rhinosinusitis etiology and early stage pathogenesis, and can be used to screen for the efficacy of emerging therapies pre-clinically.


Subject(s)
Anti-Infective Agents , Pseudomonas Infections , Rhinitis , Sinusitis , Animals , Chronic Disease , Disease Models, Animal , Humans , Immunomodulation , Mice , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa , Rhinitis/drug therapy , Sinusitis/drug therapy
5.
Article in English | MEDLINE | ID: mdl-32509598

ABSTRACT

Host defense peptides (HDPs), also known as antimicrobial peptides, are naturally occurring polypeptides (~12-50 residues) composed of cationic and hydrophobic amino acids that adopt an amphipathic conformation upon folding usually after contact with membranes. HDPs have a variety of biological activities including immunomodulatory, anti-inflammatory, anti-bacterial, and anti-biofilm functions. Although HDPs have the potential to address the global threat of antibiotic resistance and to treat immune and inflammatory disorders, they have yet to achieve this promise. Indeed, there are several challenges associated with bringing peptide-based drug candidates from the lab bench to clinical practice, including identifying appropriate indications, stability, toxicity, and cost. These challenges can be addressed in part by the development of innate defense regulator (IDR) peptides and peptidomimetics, which are synthetic derivatives of HDPs with similar or better efficacy, increased stability, and reduced toxicity and cost of the original HDP. However, one of the largest gaps between basic research and clinical application is the validity and translatability of conventional model systems, such as cell lines and animal models, for screening HDPs and their derivatives as potential drug therapies. Indeed, such translation has often relied on animal models, which have only limited validity. Here we discuss the recent development of human organoids for disease modeling and drug screening, assisted by the use of omics analyses. Organoids, developed from primary cells, cell lines, or human pluripotent stem cells, are three-dimensional, self-organizing structures that closely resemble their corresponding in vivo organs with regards to immune responses, tissue organization, and physiological properties; thus, organoids represent a reliable method for studying efficacy, formulation, toxicity and to some extent drug stability and pharmacodynamics. The use of patient-derived organoids enables the study of patient-specific efficacy, toxicogenomics and drug response predictions. We outline how organoids and omics data analysis can be leveraged to aid in the clinical translation of IDR peptides.


Subject(s)
Antimicrobial Cationic Peptides , Peptidomimetics , Animals , Bacteria , Biofilms , Humans , Organoids
6.
Sci Rep ; 9(1): 14204, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578370

ABSTRACT

Canadian Indigenous peoples (First Nations and Inuit) exhibit a high burden of infectious diseases including tuberculosis influenced by societal factors, and biological determinants. Toll-like receptor (TLR)-mediated innate immune responses are the first line of defence against infections. We examined the production of a panel of 30 cytokines in peripheral blood-derived mononuclear cells (PBMC) isolated from Indigenous and non-Indigenous participants, following stimulation with five different TLR ligands. The levels of TLR-induced pro-inflammatory cytokines such as IL-12/23p40, IL-16, and IFN-γ, and chemokines (MCP-4, MDC and eotaxin) were different between Indigenous compared to non-Indigenous participants. Antimicrobial cationic host defence peptides (CHDP) induced by TLR activation are critical for resolution of infections and modulate the TLR-to-NFκB pathway to alter downstream cytokine responses. Therefore, we examined the expression of human CHDP defensins and cathelicidin in PBMC. mRNA expression of genes encoding for def-A1 and def-B1 were significantly higher following stimulation with TLR ligands in Indigenous compared to non-Indigenous participants. The purinergic receptor P2X7 known to be activated by ATP released following TLR stimulation, is a receptor for CHDP. Therefore, we further examined single nucleotide polymorphisms (SNP) in P2X7. Indigenous participants had a significantly higher percentage of a P2X7 SNP which is associated with reduced function and lower ability to clear infections. These results suggest that a higher frequency of non-functional P2X7 receptors may influence the activity of downstream immune mediators required for resolution of infections such as pro-inflammatory cytokines and CHDP defensins, thus contributing to higher burden of infections in Indigenous population.


Subject(s)
Indigenous Peoples/genetics , Polymorphism, Genetic/genetics , Receptors, Purinergic P2X7/genetics , Toll-Like Receptors/genetics , Canada/epidemiology , Cytokines/genetics , Defensins/genetics , Humans , Immunity, Innate/genetics , Interleukin-12/genetics , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Polymorphism, Single Nucleotide/genetics , Risk Factors , Signal Transduction/genetics
7.
Biomolecules ; 9(9)2019 09 18.
Article in English | MEDLINE | ID: mdl-31540479

ABSTRACT

The anti-endotoxin activity of the cationic peptide LL-37 and its derivative IG-19 is attributed to electrostatic interaction of the peptides' positive charge with negatively charged bacterial lipopolysaccharides (LPS), and in part to the alteration of intracellular mechanisms independent of peptide binding to LPS. We examined the immunomodulatory responses induced by IG-19 and four IG-19-derived scrambled peptides (IG-19a-d), in the presence and absence of LPS, in macrophages and peripheral blood-derived mononuclear cells. All peptides had identical net charge (+5) and amino acid composition, but different hydrophobicity and α-helical propensity. Peptide IG-19 suppressed LPS-induced cytokine/chemokine production by >90%, IG-19a and IG-19b suppressed it by 40-50%, and IG-19c and IG-19d did not suppress cytokine/chemokine production at all. In silico prediction algorithms and the peptide retention time (RT) on a C18 RP HPLC column indicated a linear association between α-helical propensity and hydrophobicity with the ability of the peptides to inhibit LPS-induced responses. Peptide RT exhibited a significant correlation (>70%) between the suppression of LPS-induced cytokine/chemokine production and peptide-induced production of the anti-inflammatory cytokine IL-1RA. These results indicate that RT on a C18 column can be used as a predictor for the immunomodulatory functions of cationic peptides. Overall, we demonstrated that the immunomodulatory functions of LL-37-derived peptides with identical positive charge and amino acid composition are directly associated with the predicted α-helical propensity and hydrophobicity of the peptides.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Lipopolysaccharides/adverse effects , Peptides/pharmacology , Anti-Inflammatory Agents/chemistry , Computer Simulation , Cytokines/metabolism , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Interleukin 1 Receptor Antagonist Protein/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Macrophages/drug effects , Macrophages/immunology , Peptides/chemistry , Protein Conformation, alpha-Helical , THP-1 Cells , Cathelicidins
8.
Front Immunol ; 9: 1871, 2018.
Article in English | MEDLINE | ID: mdl-30158931

ABSTRACT

The human host defense peptide LL-37 promotes immune activation such as induction of chemokine production and recruitment of leukocytes. Conversely, LL-37 also mediates anti-inflammatory responses such as production of anti-inflammatory cytokines, e.g., IL-1RA, and the control of pro-inflammatory cytokines, e.g., TNF. The mechanisms regulating these disparate immunomodulatory functions of LL-37 are not completely understood. Rho GTPases are GTP-binding proteins that promote fundamental immune functions such as chemokine production and recruitment of leukocytes. However, recent studies have shown that distinct Rho proteins can both negatively and positively regulate inflammation. Therefore, we interrogated the role of Rho GTPases in LL-37-mediated immunomodulation. We demonstrate that LL-37-induced production of chemokines, e.g., GRO-α and IL-8 is largely dependent on Cdc42/Rac1 Rho GTPase, but independent of the Ras pathway. In contrast, LL-37-induced production of the anti-inflammatory cytokine IL-1RA is not dependent on either Cdc42/Rac1 RhoGTPase or Ras GTPase. Functional studies confirmed that LL-37-induced recruitment of leukocytes (monocytes and neutrophils) is also dependent on Cdc42/Rac1 RhoGTPase activity. We demonstrate that Cdc42/Rac1-dependent bioactivity of LL-37 involves G-protein-coupled receptors (GPCR) and JNK mitogen-activated protein kinase (MAPK) signaling, but not p38 or ERK MAPK signaling. We further show that LL-37 specifically enhances the activity of Cdc42 Rho GTPase, and that the knockdown of Cdc42 suppresses LL-37-induced production of chemokines without altering the peptide's ability to induce IL-1RA. This is the first study to demonstrate the role of Rho GTPases in LL-37-mediated responses. We demonstrate that LL-37 facilitates chemokine production and leukocyte recruitment engaging Cdc42/Rac1 Rho GTPase via GPCR and the JNK MAPK pathway. In contrast, LL-37-mediated anti-inflammatory cytokine IL-1RA production is independent of either Rho or Ras GTPase. The results of this study suggest that Cdc42 Rho GTPase may be the molecular switch that controls the opposing functions of LL-37 in the process of inflammation.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Monocytes/immunology , Neutrophils/immunology , cdc42 GTP-Binding Protein/metabolism , Antimicrobial Cationic Peptides/immunology , Cells, Cultured , Chemokine CXCL1/metabolism , Chemotaxis , Humans , Immunomodulation , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/metabolism , cdc42 GTP-Binding Protein/genetics , ras Proteins/metabolism , Cathelicidins
9.
Immunology ; 143(1): 68-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24666281

ABSTRACT

Human cathelicidin LL-37 protects against infections and endotoxin-induced inflammation. In a recent study we have shown that IG-19, an LL-37-derived peptide, protects in a murine model of arthritis. Cytokine interleukin-32 (IL-32) is elevated and directly associated with the disease severity of inflammatory arthritis. Therefore, in this study we examined the effects of LL-37 and IG-19 on IL-32-induced responses in human peripheral blood-derived mononuclear cells (PBMC) and macrophages. We showed that CD14(+) monocytes are the primary cells that produce pro-inflammatory tumour necrosis factor-α (TNF-α) following stimulation of PBMC with IL-32. We demonstrated that LL-37 and IG-19 significantly suppress IL-32-induced production of pro-inflammatory cytokines, e.g. TNF-α and IL-1ß, without altering chemokine production. In contrast, LL-37 and IG-19 enhance the production of the anti-inflammatory cytokine IL-1RA. Further mechanistic studies revealed that LL-37 and IG-19 suppress IL-32-mediated phosphorylation of Fyn (Y420) Src kinase. In contrast, IL-32-mediated phosphorylation of AKT-1 (T308) and MKP-1 (S359) is not suppressed by the peptides. LL-37 and IG-19 alone induce the phosphorylation of MKP-1 (S359), which is a known negative regulator of inflammation. Furthermore, the peptides induce the activity of p44/42 mitogen-activated protein kinase, which is known to phosphorylate MKP-1 (S359). This is the first study to demonstrate the regulation of IL-32-induced inflammation by LL-37 and its derivative peptide IG-19. The mechanistic results from this study suggest that regulation of immune-mediated inflammation by these peptides may be controlled by the dual phosphatase MKP-1. We speculate that LL-37 and its derivatives may contribute to the control of immune-mediated inflammatory diseases.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Inflammation/immunology , Interleukins/immunology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Signal Transduction/immunology , Blotting, Western , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Cathelicidins
10.
Mol Immunol ; 57(2): 86-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24091294

ABSTRACT

Current therapies for autoimmune chronic inflammatory diseases e.g. rheumatoid arthritis (RA) include inhibitors of inflammatory cytokines. However, these therapies can result in increased risk of infections. There is a need to explore alternate strategies that can control inflammation without compromising the innate ability to resolve infections. In this study, we examined the effect of small peptides derived from endogenous cathelicidin peptides in a murine model of collagen-induced arthritis (CIA). Cathelicidins are immunomodulatory peptides known to control infections. We demonstrate that the administration of the peptide IG-19, which represents an internal segment of the human cathelicidin LL-37, decreased disease severity and significantly reduced the serum levels of antibodies against collagen type II in the CIA model. IG-19 peptide reduced cellular infiltration in joints, prevented cartilage degradation and suppressed pro-inflammatory cytokines in the CIA mice. We also showed that not all cathelicidin-derived peptides exhibit similar functions. A bovine cathelicidin-derived peptide IDR-1018 did not exhibit the beneficial effects observed with the human cathelicidin LL-37-derived peptide IG-19, in the same murine model of CIA. This is the first study to provide evidence demonstrating the ability of a peptide derived from the human cathelicidin LL-37 to alleviate the arthritic disease process in a murine model of RA. Our results has lead us to propose a new approach for controlling autoimmune chronic inflammatory disorders such as RA, by using specific synthetic derivatives of endogenous host defence peptides. Cathelicidin-derived peptides are particularly attractive for their dual antimicrobial and anti-inflammatory actions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , Arthritis, Experimental/therapy , Cytokines/blood , Animals , Arthritis, Rheumatoid/therapy , Collagen/immunology , Cytokines/drug effects , Disease Models, Animal , Joints , Male , Mice , Mice, Inbred DBA , Cathelicidins
12.
J Immunol Methods ; 382(1-2): 189-95, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22698787

ABSTRACT

An impediment in the development of new therapeutic strategies for chronic inflammatory diseases is the limited understanding of underlying molecular mechanisms. The objective of this study was to identify newly synthesized (nascent) proteins induced by critical inflammatory cytokines TNF-α and IL-1ß in human monocytic THP-1 cells. We optimized methods to combine two different approaches, bio-orthogonal non-canonical amino acid tagging (BONCAT) along with proteomics using isobaric tags (iTRAQ). BONCAT employed the incorporation of l-azidohomoalanine (AHA), an analog of methionine, into TNF-α or IL-1ß induced nascent proteins. The AHA-containing nascent proteins were tagged with alkyne-biotin to allow enrichment using avidin affinity purification. The differential expressions of the enriched proteins were further determined using iTRAQ reagents and mass spectrometry (MS). The combination of BONCAT and proteomics represents a unique approach that has uncovered the nascent proteome induced by inflammatory cytokines TNF-α and IL-1ß.


Subject(s)
Alanine/analogs & derivatives , Interleukin-1beta/immunology , Membrane Transport Proteins/immunology , Proteome/analysis , Proteomics , Tumor Necrosis Factor-alpha/immunology , Alanine/chemistry , Alanine/immunology , Cell Line , Humans , Interleukin-1beta/pharmacology , Mass Spectrometry , Membrane Transport Proteins/chemistry , Monocytes/drug effects , Monocytes/metabolism , Proteome/chemistry , Proteome/drug effects , Proteome/immunology , Tumor Necrosis Factor-alpha/pharmacology
13.
J Innate Immun ; 4(4): 361-70, 2012.
Article in English | MEDLINE | ID: mdl-22739631

ABSTRACT

Host defence peptides (HDPs) are innate immune effector molecules found in diverse species. HDPs exhibit a wide range of functions ranging from direct antimicrobial properties to immunomodulatory effects. Research in the last decade has demonstrated that HDPs are critical effectors of both innate and adaptive immunity. Various studies have hypothesized that the antimicrobial property of certain HDPs may be largely due to their immunomodulatory functions. Mechanistic studies revealed that the role of HDPs in immunity is very complex and involves various receptors, signalling pathways and transcription factors. This review will focus on the multiple functions of HDPs in immunity and inflammation, with special reference to cathelicidins, e.g. LL-37, certain defensins and novel synthetic innate defence regulator peptides. We also discuss emerging concepts of specific HDPs in immune-mediated inflammatory diseases, including the potential use of cationic peptides as therapeutics for immune-mediated inflammatory disorders.


Subject(s)
Antimicrobial Cationic Peptides , Immunomodulation/immunology , Inflammation/immunology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Humans , Immunologic Factors/chemistry , Immunologic Factors/immunology , Immunologic Factors/metabolism , Molecular Sequence Data , Cathelicidins
14.
Arthritis Res Ther ; 13(4): R129, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21835002

ABSTRACT

INTRODUCTION: Innate defence regulator (IDR) peptides are synthetic cationic peptides, variants of naturally occurring innate immune effector molecules known as host defence peptides. IDR peptides were recently demonstrated to limit infection-associated inflammation selectively without compromising host innate immune functions. This study examined the impact of a 12-amino acid IDR peptide, IDR-1002, in pro-inflammatory cytokine interleukin (IL)-1ß-induced responses in synovial fibroblasts, a critical cell type in the pathogenesis of inflammatory arthritis. METHODS: Human fibroblast-like synoviocytes (FLS) were stimulated with IL-1ß in the presence and absence of IDR-1002. Production of enzyme matrix metalloproteinase-3 (MMP-3) and IL-1-receptor antagonist (IL-1RA) was monitored by enzyme-linked immunosorbent assay (ELISA), and various chemokines were evaluated by using multiplex cytometric bead array. Transcriptional responses were analyzed by quantitative real-time PCR. The impact on IL-1ß-induced proteome was investigated by quantitative proteomics by using isobaric tags. IL-1ß-induced pathways altered by IDR-1002 implicated by the proteomics analyses were further investigated by using various immunochemical assays. Cellular uptake of the peptide was monitored by using a biotinylated IDR-1002 peptide followed by microscopy probing with streptavidin-Alexa Fluor. RESULTS: This study demonstrated that IDR-1002 suppressed the production of IL-1ß-induced MMP-3 and monocyte chemotactic protein-1 (MCP-1); in contrast, IDR-1002 enhanced the production of IL-1RA, without neutralizing all chemokine responses. IDR-1002 altered the IL-1ß-induced proteome primarily by altering the expression of members of nuclear factor kappa-B (NF-κB) and c-Jun N-terminal kinase (JNK) pathways. The proteomics data also suggested that IDR-1002 was altering the transcription factor HNF-4α-mediated responses, known to be critical in metabolic regulation. With various immunochemical assays, it was further demonstrated that IL-1ß-induced NF-κB, JNK, and p38 mitogen-activated protein kinase (MAPK) activations were significantly suppressed by IDR-1002. CONCLUSIONS: This study demonstrates the ability of an innate immune-modulatory IDR-peptide to influence the IL-1ß-induced regulatory pathways and selectively to suppress inflammatory responses in synovial fibroblasts. The results of this study provide a rationale for examining the use of IDR-peptides as potential therapeutic candidates for chronic inflammatory diseases such as inflammatory arthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Fibroblasts/drug effects , Inflammation/immunology , Signal Transduction/drug effects , Blotting, Western , Cell Separation , Cells, Cultured , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Fibroblasts/immunology , Flow Cytometry , Humans , Immunohistochemistry , Inflammation/chemically induced , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , Synovial Membrane/drug effects , Synovial Membrane/immunology
15.
J Immunol ; 186(12): 7127-35, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21602493

ABSTRACT

Cytokines IL-32 and IL-17 are emerging as critical players in the pathophysiology of immune-mediated chronic inflammatory diseases. It has been speculated that the molecular mechanisms governing IL-32- and IL-17-mediated cellular responses are differentially dependent on the TNF pathway. In this study, kinome analysis demonstrated that following stimulation with cytokine IL-32, but not IL-17, there was increased phosphorylation of a peptide target corresponding to TNF-R1. Consistent with this observation, blocking TNF-R1 resulted in a suppression of IL-32-induced downstream responses, indicating that IL-32-mediated activity may be dependent on TNF-R1. In contrast, blocking TNF-R1 did not affect IL-17-induced downstream responses. Kinome analysis also implicated p300 (transcriptional coactivator) and death-associated protein kinase-1 (DAPK-1) as signaling intermediates for both IL-32 and IL-17. Phosphorylation of p300 and DAPK-1 upon stimulation with either IL-32 or IL-17 was confirmed by immunoblots. The presence of common targets was supported by results demonstrating similar downstream responses induced in the presence of IL-32 and IL-17, such as transcriptional responses and the direct activation of NF-κB. Furthermore, knockdown of p300 and DAPK-1 altered downstream responses induced by IL-32 and IL-17, and impacted certain cellular responses induced by TNF-α and IL-1ß. We hypothesize that p300 and DAPK-1 represent nodes where the inflammatory networks of IL-32 and IL-17 overlap, and that these proteins would affect both TNF-R1-dependent and -independent pathways. Therefore, p300 and DAPK-1 are viable potential therapeutic targets for chronic inflammatory diseases.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Receptors, Tumor Necrosis Factor/immunology , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases , Death-Associated Protein Kinases , Humans , Signal Transduction , p300-CBP Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL