Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Viral Immunol ; 34(9): 646-652, 2021 11.
Article in English | MEDLINE | ID: mdl-34287077

ABSTRACT

Host exposure to pathogens engage multiple pathogen recognition receptors (PRRs) including toll-like receptors (TLRs); recruit intracellular signaling adaptor proteins primarily myeloid differentiation primary response protein 88 (MyD88) for activating downstream signaling cascades, which culminate in the production of type I interferons (IFNs), proinflammatory cytokines, and chemokines; and impede pathogen replication and dissemination. However, recent studies highlight that absence of MyD88 increased antiviral type I IFN induction, and MyD88-/- mice showed a higher survival rate compared with the low survival rate of the MyD88+/+ mice, implicating MyD88 limits antiviral type I IFN response. As a single infectious agent may harbor multiple PRR agonists, which trigger different sets of TLR-initiated immune signaling, we examined whether MyD88 inhibition during stimulation of cells with more than one TLR-ligand would augment type I IFN. We stimulated human U87- and TLR3-transfected HEK293-TLR7 cells with TLR-ligands, such as lipopolysaccharides (LPS) (TLR4-ligand) plus poly I:C (TLR3-ligand) or imiquimod (R837, TLR7-ligand) plus poly I:C, in the presence of compound 4210, a previously reported MyD88 inhibitor, and measured IFN-ß response using an enzyme-linked immunosorbent assay. Our results showed that when U87- or TLR3-transfected HEK293-TLR7 cells were stimulated with TLR-ligands, such as poly I:C plus LPS or poly I:C plus R837, IFN-ß production was significantly increased with MyD88 inhibition in a dose-dependent manner. Collectively, these results indicate that during more than one TLR-ligand-induced immune signaling event, impairment of antiviral type I IFN response was restored by inhibition of MyD88 through MyD88-independent pathway of type I IFN signaling, thus, offer a MyD88-targeted approach for type I IFN induction.


Subject(s)
Interferon-beta , Myeloid Differentiation Factor 88 , Animals , HEK293 Cells , Humans , Interferon-beta/metabolism , Ligands , Lipopolysaccharides/pharmacology , Mice , Signal Transduction , Toll-Like Receptors
2.
Immunol Res ; 69(2): 117-128, 2021 04.
Article in English | MEDLINE | ID: mdl-33834387

ABSTRACT

The continuous emergence of infectious pathogens along with antimicrobial resistance creates a need for an alternative approach to treat infectious diseases. Targeting host factor(s) which are critically involved in immune signaling pathways for modulation of host immunity offers to treat a broad range of infectious diseases. Upon pathogen-associated ligands binding to the Toll-like/ IL-1R family, and other cellular receptors, followed by recruitment of intracellular signaling adaptor proteins, primarily MyD88, trigger the innate immune responses. But activation of host innate immunity strongly depends on the correct function of MyD88 which is tightly regulated. Dysregulation of MyD88 may cause an imbalance that culminates to a wide range of inflammation-associated syndromes and diseases. Furthermore, recent reports also describe that MyD88 upregulation with many viral infections is linked to decreased antiviral type I IFN response, and MyD88-deficient mice showed an increase in survivability. These reports suggest that MyD88 is also negatively involved via MyD88-independent pathways of immune signaling for antiviral type I IFN response. Because of its expanding role in controlling host immune signaling pathways, MyD88 has been recognized as a potential drug target in a broader drug discovery paradigm. Targeting BB-loop of MyD88, small molecule inhibitors were designed by structure-based approach which by blocking TIR-TIR domain homo-dimerization have shown promising therapeutic efficacy in attenuating MyD88-mediated inflammatory impact, and increased antiviral type I IFN response in experimental mouse model of diseases. In this review, we highlight the reports on MyD88-linked immune response and MyD88-targeted therapeutic approach with underlying mechanisms for controlling inflammation and antiviral type I IFN response. HIGHLIGHTS: • Host innate immunity is activated upon PAMPs binding to PRRs followed by immune signaling through TIR domain-containing adaptor proteins mainly MyD88. • Structure-based approach led to develop small-molecule inhibitors which block TIR domain homodimerization of MyD88 and showed therapeutic efficacy in limiting severe inflammation-associated impact in mice. • Therapeutic intervention of MyD88 also showed an increase in antiviral effect with strong type I IFN signaling linked to increased phosphorylation of IRFs via MyD88-independent pathway. • MyD88 inhibitors might be potentially useful as a small-molecule therapeutics for modulation of host immunity against inflammatory diseases and antiviral therapy. • However, prior clinical use of more in-depth efforts should be focused for suitability of the approach in deploying to complex diseases including COPD and COVID-19 in limiting inflammation-associated syndrome to infection.


Subject(s)
Drug Delivery Systems , Immunity, Innate/drug effects , Myeloid Differentiation Factor 88 , Virus Diseases , Animals , Disease Models, Animal , Humans , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Virus Diseases/drug therapy , Virus Diseases/immunology
3.
J Med Microbiol ; 69(10): 1240-1248, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32815800

ABSTRACT

Introduction. Melioidosis, caused by Burkholderia pseudomallei, in endemic areas, poses a challenge for treating the diseased populations without accurate diagnosis, and the disease-specific biomarkers linked with the infection have yet to be reported. Due to the invasive nature of the causative agent, Burkholderia pseudomallei, host innate effector mechanisms, including autophagy are known to be activated, resulting in differential expression of cellular proteins and immune markers. Identification of a disease-specific biomarker associated with B. pseudomallei infection will be helpful to facilitate rapid confirmation of melioidosis, which would enable early treatment and therapeutic success.Aim. We aimed to assess the levels of a host autophagy component, p62/NBR1, which function as a cargo-receptor in the process of autophagy activation leading to the degradation of ubiquitin-coated intracellular bacteria in which p62/NBR1 itself is degraded in the clearance of the pathogen. We further probed the extent of intracellular p62/NBR1 degradation and assessed its potential as a melioidosis biomarker.Methodology. We analysed peripheral blood mononuclear cell (PBMC) lysates using an ELISA-based assay for detecting cytosolic autophagy-related proteins p62/NBR1. We measured p62/NBR1 levels in diseased (confirmed B. pseudomallei infection) and non -diseased populations and utilized receiver operating characteristic (ROC) curve and max Youden index analysis for evaluating potential disease biomarker characteristics.Results. Our results revealed a three to fivefold increase in p62/NBR1 levels confirmed melioidosis cases compared to uninfected healthy donors. Comparable to p62/NBR1, levels of cytosolic LC3-I levels also increased, whereas the levels of degraded membrane bound form LC3-II was low, suggesting autophagy deficiency. Proinflammatory serum cytokine response, particularly IL-6, was consistently higher alongside B. pseudomallei infection in comparison to healthy controls.Conclusions. ROC curve and max Youden index analysis suggest that increased p62/NBR1 levels in diseased populations display characteristics of a potential disease biomarker in melioidosis and illustrates that an elevated p62/NBR1 level, in conjunction with B. pseudomallei infection associated with autophagy deficiency.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Melioidosis/metabolism , RNA-Binding Proteins/metabolism , Adult , Autophagy/physiology , Biomarkers/metabolism , Burkholderia pseudomallei/metabolism , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Melioidosis/microbiology , Sri Lanka
4.
Antiviral Res ; 181: 104854, 2020 09.
Article in English | MEDLINE | ID: mdl-32621945

ABSTRACT

Recent studies highlight that infection with Coxsackievirus B3, Venezuelan equine encephalitis virus (VEEV), Marburg virus, or stimulation using poly I:C (dsRNA), upregulates the signaling adaptor protein MyD88 and impairs the host antiviral type I interferon (IFN) responses. In contrast, MyD88 deficiency (MyD88-/-) increases the type I IFN and survivability of mice implying that MyD88 up regulation limits the type I IFN response. Reasoning that MyD88 inhibition in a virus-like manner may increase type I IFN responses, our studies revealed lipopolysaccharide stimulation of U937 cells or poly I:C stimulation of HEK293-TLR3, THP1 or U87 cells in the presence of a previously reported MyD88 inhibitor (compound 4210) augmented IFN-ß and RANTES production. Consistent with these results, overexpression of MyD88 decreased IFN-ß, whereas MyD88 inhibition rescued IFN-ß production concomitant with increased IRF3 phosphorylation, suggesting IRF-mediated downstream signaling to the IFN-ß response. Further, compound 4210 treatment inhibited MyD88 interaction with IRF3/IRF7 indicating that MyD88 restricts type I IFN signaling through sequestration of IRF3/IRF7. In cell based infection assays, compound 4210 treatment suppressed replication of VEEV, Eastern equine encephalitis virus, Ebola virus (EBOV), Rift Valley Fever virus, Lassa virus, and Dengue virus with IC50 values ranging from 11 to 42 µM. Notably, administration of compound 4210 improved survival, weight change, and clinical disease scores in mice following challenge with VEEV TC-83 and EBOV. Collectively, these results provide evidence that viral infections responsive to MyD88 inhibition lead to activation of IRF3/IRF7 and promoted a type I IFN response, thus, raising the prospect of an approach of host-directed antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Interferon Type I/genetics , Myeloid Differentiation Factor 88/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Viruses/drug effects , Animals , HEK293 Cells , Humans , Inhibitory Concentration 50 , Interferon Regulatory Factor-3/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Phosphorylation , Poly I-C/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/chemical synthesis , THP-1 Cells , Virus Diseases/drug therapy , Virus Replication/drug effects , Viruses/classification
5.
Immun Inflamm Dis ; 7(1): 7-21, 2019 03.
Article in English | MEDLINE | ID: mdl-30569531

ABSTRACT

INTRODUCTION: Burkholderia mallei (B. mallei) and Burkholderia pseudomallei (B. pseudomallei), causative agents of glanders and melioidosis, respectively, are invasive intracellular pathogens that actively multiply in phagocytic and non-phagocytic cells. Activation of cell-autonomous autophagy mechanism eliminate intracellular pathogens in which p62 a cytosolic cargo protein is selectively degraded, and an accumulation of this marker occurs if autophagy is deficient. Recurrent, relapsed and reinfection of B. pseudomallei in melioidosis patients in endemic area indicative of lack of complete of clearance and persistence of the pathogen. Reasoning that abundance in the levels of p62 may provide an indication of the intracellular infection, we sought to examine whether increase in intracellular p62 and bacterial burden with Burkholderia infection are linked to autophagy deficiency. METHODS: In this study, we investigated cell culture and mouse models of disease to identify an association between autophagy biomarkers (p62/NBR1) accumulation and intracellular persistence of B. mallei and B. pseudomallei. RESULTS: We demonstrate, that elevated levels of intracellular p62/NBR1 correlated with bacterial persistence, while pre-treatment with a pharmacological inducer of autophagy, rapamycin, reduced both intracellular p62, and bacterial survival. Our results showed an elevated p62 levels (2-5 fold) in spleen and liver cells of Burkholderia-infected BALB/c mice, as well as in spleen cells of Burkholderia-infected C57BL/6 mice, suggesting that an increase in p62/NBR1 was due to an autophagy deficiency. Similar to p62, cytosolic LC3-I levels were also elevated, while the characteristic conversion to the autophagosome-associated membrane bound form LC3-II was low in spleens of the infected mice further supporting the conclusion that autophagy was deficient. CONCLUSION: Taken together, our results suggest that an increase in intracellular p62/NBR1 may be a potential host cell biomarker of B. mallei or B. pseudomallei infections, and identifying autophagy manipulation may potentially aid to therapeutic approach for complete clearance of the pathogen.


Subject(s)
Autophagy/genetics , Burkholderia mallei/physiology , Burkholderia pseudomallei/physiology , Desensitization, Immunologic/methods , Glanders/metabolism , Melioidosis/metabolism , Animals , Chymases/metabolism , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteins/genetics , Proteins/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism
6.
Sci Rep ; 8(1): 17074, 2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30451949

ABSTRACT

A correction has been published and is appended to both the HTML and PDF versions of this paper. The error has not been fixed in the paper.

7.
Sci Rep ; 8(1): 2123, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391452

ABSTRACT

ES-62 is a protein secreted by the parasitic worm Acanthocheilonema viteae that is anti-inflammatory by virtue of covalently attached phosphorylcholine. Previously we have reported that drug-like Small Molecule Analogues (SMAs) of its phosphorylcholine moiety can mimic ES-62 in protecting against disease development in certain mouse models of autoimmune and allergic conditions, due to them causing partial degradation of the TLR/IL-1R adaptor MyD88. We have now taken a molecular modelling approach to investigating the mechanism underlying this effect and this predicts that the SMAs interact directly with the MyD88 TIR domain. Further support for this is provided by assay of LPS-induced MyD88/NF-κB-driven secreted alkaline phosphatase (SEAP) reporter activity in commercially-available stably transfected (TLR4-MD2-NF-κB-SEAP) HEK293 cells, as SMA12b-mediated inhibition of such SEAP activity is blocked by its pre-incubation with recombinant MyD88-TIR domain. Direct binding of SMA12b to the TIR domain is also shown to inhibit homo-dimerization of the adaptor, an event that can explain the observed degradation of the adaptor and inhibition of subsequent downstream signalling. Thus, these new data identify initial events by which drug-like ES-62 SMAs, which we also demonstrate are able to inhibit cytokine production by human cells, homeostatically maintain "safe" levels of MyD88 signalling.


Subject(s)
Acanthocheilonema/metabolism , Anti-Inflammatory Agents/pharmacology , Helminth Proteins/pharmacology , Inflammation/drug therapy , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Acanthocheilonema/growth & development , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Protein Domains , Tumor Necrosis Factor-alpha
8.
Chem Biol Drug Des ; 90(6): 1190-1205, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28599094

ABSTRACT

Small molecules were developed to attenuate proinflammatory cytokines resulting from activation of MyD88-mediated toll-like receptor (TLR) signaling by Francisella tularensis. Fifty-three tripeptide derivatives were synthesized to mimic a key BB-loop region involved in toll-like/interleukin-1 receptor recognition (TIR) domain interactions. Compounds were tested for inhibition of TNF-α, IFN-γ, IL-6, and IL-1ß in human peripheral blood mononuclear cells (PBMCs) and primary human bronchial epithelial cells exposed to LPS extracts from F. tularensis. From 53 compounds synthesized and tested, ten compounds were identified as effective inhibitors of F. tularensisLPS-induced cytokines. Compound stability testing in the presence of human liver microsomes and human serum resulted in the identification of tripeptide derivative 7 that was a potent, stable, and drug-like small molecule. Target corroboration using a cell-based reporter assay and competition experiments with MyD88 TIR domain protein supported that the effect of 7 was through MyD88 TIR domain interactions. Compound 7 also attenuated proinflammatory cytokines in human peripheral blood mononuclear cells and bronchial epithelial cells challenged with a live vaccine strain of F. tularensis at a multiplicity of infection of 1:5. Small molecules that target TIR domain interactions in MyD88-dependent TLR signaling represent a promising strategy toward host-directed adjunctive therapeutics for inflammation associated with biothreat agent-induced sepsis.


Subject(s)
Drug Design , Francisella tularensis/metabolism , Myeloid Differentiation Factor 88/metabolism , Peptides/chemistry , Toll-Like Receptors/metabolism , Amino Acid Sequence , Cells, Cultured , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Francisella tularensis/physiology , Genes, Reporter , HEK293 Cells , Half-Life , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Lipopolysaccharides/toxicity , Microsomes, Liver/metabolism , Myeloid Differentiation Factor 88/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , Peptides/metabolism , Peptides/pharmacology , Signal Transduction/drug effects , Toll-Like Receptors/antagonists & inhibitors , Transcriptional Activation/drug effects
9.
Curr Opin Infect Dis ; 30(3): 297-302, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28177960

ABSTRACT

PURPOSE OF REVIEW: Burkholderia mallei is a facultative intracellular pathogen that causes the highly contagious and often the fatal disease, glanders. With its high rate of infectivity via aerosol and recalcitrance toward antibiotics, this pathogen is considered a potential biological threat agent. This review focuses on the most recent literature highlighting host innate immune response to B. mallei. RECENT FINDINGS: Recent studies focused on elucidating host innate immune responses to the novel mechanisms and virulence factors employed by B. mallei for survival. Studies suggest that pathogen proteins manipulate various cellular processes, including host ubiquitination pathways, phagosomal escape, and actin-cytoskeleton rearrangement. Immune-signaling molecules such as Toll-like receptors, nucleotode-binding oligomerization domain, myeloid differentiation primary response protein 88, and proinflammatory cytokines such as interferon-gamma and tumor necrosis factor-α, play key roles in the induction of innate immune responses. Modifications in B. mallei lipopolysaccharide, in particular, the lipid A acyl groups, stimulate immune responses via Toll-like receptor4 activation that may contribute to persistent infection. SUMMARY: Mortality is high because of septicemia and immune pathogenesis with B. mallei exposure. An effective innate immune response is critical to controlling the acute phase of the infection. Both vaccination and therapeutic approaches are necessary for complete protection against B. mallei.


Subject(s)
Burkholderia mallei/immunology , Glanders/immunology , Immunity, Innate , Animals , Burkholderia mallei/pathogenicity , Cytokines/immunology , Glanders/therapy , Humans , Lipopolysaccharides/immunology , Toll-Like Receptors/immunology , Virulence Factors/immunology
10.
Sci Rep ; 5: 14246, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26381092

ABSTRACT

In this study, we used high-throughput computational screening to discover drug-like inhibitors of the host MyD88 protein-protein signaling interaction implicated in the potentially lethal immune response associated with Staphylococcal enterotoxins. We built a protein-protein dimeric docking model of the Toll-interleukin receptor (TIR)-domain of MyD88 and identified a binding site for docking small molecules. Computational screening of 5 million drug-like compounds led to testing of 30 small molecules; one of these molecules inhibits the TIR-TIR domain interaction and attenuates pro-inflammatory cytokine production in human primary cell cultures. Compounds chemically similar to this hit from the PubChem database were observed to be more potent with improved drug-like properties. Most of these 2(nd) generation compounds inhibit Staphylococcal enterotoxin B (SEB)-induced TNF-α, IFN-γ, IL-6, and IL-1ß production at 2-10 µM in human primary cells. Biochemical analysis and a cell-based reporter assay revealed that the most promising compound, T6167923, disrupts MyD88 homodimeric formation, which is critical for its signaling function. Furthermore, we observed that administration of a single dose of T6167923 completely protects mice from lethal SEB-induced toxic shock. In summary, our in silico approach has identified anti-inflammatory inhibitors against in vitro and in vivo toxin exposure with promise to treat other MyD88-related pro-inflammatory diseases.


Subject(s)
Computational Biology/methods , Computer Simulation , Drug Discovery/methods , Myeloid Differentiation Factor 88/chemistry , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Enterotoxins/immunology , Enterotoxins/metabolism , Humans , Inflammation Mediators/metabolism , Inhibitory Concentration 50 , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization/drug effects , Signal Transduction/drug effects
11.
Microb Pathog ; 78: 20-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450887

ABSTRACT

Burkholderia pseudomallei infection causes melioidosis and is often characterized by severe sepsis. Although rare in humans, Burkholderia mallei has caused infections in laboratory workers, and the early innate cellular response to B. mallei in human and nonhuman primates has not been characterized. In this study, we examined the primary cellular immune response to B. mallei in PBMC cultures of non-human primates (NHPs), Chlorocebus aethiops (African Green Monkeys), Macaca fascicularis (Cynomolgus macaque), and Macaca mulatta (Rhesus macaque) and humans. Our results demonstrated that B. mallei elicited strong primary pro-inflammatory cytokines (IFN-γ, TNF-α, IL-1ß, and IL-6) equivalent to the levels of B. pseudomallei in primary PBMC cultures of NHPs and humans. When we examined IL-1ß and other cytokine responses by comparison to Escherichia coli LPS, African Green Monkeys appears to be most responsive to B. mallei than Cynomolgus or Rhesus. Characterization of the immune signaling mechanism for cellular response was conducted by using a ligand induced cell-based reporter assay, and our results demonstrated that MyD88 mediated signaling contributed to the B. mallei and B. pseudomallei induced pro-inflammatory responses. Notably, the induced reporter activity with B. mallei, B. pseudomallei, or purified LPS from these pathogens was inhibited and cytokine production was attenuated by a MyD88 inhibitor. Together, these results show that in the scenario of severe hyper-inflammatory responses to B. mallei infection, MyD88 targeted therapeutic intervention may be a successful strategy for therapy.


Subject(s)
Burkholderia mallei/immunology , Glanders/immunology , Immunity, Innate , Leukocytes, Mononuclear/immunology , Animals , Burkholderia mallei/physiology , Chlorocebus aethiops , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Glanders/genetics , Glanders/microbiology , Humans , Immunity, Cellular , Leukocytes, Mononuclear/microbiology , Macaca fascicularis , Macaca mulatta
12.
Chem Biol Drug Des ; 86(2): 200-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25393063

ABSTRACT

Both Gram-positive and Gram-negative pathogens or pathogen-derived components, such as staphylococcal enterotoxins (SEs) and endotoxin (LPS) exposure, activate MyD88-mediated pro-inflammatory cellular immunity for host defense. However, dysregulated MyD88-mediated signaling triggers exaggerated immune response that often leads to toxic shock and death. Previously, we reported a small molecule compound 1 mimicking BB-loop structure of MyD88 was capable of inhibiting pro-inflammatory response to SEB exposure in mice. In this study, we designed a dimeric structure compound 4210 covalently linked with compound 1 by a non-polar cyclohexane linker which strongly inhibited the production of pro-inflammatory cytokines in human primary cells to SEB (IC50 1-50 µm) or LPS extracted from Francisella tularensis, Escherichia coli, or Burkholderia mallei (IC50 10-200 µm). Consistent with cytokine inhibition, in a ligand-induced cell-based reporter assay, compound 4210 inhibited Burkholderia mallei or LPS-induced MyD88-mediated NF-kB-dependent expression of reporter activity (IC50 10-30 µm). Furthermore, results from a newly expressed MyD88 revealed that 4210 inhibited MyD88 dimer formation which is critical for pro-inflammatory signaling. Importantly, a single administration of compound 4210 in mice showed complete protection from lethal toxin challenge. Collectively, these results demonstrated that compound 4210 inhibits toxin-induced inflated pro-inflammatory immune signaling, thus displays a potential bacterial toxin therapeutic.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Inflammation/drug therapy , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/chemistry , Animals , Anti-Inflammatory Agents/chemical synthesis , Biomimetic Materials/chemical synthesis , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/immunology , Drug Design , Enterotoxins/pharmacology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Shock, Septic/drug therapy , Structure-Activity Relationship
13.
PLoS One ; 7(7): e40773, 2012.
Article in English | MEDLINE | ID: mdl-22848400

ABSTRACT

Staphylococcal enterotoxin B (SEB) exposure triggers an exaggerated pro-inflammatory cytokine response that often leads to toxic shock syndrome (TSS) associated with organ failure and death. MyD88 mediates pro-inflammatory cytokine signaling induced by SEB exposure and MyD88(-/-) mice are resistant to SEB intoxication, suggesting that MyD88 may be a potential target for therapeutic intervention. We targeted the BB loop region of the Toll/IL-1 receptor (TIR) domain of MyD88 to develop small-molecule therapeutics. Here, we report that a synthetic compound (EM-163), mimic to dimeric form of BB-loop of MyD88 attenuated tumor necrosis factor (TNF)- α, interferon (IFN)-γ, interleukin (IL)-1ß, IL-2 and IL-6 production in human primary cells, whether administered pre- or post-SEB exposure. Results from a direct binding assay, and from MyD88 co-transfection/co-immunoprecipitation experiments, suggest that EM-163 inhibits TIR-TIR domain interaction. Additional results indicate that EM-163 prevents MyD88 from mediating downstream signaling. In an NF-kB-driven reporter assay of lipopolysaccharide-stimulated MyD88 signaling, EM-163 demonstrated a dose-dependent inhibition of reporter activity as well as TNF-α and IL-1ß production. Importantly, administration of EM-163 pre- or post exposure to a lethal dose of SEB abrogated pro-inflammatory cytokine responses and protected mice from toxic shock-induced death. Taken together, our results suggest that EM-163 exhibits a potential for therapeutic use against SEB intoxication.


Subject(s)
Biomimetic Materials/pharmacology , Enterotoxins/toxicity , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/drug effects , Animals , Biomimetic Materials/chemistry , Cytokines/genetics , Cytokines/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Signal Transduction/genetics
14.
J Biol Chem ; 286(36): 31385-96, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21693701

ABSTRACT

Toxic shock syndrome (TSS) is a clinical consequence of the profound amplification of host pro-inflammatory cytokine signaling that results from staphylococcal enterotoxin (SE) exposure. We recently reported that MyD88(-/-) mice were resistant to SEA or SEB toxic shock and displayed reduced levels of pro-inflammatory cytokines in their serum. Here we report that SEB stimulation of total mononuclear cells up-regulated MyD88 in monocytes and T cells. Further, MyD88 gene silencing in primary human cells using siRNA prevented SEB or SEB plus lipopolysaccharide (LPS) induction of interleukin-1ß (IL-1ß) transcriptional activation, suggesting that MyD88-mediated signaling is an essential component of SEB toxicity. We synthesized small molecules that mimic the conserved BB-loop in the Toll/IL-1 receptor (TIR) domain of MyD88. In primary human cells, these mimetics attenuated SEB-induced pro-inflammatory cytokine production. SEB stimulation of primary cells with mimetic affected newly synthesized MyD88 and downstream signaling components. Furthermore, LPS-induced MyD88 signaling was likewise inhibited in a cell-based reporter assay. More importantly, administration of mimetic reduced cytokine responses and increased survivability in a murine SEB challenge model. Collectively, these results suggest that MyD88 BB-loop mimetics interfere with SEB-induced pro-inflammatory signaling and toxicity, thus offering a potential approach in the therapy of toxic shock.


Subject(s)
Cytokines/biosynthesis , Enterotoxins/antagonists & inhibitors , Molecular Mimicry , Myeloid Differentiation Factor 88/chemistry , Peptides/pharmacology , Shock, Septic/drug therapy , Animals , Cells, Cultured , Enterotoxins/toxicity , Humans , Inflammation/prevention & control , Mice , Mice, Knockout , Peptides/chemistry , Peptides/therapeutic use , Receptors, Interleukin-1/chemistry
15.
PLoS One ; 6(1): e15985, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21283748

ABSTRACT

Ligands binding to Toll-like receptor (TLR), interleukin 1 receptor (IL-1R), or IFN-γR1 are known to trigger MyD88-mediated signaling, which activates pro-inflammatory cytokine responses. Recently we reported that staphylococcal enterotoxins (SEA or SEB), which bind to MHC class II molecules on APCs and cross link T cell receptors, activate MyD88- mediated pro-inflammatory cytokine responses. We also reported that MyD88(-/-) mice were resistant to SE- induced toxic shock and had reduced levels of serum cytokines. In this study, we investigated whether MHC class II- SE interaction by itself is sufficient to activate MyD88 in MHC class II(+) cells and induce downstream pro-inflammatory signaling and production of cytokines such as TNF-α and IL-1ß. Here we report that human monocytes treated with SEA, SEB, or anti-MHC class II monoclonal antibodies up regulated MyD88 expression, induced activation of NF-kB, and increased expression of IL-1R1 accessory protein, TNF-α and IL-1ß. MyD88 immunoprecipitated from cell extracts after SEB stimulation showed a greater proportion of MyD88 phosphorylation compared to unstimulated cells indicating that MyD88 was a component of intracellular signaling. MyD88 downstream proteins such as IRAK4 and TRAF6 were also up regulated in monocytes after SEB stimulation. In addition to monocytes, primary B cells up regulated MyD88 in response to SEA or SEB stimulation. Importantly, in contrast to primary B cells, MHC class II deficient T2 cells had no change of MyD88 after SEA or SEB stimulation, whereas MHC class II-independent activation of MyD88 was elicited by CpG or LPS. Collectively, these results demonstrate that MHC class II utilizes a MyD88-mediated signaling mechanism when in contact with ligands such as SEs to induce pro-inflammatory cytokines.


Subject(s)
Enterotoxins/metabolism , Histocompatibility Antigens Class II/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/immunology , Animals , B-Lymphocytes , Cytokines/biosynthesis , Enterotoxins/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/immunology , Humans , Inflammation , Mice , Mice, Knockout , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , Protein Binding/immunology
16.
Innate Immun ; 17(5): 451-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20699281

ABSTRACT

An elevated pro-inflammatory cytokine response is the primary cause of death by toxic shock after exposure to staphylococcal enterotoxin B (SEB). Identifying an intracellular signal mediator that predominantly controls the pro-inflammatory response is important for developing a therapeutic strategy. We examined the role of the signaling adaptor MyD88 in cell culture and in a mouse model of toxic shock. Our results indicated that elevated tumor necrosis factor-α, interferon-γ, interleukin (IL)-1α/ß and IL-6 production from mouse spleen cells treated with SEB alone or in combination with lipopolysaccharide (LPS) was regulated by MyD88. Elevated levels of MyD88 protein in spleen cells, as well as in CD11c(+) or Mac3(+) cells, and activation of nuclear factor-κB in spleen cells were observed in mice treated with SEB. An SEB-dose dependent lethality was observed in LPS-potentiated and in D-galactosamine-sensitized mice. D-Galactosamine treatment of spleen cells had no effect in cytokine induction but rather increased the sensitivity to toxic shock in mice. Our results demonstrated an impaired pro-inflammatory cytokine production by spleen cells of MyD88(-/-) mice in response to SEB or SEB plus LPS. Most importantly, MyD88(-/-) mice were resistant to SEB-induced death. These results demonstrate that MyD88-dependent pro-inflammatory signaling is responsible for SEB intoxication. In addition, our studies also demonstrated that LPS potentiation, in comparison to D-galactosamine sensitization, contributes to a stronger SEB-induced lethality. This is due to the pro-inflammatory cytokine response elicited by MyD88 after exposure to SEB and LPS. These findings offer an important insight upon SEB intoxication and subsequent therapy targeting MyD88.


Subject(s)
Enterotoxins/administration & dosage , Myeloid Differentiation Factor 88/metabolism , Shock, Septic/immunology , Spleen/metabolism , Staphylococcal Infections/immunology , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Enterotoxins/adverse effects , Female , Galactosamine/administration & dosage , Gene Expression Regulation , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Shock, Septic/chemically induced , Shock, Septic/etiology , Spleen/immunology , Spleen/microbiology , Spleen/pathology , Staphylococcal Infections/complications
17.
Immunology ; 130(4): 516-26, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20465563

ABSTRACT

Staphylococcal enterotoxin (SE) -induced toxic shock is triggered by inflammatory cytokine signal amplification after SE binding to major histocompatibility complex class II molecules on antigen-presenting cells and T-cell receptors. Identifying host cellular elements contributing to this pro-inflammatory signal amplification is critical for developing a strategy for therapeutic intervention. Myeloid differentiation primary-response protein 88 (MyD88) is an intracellular signalling adaptor protein primarily known for mediating pro-inflammatory cytokine responses. We investigated the role of MyD88 in staphylococcal enterotoxin A (SEA) -treated cell cultures and mouse models of toxic shock. Our results demonstrated that elevated levels of tumour necrosis factor-alpha, interferon-gamma, interleukin-1alpha/beta (IL-1alpha/beta), IL-2 and IL-6 production correlated with up-regulation of MyD88 after treatment of spleen cells and mice with SEA alone or in combination with lipopolysaccharide (LPS). The SEA-induced lethality was also observed in (LPS-independent) D-galactosamine-sensitized mice. While LPS potentiated SEA-induced cytokine responses, D-galactosamine treatment had no additive effect. Most importantly, our results demonstrated that MyD88(-/-) mice were resistant to SEA-induced toxic shock and had reduced pro-inflammatory cytokine responses. These results suggest that SEA-induced lethality is primarily dependent on MyD88. Our findings offer an important insight on potential therapeutic treatment of SEA-induced toxic shock targeting MyD88.


Subject(s)
Cytokines/immunology , Enterotoxins/toxicity , Myeloid Differentiation Factor 88/immunology , Staphylococcus/immunology , Animals , Cells, Cultured , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , NF-kappa B/immunology , NF-kappa B/metabolism , Spleen/immunology , Staphylococcus/drug effects
18.
Clin Vaccine Immunol ; 15(1): 131-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18045883

ABSTRACT

Interleukin-15 (IL-15) is generally considered to sustain T-cell memory and to be a growth factor for natural killer cells. Previous data from our laboratory demonstrated that IL-15 is also an important factor for developing human dendritic cells. For this study, we investigated the effects of IL-15 on antibody responses in mice to a recombinant staphylococcal enterotoxin B (SEB) vaccine (STEBVax) in a preclinical model of toxic shock syndrome induced by SEB. We observed that mouse spleen cells treated with IL-15 in ex vivo culture gained a dendritic cell-like phenotype. Administration of IL-15 to mice also resulted in an increased number of mature CD11c+ dendritic cells in mouse spleens. A significant, IL-15 dose-dependent increase in antigen-specific antibody was observed after coadministration with the vaccine and an aluminum-based adjuvant (alhydrogel). Furthermore, the coadministration of IL-15 with STEBVax and alhydrogel also protected mice from lethal toxic shock above the levels that obtained without IL-15. Thus, the vaccine response enhanced by IL-15 appears to be mediated by mature dendritic cells and results in prevalent seroconversion to Th2-dependent antibodies. This suggests a potential use of IL-15 as an adjuvant for antibody-dependent responses to vaccines.


Subject(s)
Bacterial Vaccines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Enterotoxins/immunology , Interleukin-15/pharmacology , Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibody Formation , Bacterial Vaccines/therapeutic use , CD11c Antigen/biosynthesis , CD11c Antigen/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/cytology , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Interleukin-15/immunology , Male , Mice , Mice, Inbred BALB C , Shock, Septic/immunology , Shock, Septic/prevention & control , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Staphylococcus aureus/immunology , Th2 Cells/immunology , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use
19.
J Infect Dis ; 194(12): 1753-60, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17109349

ABSTRACT

Cell contact by the plague bacterium Yersinia pestis initiates the injection of several virulence factors that target biochemical pathways critical for host clearance of bacteria. Despite this impairment of innate immunity, it is unclear whether antigen recognition by T cells is equally affected. We present evidence that human cytolytic T cells respond to Y. pestis virulence proteins presented by infected monocytes and dendritic cells. These T cell antigens consisted of a panel of proteins encoded by pCD1, a 70-kDa plasmid that harbors virulence factors and transport proteins of the cell contact-dependent, type III secretion system. Infected cells retained the ability to process and present tetanus toxoid to T cells, which indicates that responses to unrelated antigens were also maintained. Our results indicate that T cell immunity remains functional during Y. pestis infection, which thus suggests the potential benefits of therapeutic vaccination and strategies that emphasize the inclusion of cytotoxic T lymphocyte responses.


Subject(s)
Immunity, Innate , Plague/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Humans , Leukocytes, Mononuclear , Molecular Weight , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Plasmids/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Tetanus Toxoid/immunology , Transcription Factors/chemistry , Transcription Factors/immunology , Virulence Factors/immunology , Yersinia pestis
20.
J Infect Dis ; 191(2): 278-88, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15609239

ABSTRACT

A new anthrax vaccine under clinical investigation is based on recombinant Bacillus anthracis protective antigen (rPA). Here, we investigated microneedle-based cutaneous and nasal mucosal delivery of rPA in mice and rabbits. In mice, intradermal (id) delivery achieved up to 90% seroconversion after a single dose, compared with 20% after intramuscular (im) injection. Intranasal (inl) delivery of a liquid formulation required 3 doses to achieve responses that were comparable with those achieved via the id or im routes. In rabbits, id delivery provided complete protection against aerosol challenge with anthrax spores; in addition, novel powder formulations administered inl provided complete protection, whereas a liquid formulation provided only partial protection. These results demonstrate, for the first time, that cutaneous or nasal mucosal administration of rPA provides complete protection against inhalational anthrax in rabbits. The novel vaccine/device combinations described here have the potential to improve the efficacy of rPA and other biodefense vaccines.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax/prevention & control , Antigens, Bacterial/immunology , Vaccination , Administration, Cutaneous , Administration, Intranasal , Animals , Anthrax Vaccines/immunology , Antibodies, Bacterial/blood , Dose-Response Relationship, Immunologic , Drug Delivery Systems , Mice , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...