Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 7(1): 244, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511088

ABSTRACT

BACKGROUND: Immune checkpoint blockade (ICB) promotes adaptive immunity and tumor regression in some cancer patients. However, in patients with immunologically "cold" tumors, tumor-resident innate immune cell activation may be required to prime an adaptive immune response and so exploit the full potential of ICB. Whilst Toll-like receptor (TLR) agonists have been used topically to successfully treat some superficial skin tumors, systemic TLR agonists have not been well-tolerated. METHODS: The response of human immune cells to TLR7 and 8 agonism was measured in primary human immune cell assays. MEDI9197 (3M-052) was designed as a novel lipophilic TLR7/8 agonist that is retained at the injection site, limiting systemic exposure. Retention of the TLR7/8 agonist at the site of injection was demonstrated using quantitative whole-body autoradiography, HPLC-UV, and MALDI mass spectrometry imaging. Pharmacodynamic changes on T cells from TLR7/8 agonist treated B16-OVA tumors was assessed by histology, quantitative real time PCR, and flow cytometry. Combination activity of TLR7/8 agonism with immunotherapies was assessed in vitro by human DC-T cell MLR assay, and in vivo using multiple syngeneic mouse tumor models. RESULTS: Targeting both TLR7 and 8 triggers an innate and adaptive immune response in primary human immune cells, exemplified by secretion of IFNα, IL-12 and IFNγ. In contrast, a STING or a TLR9 agonist primarily induces release of IFNα. We demonstrate that the TLR7/8 agonist, MEDI9197, is retained at the sight of injection with limited systemic exposure. This localized TLR7/8 agonism leads to Th1 polarization, enrichment and activation of natural killer (NK) and CD8+ T cells, and inhibition of tumor growth in multiple syngeneic models. The anti-tumor activity of this TLR7/8 agonist is enhanced when combined with T cell-targeted immunotherapies in pre-clinical models. CONCLUSION: Localized TLR7/8 agonism can enhance recruitment and activation of immune cells in tumors and polarize anti-tumor immunity towards a Th1 response. Moreover, we demonstrate that the anti-tumor effects of this TLR7/8 agonist can be enhanced through combination with checkpoint inhibitors and co-stimulatory agonists.


Subject(s)
Dendritic Cells/immunology , Heterocyclic Compounds, 3-Ring/pharmacology , Killer Cells, Natural/immunology , Melanoma, Experimental/drug therapy , Stearic Acids/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Tumor Microenvironment/immunology , Adaptive Immunity , Adjuvants, Immunologic/pharmacology , Animals , Apoptosis , Cell Proliferation , Female , Humans , Immunotherapy , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Rats, Sprague-Dawley , Tumor Cells, Cultured
2.
J Immunol ; 198(11): 4413-4424, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28468970

ABSTRACT

Mature peripheral double negative T (DNT) cells expressing αß TCR but lacking CD4/CD8 coreceptors play protective as well as pathogenic roles. To better understand their development and functioning in vivo, we concomitantly inactivated CD4 and CD8 genes in mice with intact MHC class I and class II molecules with the hypothesis that this would enable the development of DNT cells. We also envisaged that these DNT cells could be activated by bacterial superantigens in vivo as activation of T cells by superantigens does not require CD4 and CD8 coreceptors. Because HLA class II molecules present superantigens more efficiently than murine MHC class II molecules, CD4 CD8 double knockout (DKO) mice transgenically expressing HLA-DR3 or HLA-DQ8 molecules were generated. Although thymic cellularity was comparable between wild type (WT) and DKO mice, CD3+ αß TCR+ thymocytes were significantly reduced in DKO mice, implying defects in thymic-positive selection. Splenic CD3+ αß TCR+ cells and Foxp3+ T regulatory cells were present in DKO mice but significantly reduced. However, the in vivo inflammatory responses and immunopathology elicited by acute challenge with the staphylococcal superantigen enterotoxin B were comparable between WT and DKO mice. Choric exposure to staphylococcal enterotoxin B precipitated a lupus-like inflammatory disease with characteristic lympho-monocytic infiltration in lungs, livers, and kidneys, along with production of anti-nuclear Abs in DKO mice as in WT mice. Overall, our results suggest that DNT cells can develop efficiently in vivo and chronic exposure to bacterial superantigens may precipitate a lupus-like autoimmune disease through activation of DNT cells.


Subject(s)
CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Enterotoxins/immunology , Superantigens/immunology , T-Lymphocyte Subsets/immunology , Animals , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/cytology , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/immunology
3.
J Pharm Sci ; 106(8): 2037-2045, 2017 08.
Article in English | MEDLINE | ID: mdl-28456734

ABSTRACT

Toll-like receptor (TLR) agonists TLR 7/8, MEDI9197, is a imidazoquinoline analogue that can be used for cancer immunotherapy based on its efficacy toward a variety of tumors. Systemic administration of TLR agonists results in stimulation of the immune system throughout the entire body causing undesirable side effects. To minimize these adverse events, local administration of TLR agonists including intratumoral (IT) delivery has been introduced. Here, a poloxamer 407 thermogel formulation for IT delivery of a TLR 7/8 dual agonist, MEDI9197, is described in which the combination of the aqueous thermogel and the ethanolic TLR 7/8 dual agonist, MEDI9197, solution leads to precipitated drug particles within the gel. The in vitro release profile showed an initial burst followed by sustained release. A B16-OVA mouse tumor model was used to assess the in vivo pharmacokinetics, efficacy, and systemic cytokine and chemokine (cytokine) production of the poloxamer 407-based thermogel formulation. The pharmacokinetic evaluation showed that the agonist level within the tumor was reduced by ∼70% over 14 days while serum agonist levels indicated an initial burst at the 6-h time point followed by a drop in serum drug levels over the 14 days of the experiment. The tumor growth inhibition, survival, and serum cytokines for post-IT injection of the poloxamer 407 formulation showed that it slowly released TLR 7/8 agonist, MEDI9197, resulting in more efficacious tumor growth inhibition compared with control groups. In addition, the cytokine levels in circulation indicated that a dose increase led to a decrease in the serum inflammatory and interferon-inducible cytokines levels. This observation could be due to a reduction of drug diffusion and escape from the tumor site due to the precipitation of the drug inside the tumor leading to sustained release. IT delivery of TLR 7/8 dual agonist, MEDI9197, via a thermosensitive gel-based formulation was efficacious and could offer an alternate method of local drug delivery.


Subject(s)
Antineoplastic Agents/administration & dosage , Delayed-Action Preparations/chemistry , Heterocyclic Compounds, 3-Ring/administration & dosage , Melanoma, Experimental/drug therapy , Poloxamer/chemistry , Stearic Acids/administration & dosage , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cytokines/blood , Cytokines/immunology , Drug Delivery Systems , Female , Gels/chemistry , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Melanoma, Experimental/blood , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Stearic Acids/pharmacokinetics , Stearic Acids/therapeutic use , Temperature , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology
4.
Toxins (Basel) ; 7(12): 5308-19, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26670252

ABSTRACT

Staphylococcus aureus, the most common cause of wound infection, produces several exotoxins, including superantigens (SAgs). SAgs are the potent activators of the immune system. Given this unique property, we hypothesized that SAgs produced by S. aureus in wounds would have local, as well as systemic immunologic effects. We tested our hypothesis using a novel staphylococcal skin wound infection model in transgenic mice expressing HLA-DR3. Skin wounds were left uninfected or colonized with S. aureus strains producing SAgs or an isogenic strain not producing any SAg. Animals with wounds challenged with SAg-producing S. aureus had increased morbidity and lower serum IL-17 levels compared to those challenged with the SAg non-producing S. aureus (p = 0.027 and p = 0.032, respectively). At Day 8 following microbial challenge, compared to mice with uninfected wounds, the proportion of Vß8⁺CD4⁺ T cells was increased, while the proportion of Vß8⁺CD8⁺ T cells was decreased only in the spleens of mice challenged with SAg-producing S. aureus (p < 0.001). No such changes were measured in mice challenged with SAg non-producing S. aureus. Lungs, livers and kidneys from mice challenged with SAg-producing, but not SAg non-producing, S. aureus showed inflammatory changes. Overall, SAg-mediated systemic immune activation in wounds harboring S. aureus may have clinical implications.


Subject(s)
Antigens, Bacterial/immunology , Skin Diseases/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Wound Healing/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-17/blood , Kidney/pathology , Liver/pathology , Lung/pathology , Mice, Transgenic , Skin/immunology , Skin/injuries , Skin Diseases/blood , Staphylococcal Infections/blood
5.
J Immunol ; 195(10): 4660-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26475924

ABSTRACT

MHC, especially HLA-DR3 and HLA-DR2, is one of the most important genetic susceptibility regions for systemic lupus erythematosus. Human studies to understand the role of specific HLA alleles in disease pathogenesis have been hampered by the presence of strong linkage disequilibrium in this region. To overcome this, we produced transgenic mice expressing HLA-DR3 (DRß1*0301) and devoid of endogenous class II (both I-A and I-E genes, AE(0)) on a lupus-prone NZM2328 background (NZM2328.DR3(+)AE(0)). Both NZM2328 and NZM2328.DR3(+)AE(0) mice developed anti-dsDNA and glomerulonephritis, but anti-dsDNA titers were higher in the latter. Although kidney histological scores were similar in NZM2328 and NZM2328.DR3(+)AE(0) mice (7.2 ± 4.3 and 8.6 ± 5.7, respectively, p = 0.48), the onset of severe proteinuria occurred earlier in NZM2328.DR3(+)AE(0) mice compared with NZM2328 mice (median, 5 and 9 mo respectively, p < 0.001). Periarterial lymphoid aggregates, classic wire loop lesions, and occasional crescents were seen only in kidneys from NZM2328.DR3(+)AE(0) mice. Interestingly, NZM2328.DR3(+)AE(0) mice, but not NZM2328 mice, spontaneously developed anti-Smith (Sm) Abs. The anti-Sm Abs were seen in NZM2328.DR3(+)AE(0) mice that were completely devoid of endogenous class II (AE(-/) (-)) but not in mice homozygous (AE(+/+)) or heterozygous (AE(+/-)) for endogenous MHC class II. It appears that only HLA-DR3 molecules can preferentially select SmD-reactive CD4(+) T cells for generation of the spontaneous anti-Sm immune response. Thus, our mouse model unravels a critical role for HLA-DR3 in generating an autoimmune response to SmD and lupus nephritis in the NZM2328 background.


Subject(s)
Antibodies, Antinuclear/immunology , Glomerulonephritis/immunology , HLA-DR3 Antigen/immunology , Lupus Nephritis/immunology , snRNP Core Proteins/immunology , Animals , Antibodies, Antinuclear/genetics , CD4-Positive T-Lymphocytes/immunology , DNA/immunology , Disease Models, Animal , Genetic Predisposition to Disease , Glomerulonephritis/genetics , HLA-DR2 Antigen/immunology , HLA-DR3 Antigen/genetics , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/genetics , Mice , Mice, Knockout
6.
J Leukoc Biol ; 98(2): 271-81, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25979434

ABSTRACT

SAgs, produced by Staphylococcus aureus, play a major role in the pathogenesis of invasive staphylococcal diseases by inducing potent activation of the immune system. However, the role of SAgs, produced by S. aureus, associated with indwelling devices or tissues, are not known. Given the prevalence of device-associated infection with toxigenic S. aureus in clinical settings and the potency of SAgs, we hypothesized that continuous exposure to SAgs produced by catheter-associated S. aureus could have systemic consequences. To investigate these effects, we established a murine in vivo catheter colonization model. One centimeter long intravenous catheters were colonized with a clinical S. aureus isolate producing SAgs or isogenic S. aureus strains, capable or incapable of producing SAg. Catheters were subcutaneously implanted in age-matched HLA-DR3, B6, and AE(o) mice lacking MHC class II molecules and euthanized 7 d later. There was no evidence of systemic infection. However, in HLA-DR3 transgenic mice, which respond robustly to SSAgs, the SSAg-producing, but not the nonproducing strains, caused a transient increase in serum cytokine levels and a protracted expansion of splenic CD4(+) T cells expressing SSAg-reactive TCR Vß8. Lungs, livers, and kidneys from these mice showed infiltration with CD4(+) and CD11b(+) cells. These findings were absent in B6 and AE(o) mice, which are known to respond poorly to SSAgs. Overall, our novel findings suggest that systemic immune activation elicited by SAgs, produced by S. aureus colonizing foreign bodies, could have clinical consequences in humans.


Subject(s)
Catheter-Related Infections/immunology , Enterotoxins/biosynthesis , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , Superantigens/biosynthesis , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , Catheter-Related Infections/genetics , Catheter-Related Infections/microbiology , Catheter-Related Infections/pathology , Catheters, Indwelling , Enterotoxins/immunology , Gene Deletion , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Humans , Kidney/immunology , Kidney/microbiology , Kidney/pathology , Liver/immunology , Liver/microbiology , Liver/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/immunology , Spleen/microbiology , Spleen/pathology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Superantigens/immunology
7.
Diagn Microbiol Infect Dis ; 81(3): 201-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25619753

ABSTRACT

Staphylococcus aureus is a common cause of prosthetic joint infection (PJI). The prevalence of superantigens (SAgs) among PJI-associated S. aureus is unknown. Eighty-four S. aureus isolates associated with PJI isolated between 1999 and 2006 were studied. SAg genes, sea, seb, sec, sed, see, seg, seh, sei, and tst, were assayed by PCR. Seventy-eight (92.9%) isolates carried at least 1 SAg gene studied, with 61 (72.6%) harboring more than 1. seg was most commonly (70.2%), and seh was least frequently (4.8%) detected. tst-positive isolates were associated with early infection and increased erythrocyte sedimentation rate at diagnosis (P=0.006 and P=0.021, respectively). seg and sei were associated with methicillin resistance (P=0.008 and P=0.002, respectively). A majority of PJI-associated isolates studied produced biologically active SAgs in both planktonic and biofilm growth modes. SAg genes are prevalent in S. aureus causing PJI.


Subject(s)
Arthritis/microbiology , Prosthesis-Related Infections/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Superantigens/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Genotype , Humans , Leukocytes, Mononuclear/drug effects , Male , Mice, Transgenic , Middle Aged , Polymerase Chain Reaction , Staphylococcus aureus/isolation & purification , Superantigens/analysis
8.
J Immunol ; 193(6): 2919-30, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25092888

ABSTRACT

Life-threatening infections caused by Staphylococcus aureus, particularly the community-acquired methicillin-resistant strains of S. aureus, continue to pose serious problems. Greater virulence and increased pathogenicity of certain S. aureus strains are attributed to higher prevalence of exotoxins. Of these exotoxins, the superantigens (SAg) are likely most pathogenic because of their ability to rapidly and robustly activate the T cells even in extremely small quantities. Therefore, countering SAg-mediated T cell activation using T regulatory cells (Tregs) might be beneficial in diseases such as toxic shock syndrome (TSS). As the normal numbers of endogenous Tregs in a typical host are insufficient, we hypothesized that increasing the Treg numbers by administration of IL-2/anti-IL-2 Ab immune complexes (IL2C) or by adoptive transfer of ex vivo expanded Tregs might be more effective in countering SAg-mediated immune activation. HLA-DR3 transgenic mice that closely recapitulate human TSS were treated with IL2C to increase endogenous Tregs or received ex vivo expanded Tregs. Subsequently, they were challenged with SAg to induce TSS. Analyses of various parameters reflective of TSS (serum cytokine/chemokine levels, multiple organ pathology, and SAg-induced peripheral T cell expansion) indicated that increasing the Tregs failed to mitigate TSS. On the contrary, serum IFN-γ levels were increased in IL2C-treated mice. Exploration into the reasons behind the lack of protective effect of Tregs revealed IL-17 and IFN-γ-dependent loss of Tregs during TSS. In addition, significant upregulation of glucocorticoid-induced TNFR family-related receptor on conventional T cells during TSS could render them resistant to Treg-mediated suppression, contributing to failure of Treg-mediated immune regulation.


Subject(s)
Enterotoxins/immunology , Shock, Septic/immunology , Superantigens/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Adoptive Transfer , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/biosynthesis , Glucocorticoids , HLA-DR alpha-Chains/genetics , HLA-DR alpha-Chains/immunology , HLA-DR beta-Chains/genetics , HLA-DR beta-Chains/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Transgenic , Receptors, Tumor Necrosis Factor/biosynthesis , Shock, Septic/microbiology , Staphylococcal Infections/immunology , Up-Regulation
9.
Mediators Inflamm ; 2014: 468285, 2014.
Article in English | MEDLINE | ID: mdl-25024509

ABSTRACT

Staphylococcus aureus is capable of causing a spectrum of human illnesses. During serious S. aureus infections, the staphylococcal pathogen-associated molecular patterns (PAMPs) such as peptidoglycan, lipoteichoic acid, and lipoproteins and even intact S. aureus, are believed to act in conjunction with the staphylococcal superantigens (SSAg) to activate the innate and adaptive immune system, respectively, and cause immunopathology. However, recent studies have shown that staphylococcal PAMPs could suppress inflammation by several mechanisms and protect from staphylococcal toxic shock syndrome, a life-threatening systemic disease caused by toxigenic S. aureus. Given the contradictory pro- and anti-inflammatory roles of staphylococcal PAMPs, we examined the effects of S. aureus-derived molecular patterns on immune responses driven by SSAg in vivo using HLA-DR3 and HLA-DQ8 transgenic mice. Our study showed that neither S. aureus-derived peptidoglycans (PGN), lipoteichoic acid (LTA), nor heat-killed Staphylococcus aureus (HKSA) inhibited SSAg-induced T cell proliferation in vitro. They failed to antagonize the immunostimulatory effects of SSAg in vivo as determined by their inability to attenuate systemic cytokine/chemokine response and reduce SSAg-induced T cell expansion. These staphylococcal PAMPs also failed to protect HLA-DR3 as well as HLA-DQ8 transgenic mice from either SSAg-induced toxic shock or pneumonia induced by a SSAg-producing strain of S. aureus.


Subject(s)
Pneumonia/immunology , Pneumonia/metabolism , Shock, Septic/immunology , Shock, Septic/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Superantigens/immunology , Animals , HLA-DQ Antigens/genetics , HLA-DR3 Antigen/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Transgenic , Pneumonia/chemically induced , Shock, Septic/chemically induced
10.
Diagn Microbiol Infect Dis ; 79(2): 119-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24745820

ABSTRACT

The frequency of superantigen production among Staphylococcus aureus isolates associated with endocarditis is not well defined. We tested 154 S. aureus isolates from definite infective endocarditis cases for the presence of staphylococcal enterotoxins A-E, H, and TSST-1 by PCR, enzyme-linked immunosorbent assay, and using an HLA-DR3 transgenic mouse splenocyte proliferation assay. Sixty-three isolates (50.8%) tested positive for at least 1 superantigen gene, with 21 (16.9%) testing positive for more than 2. tst (28.6%) was most common, followed by seb (27%), sea (22.2%), sed (20.6%), see (17.5%), and sec (11.1%). Of 41 methicillin-resistant S. aureus, 21 had superantigen genes, with sed being more frequently detected in this group compared to methicillin-susceptible S. aureus (P < 0.05). Superantigen genes were not associated with mortality (P = 0.81). 75% of PCR-positive isolates induced robust splenocyte proliferation. Overall, more than half of S. aureus isolates causing endocarditis carry superantigen genes, of which most are functional.


Subject(s)
Endocarditis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Superantigens/analysis , Superantigens/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , Leukocytes, Mononuclear/immunology , Male , Mice, Transgenic , Middle Aged , Polymerase Chain Reaction , Young Adult
11.
PLoS One ; 8(11): e78687, 2013.
Article in English | MEDLINE | ID: mdl-24236037

ABSTRACT

Human and animal studies strongly suggest that dietary gluten could play a causal role in the etiopathogenesis of type 1 diabetes (T1D). However, the mechanisms have not been elucidated. Recent reports indicate that the intestinal microbiome has a major influence on the incidence of T1D. Since diet is known to shape the composition of the intestinal microbiome, we investigated using non-obese diabetic (NOD) mice whether changes in the intestinal microbiome could be attributed to the pro- and anti-diabetogenic effects of gluten-containing and gluten-free diets, respectively. NOD mice were raised on gluten-containing chows (GCC) or gluten-free chows (GFC). The incidence of diabetes was determined by monitoring blood glucose levels biweekly using a glucometer. Intestinal microbiome composition was analyzed by sequencing 16S rRNA amplicons derived from fecal samples. First of all, GCC-fed NOD mice had the expected high incidence of hyperglycemia whereas NOD mice fed with a GFC had significantly reduced incidence of hyperglycemia. Secondly, when the fecal microbiomes were compared, Bifidobacterium, Tannerella, and Barnesiella species were increased (p = 0.03, 0.02, and 0.02, respectively) in the microbiome of GCC mice, where as Akkermansia species was increased (p = 0.02) in the intestinal microbiomes of NOD mice fed GFC. Thirdly, both of the gluten-free chows that were evaluated, either egg white based (EW-GFC) or casein based (C-GFC), significantly reduced the incidence of hyperglycemia. Interestingly, the gut microbiome from EW-GFC mice was similar to C-GFC mice. Finally, adding back gluten to the gluten-free diet reversed its anti-diabetogenic effect, reduced Akkermansia species and increased Bifidobacterium, Tannerella, and Barnesiella suggesting that the presence of gluten is directly responsible for the pro-diabetogenic effects of diets and it determines the gut microflora. Our novel study thus suggests that dietary gluten could modulate the incidence of T1D by changing the gut microbiome.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diet, Gluten-Free , Glutens/adverse effects , Intestines/microbiology , Microbiota , Animals , Autoantibodies/blood , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Dietary Proteins/adverse effects , Feces/microbiology , Female , Humans , Immunoglobulin G/blood , Incidence , Insulin/immunology , Male , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/immunology
12.
Antimicrob Agents Chemother ; 56(10): 5401-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22850509

ABSTRACT

Superantigens (SAg), the potent activators of the immune system, are important determinants of Staphylococcus aureus virulence and pathogenicity. Superior response to SAg in human leukocyte antigen (HLA)-DR3 transgenic mice rendered them more susceptible than C57BL/6 mice to pneumonia caused by SAg-producing strains of S. aureus. Linezolid, a bacterial protein synthesis inhibitor, was superior to vancomycin in inhibiting SAg production by S. aureus in vitro and conferred greater protection from pneumonia caused by SAg-producing staphylococci.


Subject(s)
Acetamides/therapeutic use , Oxazolidinones/therapeutic use , Pneumonia, Bacterial/drug therapy , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Superantigens/metabolism , Vancomycin/therapeutic use , Animals , Linezolid , Mice , Mice, Inbred BALB C , Mice, Transgenic
13.
J Immunol ; 189(4): 2054-62, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22798666

ABSTRACT

Chronic nasal and skin colonization with superantigen (SAg)-producing Staphylococcus aureus is well documented in humans. Given that trans-mucosal and trans-cutaneous absorption of SAgs can occur, we determined whether chronic exposure to small amounts of SAg per se could activate autoreactive CD4(+) and CD8(+) T cells and precipitate any autoimmune disease without further external autoantigenic stimulation. Because HLA class II molecules present SAg more efficiently than do mouse MHC class II molecules, HLA-DQ8 transgenic mice were implanted s.c. with mini-osmotic pumps capable of continuously delivering the SAg, staphylococcal enterotoxin B (total of 10 µg/mouse), or PBS over 4 wk. Chronic exposure to staphylococcal enterotoxin B resulted in a multisystem autoimmune inflammatory disease with features similar to systemic lupus erythematosus. The disease was characterized by mononuclear cell infiltration of lungs, liver, and kidneys, accompanied by the production of anti-nuclear Abs and deposition of immune complexes in the renal glomeruli. The inflammatory infiltrates in various organs predominantly consisted of CD4(+) T cells bearing TCR Vß8. The extent of immunopathology was markedly reduced in mice lacking CD4(+) T cells and CD28, indicating that the disease is CD4(+) T cell mediated and CD28 dependent. The absence of disease in STAT4-deficient, as well as IFN-γ-deficient, HLA-DQ8 mice suggested the pathogenic role of Th1-type cytokines, IL-12 and IFN-γ. In conclusion, our study suggests that chronic exposure to extremely small amounts of bacterial SAg could be an etiological factor for systemic lupus erythematosus.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Superantigens/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/pathology , CD28 Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , HLA-DQ Antigens/immunology , Humans , Infusions, Subcutaneous , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Transgenic , Staphylococcus aureus/immunology , Superantigens/administration & dosage
14.
Microbes Infect ; 14(6): 528-36, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22273732

ABSTRACT

Staphylococcal superantigens (SAg) are a family of potent exotoxins produced by Staphylococcus aureus. They play an important role in the pathogenesis of staphylococcal shock and pneumonia by causing a robust activation of the immune system and eliciting a strong surge in systemic cytokine and chemokine levels. Given the biological functions of SAg, we evaluated the efficacy of tacrolimus, a potent immunosuppressive agent, in the prophylaxis and therapy of staphylococcal TSS and pneumonia using human leukocyte antigen (HLA)-DR3 transgenic mice. Tacrolimus significantly inhibited staphylococcal SAg induced T cell activation in vitro. In vivo, tacrolimus significantly suppressed the SAg-induced elevation in serum cytokine and chemokine levels when given prophylactically, when administered immediately or even 2 h following systemic SAg challenge. Paradoxically, neither the prophylactic nor post-exposure treatment with tacrolimus protected mice from lethal SAg-induced TSS. A closer examination revealed that tacrolimus failed to suppress SAg-induced T cell proliferation and systemic pathology, including gut dysfunction. Tacrolimus also failed to protect from lethal pneumonia induced by a SAg-producing S. aureus strain. Thus, our study showed that even though T cell activation by SAg plays a major role in the immunopathogenesis of TSS and pneumonia, tacrolimus alone has no beneficial effect.


Subject(s)
Enterotoxins/immunology , Immunosuppressive Agents/therapeutic use , Pneumonia, Staphylococcal/drug therapy , Staphylococcus aureus/drug effects , Superantigens/immunology , Systemic Inflammatory Response Syndrome/drug therapy , Tacrolimus/therapeutic use , Animals , Cytokines/biosynthesis , Cytokines/immunology , HLA-DR3 Antigen/genetics , Humans , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/physiopathology , Staphylococcus aureus/pathogenicity , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/physiopathology , T-Lymphocytes/immunology , Tacrolimus/pharmacology , Treatment Outcome
15.
PLoS One ; 6(11): e27203, 2011.
Article in English | MEDLINE | ID: mdl-22102880

ABSTRACT

Staphylococcal enterotoxin B (SEB) is one of a family of toxins secreted by Staphylococcus aureus that act as superantigens, activating a large fraction of the T-cell population and inducing production of high levels of inflammatory cytokines that can cause toxic shock syndrome (TSS) and death. Extracellular engagement of the TCR of T-cells and class II MHC of antigen presenting cells by SEB triggers the activation of many intracellular signaling processes. We engineered chimeric antibodies to block the extracellular engagement of cellular receptors by SEB and used a statin to inhibit intracellular signaling. Chimeric human-mouse antibodies directed against different neutralizing epitopes of SEB synergistically inhibited its activation of human T-cells in vitro. In the in vivo model of lethal toxic shock syndrome (TSS) in HLA-DR3 transgenic mice, two of these antibodies conferred significant partial protection when administered individually, but offered complete protection in a synergistic manner when given together. Similarly, in vivo, lovastatin alone conferred only partial protection from TSS similar to single anti-SEB antibodies. However, used in combination with one chimeric neutralizing anti-SEB antibody, lovastatin provided complete protection against lethal TSS in HLA-DR3 transgenic mice. These experiments demonstrate that in vivo protection against lethal doses of SEB can be achieved by a statin of proven clinical safety and chimeric human-mouse antibodies, agents now widely used and known to be of low immunogenicity in human hosts.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Anticholesteremic Agents/therapeutic use , Enterotoxins/therapeutic use , Lovastatin/therapeutic use , Shock, Septic/prevention & control , Animals , Antibodies, Bacterial/therapeutic use , Antibodies, Neutralizing/therapeutic use , Drug Synergism , Enterotoxins/genetics , Enterotoxins/immunology , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Shock, Septic/immunology , Shock, Septic/mortality , Superantigens/immunology , Survival Rate
16.
Am J Pathol ; 178(6): 2760-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641398

ABSTRACT

Among the exotoxins produced by Staphylococcus aureus and Streptococcus pyogenes, the superantigens (SAgs) are the most potent T-cell activators known to date. SAgs are implicated in several serious diseases including toxic shock syndrome (TSS), Kawasaki disease, and sepsis. However, the immunopathogenesis of TSS and other diseases involving SAgs are still not completely understood. The commonly used conventional laboratory mouse strains do not respond robustly to SAgs in vivo. Therefore, they must be artificially rendered susceptible to TSS by using sensitizing agents such as d-galactosamine (d-galN), which skews the disease exclusively to the liver and, hence, is not representative of the disease in humans. SAg-induced TSS was characterized using transgenic mice expressing HLA class II molecules that are extremely susceptible to TSS without d-galN. HLA-DR3 transgenic mice recapitulated TSS in humans with extensive multiple-organ inflammation affecting the lung, liver, kidneys, heart, and small intestines. Heavy infiltration with T lymphocytes (both CD4(+) and CD8+), neutrophils, and macrophages was noted. In particular, the pathologic changes in the small intestines were extensive and accompanied by significantly altered absorptive functions of the enterocytes. In contrast to massive liver failure alone in the d-galN sensitization model of TSS, findings of the present study suggest that gut dysfunction might be a key pathogenic event that leads to high morbidity and mortality in humans with TSS.


Subject(s)
HLA-DR3 Antigen/immunology , Liver/immunology , Liver/pathology , Shock, Septic/immunology , Shock, Septic/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Enterotoxins/immunology , Galactosamine/immunology , Gastrointestinal Tract/pathology , Humans , Imaging, Three-Dimensional , Immunization , Mice , Mice, Transgenic , Organ Specificity/immunology , Permeability , Shock, Septic/blood , Shock, Septic/etiology , Time Factors
17.
PLoS One ; 6(2): e16764, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21304813

ABSTRACT

Toxic shock syndrome (TSS) caused by the superantigen exotoxins of Staphylococcus aureus and Streptococcus pyogenes is characterized by robust T cell activation, profound elevation in systemic levels of multiple cytokines, including interferon-γ (IFN-γ), followed by multiple organ dysfunction and often death. As IFN-γ possesses pro- as well as anti-inflammatory properties, we delineated its role in the pathogenesis of TSS. Antibody-mediated in vivo neutralization of IFN-γ or targeted disruption of IFN-γ gene conferred significant protection from lethal TSS in HLA-DR3 transgenic mice. Following systemic high dose SEB challenge, whereas the HLA-DR3.IFN-γ(+/+) mice became sick and succumbed to TSS, HLA-DR3.IFN-γ(-/-) mice appeared healthy and were significantly protected from SEB-induced lethality. SEB-induced systemic cytokine storm was significantly blunted in HLA-DR3.IFN-γ(-/-) transgenic mice. Serum concentrations of several cytokines (IL-4, IL-10, IL-12p40 and IL-17) and chemokines (KC, rantes, eotaxin and MCP-1) were significantly lower in HLA-DR3.IFN-γ(-/-) transgenic mice. However, SEB-induced T cell expansion in the spleens was unaffected and expansion of SEB-reactive TCR Vß8(+) CD4(+) and CD8(+) T cells was even more pronounced in HLA-DR3.IFN-γ(-/-) transgenic mice when compared to HLA-DR3.IFN-γ(+/+) mice. A systematic histopathological examination of several vital organs revealed that both HLA-DR3.IFN-γ(+/+) and HLA-DR3.IFN-γ(-/-) transgenic mice displayed comparable severe inflammatory changes in lungs, and liver during TSS. Remarkably, whereas the small intestines from HLA-DR3.IFN-γ(+/+) transgenic mice displayed significant pathological changes during TSS, the architecture of small intestines in HLA-DR3.IFN-γ(-/-) transgenic mice was preserved. In concordance with these histopathological changes, the gut permeability to macromolecules was dramatically increased in HLA-DR3.IFN-γ(+/+) but not HLA-DR3.IFN-γ(-/-) mice during TSS. Overall, IFN-γ seemed to play a lethal role in the immunopathogenesis of TSS by inflicting fatal small bowel pathology. Our study thus identifies the important role for IFN-γ in TSS.


Subject(s)
Interferon-gamma/physiology , Intestinal Diseases/complications , Intestinal Diseases/genetics , Shock, Septic/etiology , Superantigens/toxicity , Animals , Antibodies, Neutralizing/pharmacology , Chemokines/metabolism , Cytokines/metabolism , Disease Progression , Enterotoxins/toxicity , HLA-DR3 Antigen/genetics , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Diseases/mortality , Intestinal Diseases/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/mortality
18.
Mol Ther ; 18(6): 1143-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20372109

ABSTRACT

Bacterial superantigen (BSAg)-induced toxic shock syndrome (TSS) and bacterial lipopolysaccharide (LPS)-induced shock are characterized by severe systemic inflammation. As nuclear factor kappaB (NF kappaB) plays an important role in inflammation and bortezomib, a proteasome inhibitor widely used in cancer chemotherapy, is a potent inhibitor of NF kappaB activation, we evaluated the therapeutic and prophylactic use of bortezomib in these conditions using murine models. Bortezomib prophylaxis significantly reduced serum levels of many cytokines and chemokines induced by BSAg. However, at 3 hours, serum level of TNF-a, an important cytokine implicated in TSS, was significantly reduced but not abolished. At 6 hours, there was no difference in the serum TNF-a levels between bortezomib treated and untreated mice challenged with staphylococcal enterotoxin B (SEB). Paradoxically, all mice treated with bortezomib either before or after BSAg challenge succumbed to TSS. Neither bortezomib nor BSAg was lethal if given alone. Serum biochemical parameters and histopathological findings suggested acute liver failure as the possible cause of mortality. Liver tissue from SEB-challenged mice treated with bortezomib showed a significant reduction in NF kappaB activation. Because NF kappaB-dependent antiapoptotic pathways protect hepatocytes from TNF-alpha-induced cell death, inhibition of NF kappaB brought forth by bortezomib in the face of elevated TNF-alpha levels caused by BSAg or LPS is detrimental.


Subject(s)
Boronic Acids/adverse effects , Cysteine Proteinase Inhibitors/adverse effects , Lipopolysaccharides/toxicity , Proteasome Inhibitors , Pyrazines/adverse effects , Superantigens/toxicity , Systemic Inflammatory Response Syndrome/chemically induced , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Apoptosis , Bortezomib , Cytokines/biosynthesis , HLA-DR3 Antigen/genetics , Liver/drug effects , Liver/physiopathology , Mice , Mice, Transgenic , NF-kappa B/metabolism
19.
Infect Immun ; 78(6): 2801-11, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308304

ABSTRACT

Staphylococcal enterotoxin B (SEB), a shock-inducing exotoxin synthesized by Staphylococcus aureus, is an important cause of food poisoning and is a class B bioterrorism agent. SEB mediates antigen-independent activation of a major subset of the T-cell population by cross-linking T-cell receptors (TCRs) with class II major histocompatibility complex (MHC-II) molecules of antigen-presenting cells, resulting in the induction of antigen independent proliferation and cytokine secretion by a significant fraction of the T-cell population. Neutralizing antibodies inhibit SEB-mediated T-cell activation by blocking the toxin's interaction with the TCR or MHC-II and provide protection against the debilitating effects of this superantigen. We derived and searched a set of monoclonal mouse anti-SEB antibodies to identify neutralizing anti-SEB antibodies that bind to different sites on the toxin. A pair of non-cross-reactive, neutralizing anti-SEB monoclonal antibodies (MAbs) was found, and a combination of these antibodies inhibited SEB-induced T-cell proliferation in a synergistic rather than merely additive manner. In order to engineer antibodies more suitable than mouse MAbs for use in humans, the genes encoding the VL and VH gene segments of a synergistically acting pair of mouse MAbs were grafted, respectively, onto genes encoding the constant regions of human Igkappa and human IgG1, transfected into mammalian cells, and used to generate chimeric versions of these antibodies that had affinity and neutralization profiles essentially identical to their mouse counterparts. When tested in cultures of human peripheral blood mononuclear cells or splenocytes derived from HLA-DR3 transgenic mice, the chimeric human-mouse antibodies synergistically neutralized SEB-induced T-cell activation and cytokine production.


Subject(s)
Antibodies, Bacterial/immunology , Enterotoxins/antagonists & inhibitors , Staphylococcus aureus/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antitoxins/genetics , Antitoxins/immunology , Blood/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/immunology
20.
Physiol Genomics ; 37(3): 279-93, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19336531

ABSTRACT

Toxic shock syndrome (TSS) is an acute, serious systemic illness caused by bacterial superantigens. Nonavailability of a suitable animal model until recently has hampered an in-depth understanding of the pathogenesis of TSS. In the current study, we characterized the early molecular events underlying TSS using our HLA-DR3 transgenic mouse model. Gene expression profiling using DNA microarrays identified a rapid and significant upregulation of several pro- as well as anti-inflammatory mediators, many of which have never been previously described in TSS. In vivo administration of staphylococcal enterotoxin B (SEB) led to an increase in the expression of Th0- (IL-2, 240-fold); Th1- (IFN-gamma, 360-fold; IL-12, 8-fold); Th2- (IL-4, 53-fold; IL-5, 4-fold) as well as Th17-type cytokines (IL-21, 19-fold; IL-17, 5-fold). The immunoregulatory cytokines (IL-6, 700-fold; IL-10, 18-fold); CC chemokines (such as CCL 2, 11, 3, 24, 17, 12, 7), CXC chemokines (such as CXCL 1, 2, 5, 11, 10, 19); and several proteases (matrix metalloproteinases 13, 8, 3, and 9) were also upregulated. Serum levels of several of these cytokines/chemokines were also significantly elevated. Pathway analyses revealed significant modulation in a variety of biochemical and cellular functions, providing molecular insights into the pathogenesis of TSS. Administration of bortezomib, a clinically approved proteasome inhibitor capable of blocking NF-kappaB pathway, was able to significantly modulate the expression of a variety of genes induced by SEB. Thus, our study showed that TSS is a complex process and emphasized the potential of use of bortezomib in the therapy of superantigen-induced TSS.


Subject(s)
Enterotoxins/immunology , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Superantigens/immunology , Animals , Boronic Acids/pharmacology , Bortezomib , Cytokines/blood , Cytokines/genetics , Enterotoxins/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DR alpha-Chains , Inflammation Mediators/metabolism , Mice , Mice, Transgenic , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Receptors, Chemokine/genetics , Receptors, Cytokine/genetics , Signal Transduction/genetics , Superantigens/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...