Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-37511553

RESUMO

As a biological macromolecule, the superantigen staphylococcal enterotoxin C2 (SEC2) is one of the most potent known T-cell activators, and it induces massive cytotoxic granule production. With this property, SEC2 and its mutants are widely regarded as immunomodulating agents for cancer therapy. In a previous study, we constructed an MHC-II-independent mutant of SEC2, named ST-4, which exhibits enhanced immunocyte stimulation and antitumor activity. However, tumor cells have different degrees of sensitivity to SEC2/ST-4. The mechanisms of immune resistance to SEs in cancer cells have not been investigated. Herein, we show that ST-4 could activate more powerful human lymphocyte granule-based cytotoxicity than SEC2. The results of RNA-seq and atomic force microscopy (AFM) analysis showed that, compared with SKOV3 cells, the softer ES-2 cells could escape from SEC2/ST-4-induced cytotoxic T-cell-mediated apoptosis by regulating cell softness through the CDC42/MLC2 pathway. Conversely, after enhancing the stiffness of cancer cells by a nonmuscle myosin-II-specific inhibitor, SEC2/ST-4 exhibited a significant antitumor effect against ES-2 cells by promoting perforin-dependent apoptosis and the S-phase arrest. Taken together, these data suggest that cell stiffness could be a key factor of resistance to SEs in ovarian cancer, and our findings may provide new insight for SE-based tumor immunotherapy.


Assuntos
Antineoplásicos , Enterotoxinas , Humanos , Enterotoxinas/farmacologia , Enterotoxinas/metabolismo , Superantígenos/farmacologia , Antineoplásicos/farmacologia , Linfócitos T , Ativação Linfocitária
2.
BMC Biotechnol ; 22(1): 31, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307814

RESUMO

BACKGROUND: Staphylococcal superantigens are virulence factors that help the pathogen escape the immune system and develop an infection. Toxic shock syndrome toxin (TSST)-1 is one of the most studied superantigens whose role in toxic shock syndrome and some particular disorders have been demonstrated. Inhibiting TSST-1 production with antibiotics and targeting TSST-1 with monoclonal antibodies might be one of the best strategies to prevent TSST-1-induced cytokines storm followed by lethality. RESULTS: A novel single-chain variable fragment (scFv), MS473, against TSST-1 was identified by selecting an scFv phage library on the TSST-1 protein. The MS473 scFv showed high affinity and specificity for TSST-1. Moreover, MS473 could significantly prevent TSST-1-induced mitogenicity (the IC50 value: 1.5 µM) and cytokine production. CONCLUSION: Using traditional antibiotics with an anti-TSST-1 scFv as a safe and effective agent leads to deleting the infection source and preventing the detrimental effects of the toxin disseminated into the whole body.


Assuntos
Anticorpos de Cadeia Única , Humanos , Anticorpos de Cadeia Única/farmacologia , Anticorpos de Cadeia Única/metabolismo , Staphylococcus aureus , Superantígenos/metabolismo , Superantígenos/farmacologia , Enterotoxinas , Citocinas/metabolismo , Antibacterianos/farmacologia
3.
J Leukoc Biol ; 111(3): 597-609, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34114693

RESUMO

Staphylococcal enterotoxins (SE) pose a great threat to human health due to their ability to bypass antigen presentation and activate large amounts of conventional T cells resulting in a cytokine storm potentially leading to toxic shock syndrome. Unconventional T- and NK cells are also activated by SE but the mechanisms remain poorly understood. In this study, the authors aimed to explore the underlying mechanism behind SE-mediated activation of MAIT-, γδ T-, and NK cells in vitro. CBMC or PBMC were stimulated with the toxins SEA, SEH, and TSST-1, and cytokine and cytotoxic responses were analyzed with ELISA and flow cytometry. All toxins induced a broad range of cytokines, perforin and granzyme B, although SEH was not as potent as SEA and TSST-1. SE-induced IFN-γ expression in MAIT-, γδ T-, and NK cells was clearly reduced by neutralization of IL-12, while cytotoxic compounds were not affected at all. Kinetic assays showed that unconventional T cell and NK cell-responses are secondary to the response in conventional T cells. Furthermore, co-cultures of isolated cell populations revealed that the ability of SEA to activate γδ T- and NK cells was fully dependent on the presence of both monocytes and αß T cells. Lastly, it was found that SE provoked a reduced and delayed cytokine response in infants, particularly within the unconventional T and NK cell populations. This study provides novel insights regarding the activation of unconventional T- and NK cells by SE, which contribute to understanding the vulnerability of young children towards Staphylococcus aureus infections.


Assuntos
Monócitos , Linfócitos T , Criança , Pré-Escolar , Citocinas , Enterotoxinas/farmacologia , Humanos , Células Matadoras Naturais , Leucócitos Mononucleares , Staphylococcus aureus , Superantígenos/farmacologia
4.
mSphere ; 6(4): e0060821, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34319127

RESUMO

Atopic dermatitis (AD) is a condition affecting 30 million persons in the United States. AD patients are heavily infected with Staphylococcus aureus on the skin. A particularly severe form of AD is eczema herpeticum (ADEH), where the patients' AD is complicated by S. aureus and herpes simplex virus (HSV) infection. This study examined the S. aureus strains from 15 ADEH patients, provided blinded, and showed a high association of ADEH with strains that produce toxic shock syndrome toxin-1 (TSST-1; 73%) compared to 10% production by typical AD isolates from patients without EH and those from another unrelated condition, cystic fibrosis. The ADEH isolates produced the superantigens associated with TSS (TSST-1 and staphylococcal enterotoxins A, B, and C). This association may in part explain the potential severity of ADEH. We also examined the effect of TSST-1 and HSV-1 on human epithelial cells and keratinocytes. TSST-1 used CD40 as its receptor on epithelial cells, and HSV-1 either directly or indirectly interacted with CD40. The consequence of these interactions was chemokine production, which is capable of causing harmful inflammation, with epidermal/keratinocyte barrier disruption. Human epithelial cells treated first with TSST-1 and then HSV-1 resulted in enhanced chemokine production. Finally, we showed that TSST-1 modestly increased HSV-1 replication but did not increase viral plaque size. Our data suggest that ADEH is associated with production of the major TSS-associated superantigens, together with HSV reactivation. The superantigens plus HSV may damage the skin barrier by causing harmful inflammation, thereby leading to increased symptoms. IMPORTANCE Atopic dermatitis (eczema, AD) with concurrent herpes simplex virus infection (eczema herpeticum, ADEH) is a severe form of AD. We show that ADEH patients are colonized with Staphylococcus aureus that primarily produces the superantigen toxic shock syndrome toxin-1 (TSST-1); however, significantly but to a lesser extent the superantigens staphylococcal enterotoxins A, B, and C are also represented in ADEH. Our studies showed that TSST-1 uses the immune costimulatory molecule CD40 as its epithelial cell receptor. Herpes simplex virus (HSV) also interacted directly or indirectly with CD40 on epithelial cells. Treatment of epithelial cells with TSST-1 and then HSV-1 resulted in enhanced chemokine production. We propose that this combination of exposures (TSST-1 and then HSV) leads to opening of epithelial and skin barriers to facilitate potentially serious ADEH.


Assuntos
Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Enterotoxinas/genética , Enterotoxinas/metabolismo , Herpesvirus Humano 1/metabolismo , Erupção Variceliforme de Kaposi/microbiologia , Staphylococcus aureus/patogenicidade , Superantígenos/genética , Superantígenos/metabolismo , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/farmacologia , Antígenos CD40/imunologia , Quimiocinas/imunologia , Enterotoxinas/imunologia , Enterotoxinas/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Células HaCaT , Herpesvirus Humano 1/imunologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/imunologia , Queratinócitos/microbiologia , Queratinócitos/virologia , Staphylococcus aureus/metabolismo , Superantígenos/imunologia , Superantígenos/farmacologia
5.
mSphere ; 5(5)2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-33028686

RESUMO

Staphylococcus aureus and Streptococcus pyogenes are significant human pathogens, causing infections at multiple body sites, including across the skin. Both are organisms that cause human diseases and secrete superantigens, including toxic shock syndrome toxin-1 (TSST-1), staphylococcal enterotoxins (SEs), and streptococcal pyrogenic exotoxins (SPEs). On the skin, human keratinocytes represent the first cell type to encounter these superantigens. We employed transcriptome sequencing (RNA-seq) to evaluate the human primary keratinocyte response to both TSST-1 and staphylococcal enterotoxin B (SEB) in triplicate analyses. Both superantigens caused large numbers of genes to be up- and downregulated. The genes that exhibited 2-fold differential gene expression compared to vehicle-treated cells, whether up- or downregulated, totaled 5,773 for TSST-1 and 4,320 for SEB. Of these, 4,482 were significantly upregulated by exposure of keratinocytes to TSST-1, whereas 1,291 were downregulated. For SEB, expression levels of 3,785 genes were upregulated, whereas those of 535 were downregulated. There was the expected high overlap in both upregulation (3,412 genes) and downregulation (400 genes). Significantly upregulated genes included those associated with chemokine production, with the possibility of stimulation of inflammation. We also tested an immortalized human keratinocyte line, from a different donor, for chemokine response to four superantigens. TSST-1 and SEB caused production of interleukin-8 (IL-8), MIP-3α, and IL-33. SPEA and SPEC were evaluated for stimulation of expression of IL-8 as a representative chemokine; both stimulated production of IL-8.IMPORTANCEStaphylococcus aureus and Streptococcus pyogenes are common human pathogens, causing infections that include the skin. Both pathogens produce a family of secreted toxins called superantigens, which have been shown to be important in human diseases. The first cell types encountered by superantigens on skin are keratinocytes. Our studies demonstrated, that the human keratinocyte pathway, among other pathways, responds to superantigens with production of chemokines, setting off inflammation. This inflammatory response may be harmful, facilitating opening of the skin barrier.


Assuntos
Queratinócitos/efeitos dos fármacos , Queratinócitos/imunologia , Superantígenos/imunologia , Superantígenos/farmacologia , Linhagem Celular Transformada , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Enterotoxinas/farmacologia , Perfilação da Expressão Gênica , Humanos , Inflamação , RNA-Seq , Staphylococcus aureus/química , Staphylococcus aureus/imunologia , Streptococcus pyogenes/química , Streptococcus pyogenes/imunologia
6.
Biochem Biophys Res Commun ; 532(2): 200-204, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32859377

RESUMO

S. aureus is associated with atopic dermatitis (AD). Several staphylococcal products including cell wall components, protease, and exotoxins, are thought to be involved in allergic inflammation of AD via activating immune cells such as T cells and mast cells. None of the staphylococcal exotoxins has been reported to activate a primary IL-4 inducer, basophils, that are known to produce large amounts of IL-4 in response to allergens as well as IgE-independent stimuli such as mites and helminth proteases. In this study, we investigated the ability of staphylococcal superantigen-like (SSL) family to activate basophils. SSL12, reported its activity to activate mast cells, induced the production of IL-4 in bone marrow derived basophils. SSL12 also evoked the release of IL-4 in freshly isolated murine basophils in bone marrow cells, as the depletion of basophils by basophils-specific antibodies against high-affinity IgE receptor and CD49b diminished the responsiveness of bone marrow cells for SSL12. These results propose the novel immune regulatory activity of SSL12 by inducing IL-4 in basophils, that contributes to the development of allergic inflammation disorders and the immune evasion of the cocci.


Assuntos
Basófilos/metabolismo , Interleucina-4/metabolismo , Staphylococcus aureus/imunologia , Superantígenos/farmacologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/farmacologia , Basófilos/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Interações Hospedeiro-Patógeno , Camundongos Endogâmicos C57BL , Staphylococcus aureus/patogenicidade , Superantígenos/genética , Superantígenos/imunologia
7.
Biomed Pharmacother ; 115: 108905, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31060004

RESUMO

Superantigens (SAgs) are a class of antigens that cause non-specific activation of T-cells resulting in polyclonal T cell activation and massive cytokine release and causing symptoms similar to sepsis, e.g. hypotension and subsequent hyporeactivity. We investigated the direct effect of SAgs on vascular tone using two recombinant SAgs, SEA and SPEA. The roles of Nitric Oxide (NO) and potentially hyperpolarization, which is dependent on the K+ channel activation, were also explored. The data show that SEA and SPEA have direct vasodilatory effects that were in part NO-dependent, but completely dependent on activation of K+ channels. Our work also identified the functional regions of one of the superantigens, SPEA, that are involved in causing the vasodilation and possible hypotension. A series of 20 overlapping peptides, spanning the entire sequence of SPEA, were designed and synthesized. The vascular response of each peptide was measured, and the active peptides were identified. Our results implicate the regions, (61-100), (101-140) and (181-220) which cause the vasodilation and possible hypotension effects of SPEA. The data also shows that the peptide 181-220 exert the highest vasodilation effect. This work therefore, demonstrates the direct effect of SAgs on vascular tone and identify the active region causing this vasodilation. We propose that these three peptides could be effective novel antihypertensive drugs. We also overexpressed, in E.coli, four superantigens from codon optimized genes.


Assuntos
Ativação Linfocitária/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Peptídeos/farmacologia , Superantígenos/farmacologia , Linfócitos T/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Animais , Escherichia coli/genética , Imunoterapia , Músculo Liso Vascular/efeitos dos fármacos , Neoplasias/imunologia , Peptídeos/genética , Ovinos , Superantígenos/genética , Linfócitos T/imunologia
8.
Sci Rep ; 9(1): 4211, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30862940

RESUMO

Methicillin resistant Staphylococcus aureus (MRSA) is a major human pathogen, which causes superficial to lethal clinical infections. Neutrophils are the most abundant leukocytes in the blood and are the first defense mechanism against S. aureus infections. Here we show Staphylococcal Superantigen-Like protein 11 (SSL11) from MRSA USA300_FPR3757 mediated differentiated human neutrophil-like cells (dHL60) motility arrest by inducing cell adhesion and "locking" cells in adhesion stage, without inducing oxidative burst. Pre-incubation of SSL11 with the glycan Sialyl Lewis X blocked SSL11 function and de-glycosylation of dHL60 cells by PNGase F abolished SSL11 binding, suggesting that SSL11 functions via interacting with glycans. This is the first description of a bacterial toxin inhibiting neutrophil motility by inducing adhesion and "locking" cells in an adhesion stage. Therefore, this study might provide a new target against S. aureus infections.


Assuntos
Proteínas de Bactérias/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/química , Neutrófilos/metabolismo , Superantígenos/farmacologia , Proteínas de Bactérias/química , Células HL-60 , Humanos , Neutrófilos/patologia , Superantígenos/química
9.
J Biol Chem ; 294(16): 6294-6305, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30782846

RESUMO

Several antibody-targeting cancer immunotherapies have been developed based on T cell activation at the target cells. One of the most potent activators of T cells are bacterial superantigens, which bind to major histocompatibility complex class II on antigen-presenting cells and activate T cells through T cell receptor. Strong T cell activation is also one of the main weaknesses of this strategy as it may lead to systemic T cell activation. To overcome the limitation of conventional antibody-superantigen fusion proteins, we have split a superantigen into two fragments, individually inactive, until both fragments came into close proximity and reassembled into a biologically active form capable of activating T cell response. A screening method based on fusion between SEA and coiled-coil heterodimers was developed that enabled detection of functional split SEA designs. The split SEA design that demonstrated efficacy in fusion with coiled-coil dimer forming polypeptides was fused to a single chain antibody specific for tumor antigen CD20. This design selectively activated T cells by split SEA-scFv fusion binding to target cells.


Assuntos
Enterotoxinas/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Cadeia Única/farmacocinética , Superantígenos/farmacologia , Linfócitos T/imunologia , Antígenos CD20/imunologia , Linhagem Celular Tumoral , Enterotoxinas/genética , Células HEK293 , Humanos , Anticorpos de Cadeia Única/genética , Superantígenos/genética , Linfócitos T/patologia
10.
Vet Res ; 49(1): 115, 2018 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-30486901

RESUMO

Staphylococcus aureus is a versatile opportunistic pathogen, causing disease in human and animal species. Its pathogenicity is linked to the ability of S. aureus to secrete immunomodulatory molecules. These evasion proteins bind to host receptors or their ligands, resulting in inhibitory effects through high affinity protein-protein interactions. Staphylococcal evasion molecules are often species-specific due to differences in host target proteins between species. We recently solved the crystal structure of murine TLR2 in complex with immunomodulatory molecule staphylococcal superantigen-like protein 3 (SSL3), which revealed the essential residues within SSL3 for TLR2 inhibition. In this study we aimed to investigate the molecular basis of the interaction on the TLR2 side. The SSL3 binding region on murine TLR2 was compared to that of other species through sequence alignment and homology modeling, which identified interspecies differences. To examine whether this resulted in altered SSL3 activity on the corresponding TLR2s, bovine, equine, human, and murine TLR2 were stably expressed in HEK293T cells and the ability of SSL3 to inhibit TLR2 was assessed. We found that SSL3 was unable to inhibit bovine TLR2. Subsequent loss and gain of function mutagenesis showed that the lack of inhibition is explained by the absence of two tyrosine residues in bovine TLR2 that play a prominent role in the SSL3-TLR2 interface. We found no evidence for the existence of allelic SSL3 variants that have adapted to the bovine host. Thus, within this paper we reveal the molecular determinants of the TLR2-SSL3 interaction which adds to our understanding of staphylococcal host specificity.


Assuntos
Proteínas de Bactérias/farmacologia , Superantígenos/farmacologia , Receptor 2 Toll-Like/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Bovinos , Clonagem Molecular , Simulação por Computador , Proteínas de Ligação a DNA , Células HEK293 , Cavalos , Humanos , Modelos Químicos , Modelos Moleculares , Conformação Proteica , Especificidade da Espécie , Staphylococcus aureus/fisiologia
11.
Toxins (Basel) ; 10(4)2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29584685

RESUMO

Staphylococcal Enterotoxins (SEs) are superantigens (SAg) originally produced by S. aureus, but their presence in coagulase negative staphylococci (CNS) has long been suspected. This study aims to better characterize a novel C-like enterotoxin expressed by clinical S. epidermidis strains, called SECepi. We isolated and characterized SECepi for its molecular and functional properties. The toxin was structurally modeled according to its significant similarity with S. aureus SEC3. Most of SEC amino acid residues important for the formation of the trimolecular Major Histocompatibility Complex II MHCII-SEC-T Cell Receptor TCR complex are conserved in SECepi. The functional properties of SECepi were estimated after cloning, expression in E. coli, and purification. The recombinant SECepi toxin exhibits biological characteristics of a SAg including stimulation of human T-cell mitogenicity, inducing and releasing high cytokines levels: IL-2, -4, -6, -8, -10, IFN-γ, TNF-α and GM-CSF at a dose as low as 3.7 pM. Compared to SECaureus, the production of pro-sepsis cytokine IL-6 is significantly higher with SECepi-activated lymphocytes. Furthermore, SECepi is stable to heat, pepsin or trypsin hydrolysis. The SECepi superantigen produced by CNS is functionally very close to that of S. aureus, possibly inducing a systemic inflammatory response at least comparable to that of SECaureus, and may account for S. epidermidis pathogenicity.


Assuntos
Enterotoxinas , Staphylococcus epidermidis/fisiologia , Superantígenos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Enterotoxinas/química , Enterotoxinas/metabolismo , Enterotoxinas/farmacologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Superantígenos/química , Superantígenos/metabolismo , Superantígenos/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/fisiologia
12.
Technol Cancer Res Treat ; 17: 1533033818762910, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29551087

RESUMO

Staphylococcal enterotoxin A is well known as a superantigen and able to be used for cancer immunotherapy. In this study, recombinant Staphylococcal enterotoxin A was genetically conjugated to epidermal growth factor to produce a chimeric protein recombinant Staphylococcal enterotoxin A-epidermal growth factor expressed in Escherichia coli. The recombinant Staphylococcal enterotoxin A-epidermal growth factor protein was purified using Strep-Tactin affinity chromatography and Endotoxin Removal Resin and identified by sodium dodecyl sulfate-polyacrylamide gel electropheresis and liquid chromatography-tandem mass spectrometry analysis. Furthermore, in vitro experiments showed purified recombinant Staphylococcal enterotoxin A-epidermal growth factor could successfully bind to the human nasopharyngeal carcinoma cell line CNE2, significantly promote the proliferation of human peripheral blood mononuclear cells, and enhance the secretion of several cytokines that have broad antitumor activities, such as interferon-γ, tumor necrosis factor-α, and interleukin-2 . Importantly, recombinant Staphylococcal enterotoxin A-epidermal growth factor significantly inhibited proliferation of CNE2 cells and promoted apoptosis in CNE2 cells when cocultured with peripheral blood mononuclear cells. Finally, both the binding of recombinant Staphylococcal enterotoxin A-epidermal growth factor and the toxicity of recombinant Staphylococcal enterotoxin A-epidermal growth factor-activated peripheral blood mononuclear cells were demonstrated as specific and only effective on high epidermal growth factor receptor-expressing cell lines. In all, our work suggests that recombinant Staphylococcal enterotoxin A-epidermal growth factor serves as a promising novel immunotherapeutic agent. More in vivo and in vitro studies are needed to verify its antitumor potency, as well as investigate the underlying mechanisms in cancer immunotherapy.


Assuntos
Antineoplásicos/farmacologia , Enterotoxinas/imunologia , Fator de Crescimento Epidérmico/farmacologia , Imunoterapia/métodos , Carcinoma Nasofaríngeo , Antineoplásicos/síntese química , Antineoplásicos/imunologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Citocinas/imunologia , Enterotoxinas/farmacologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Superantígenos/farmacologia
13.
Exp Dermatol ; 27(6): 681-684, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29498775

RESUMO

In this study, we investigated the role of IL-26 in allergic contact dermatitis (ACD), highlighting its' contribute in the cytotoxic mechanism responsible for the tissue injury. IL-26 is a signature Th17 cytokine, and immune cells are its predominant sources. Recently, it has shown that Th17 cell-derived-IL-26 functions like an antimicrobial peptide. Here, we hypothesized that IL-26 could be involved in cytotoxicity mechanism that underlies ACD. Indeed, we have attributed a role to IL-26 in this context, through PBMC cytotoxicity assays vs HaCat. To demonstrate that IL-26 was effectively involved in this activity, we performed the assay using transfected ACD PBMCs by siRNA for IL-26. Indeed, we demonstrated that these cells were less able to kill keratinocytes compared with ACD PBMCs (P < .01). In conclusion, our findings support the idea that this emergent cytokine, IL-26, is implicated in the killing mechanisms of KC observed during ACD.


Assuntos
Dermatite Alérgica de Contato/imunologia , Interleucinas/sangue , Interleucinas/imunologia , Leucócitos Mononucleares/metabolismo , Toxinas Bacterianas/farmacologia , Linhagem Celular , Sobrevivência Celular , Testes Imunológicos de Citotoxicidade , Dermatite Alérgica de Contato/sangue , Dermatite Alérgica de Contato/genética , Dermatite Atópica/genética , Enterotoxinas/farmacologia , Expressão Gênica , Inativação Gênica , Humanos , Interleucinas/genética , Queratinócitos , Níquel/farmacologia , Psoríase/genética , Superantígenos/farmacologia , Transfecção
14.
PLoS Pathog ; 13(9): e1006461, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28880920

RESUMO

Bacterial superantigens (SAgs) cause Vß-dependent T-cell proliferation leading to immune dysregulation associated with the pathogenesis of life-threatening infections such as toxic shock syndrome, and necrotizing pneumonia. Previously, we demonstrated that staphylococcal enterotoxin-like toxin X (SElX) from Staphylococcus aureus is a classical superantigen that exhibits T-cell activation in a Vß-specific manner, and contributes to the pathogenesis of necrotizing pneumonia. Here, we discovered that SElX can also bind to neutrophils from human and other mammalian species and disrupt IgG-mediated phagocytosis. Site-directed mutagenesis of the conserved sialic acid-binding motif of SElX abolished neutrophil binding and phagocytic killing, and revealed multiple glycosylated neutrophil receptors for SElX binding. Furthermore, the neutrophil binding-deficient mutant of SElX retained its capacity for T-cell activation demonstrating that SElX exhibits mechanistically independent activities on distinct cell populations associated with acquired and innate immunity, respectively. Finally, we demonstrated that the neutrophil-binding activity rather than superantigenicity is responsible for the SElX-dependent virulence observed in a necrotizing pneumonia rabbit model of infection. Taken together, we report the first example of a SAg, that can manipulate both the innate and adaptive arms of the human immune system during S. aureus pathogenesis.


Assuntos
Enterotoxinas/metabolismo , Exfoliatinas/farmacologia , Neutrófilos/efeitos dos fármacos , Infecções Estafilocócicas , Superantígenos/farmacologia , Animais , Exfoliatinas/metabolismo , Humanos , Ativação Linfocitária/imunologia , Neutrófilos/imunologia , Coelhos , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Superantígenos/imunologia
15.
Biosci Biotechnol Biochem ; 81(9): 1741-1746, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28715277

RESUMO

Superantigens can induce cell-mediated cytotoxicity preferentially against MHC II-positive target cells with large amounts of inflammatory cytokines releasing. In this study, superantigen staphylococcal enterotoxin C (SEC) 1 was investigated to evaluate its potential in bladder cancer immunotherapy in vitro and in vivo. Our results revealed that SEC1 could stimulate the proliferation of human peripheral blood mononuclear cells (PBMCs) in a dose-dependent manner, accompanied with the release of interleukin-2, interferon-γ, and tumor necrosis factor-α, and increased the population of CD4+ T cells and CD8+ T cells. PBMCs stimulated by SEC1 could initiate significant cytotoxicity towards human bladder cancer cells in vitro. The results of in vivo antitumor experiment indicated that SEC1 could decrease the rate of tumor formation and prolong the survival time of tumor-bearing mice. Our study demonstrated that SEC1 inhibited the growth of bladder cancer. And it is also suggested that SEC1 may become a candidate for bladder cancer immunotherapy.


Assuntos
Enterotoxinas/farmacologia , Superantígenos/farmacologia , Neoplasias da Bexiga Urinária/patologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Interferon gama/metabolismo , Interleucina-2/metabolismo , Camundongos , Fator de Necrose Tumoral alfa/metabolismo
16.
Toxins (Basel) ; 8(11)2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27801832

RESUMO

Among the toxin family of bacterial superantigens, the six members of the enterotoxin gene cluster (egc) seem to have unusual characteristics. They are present in the majority of Staphylococcus aureus strains, but their role in disease remains uncertain. We assessed secretion levels, immunogenicity, and toxicity of native and recombinant egc proteins. After having developed enzyme-linked immunosorbent assays, we found different quantities of egc proteins secreted by bacterial isolates. Supernatants induced proliferation of human peripheral blood mononuclear cells. However, purified recombinant egc proteins were shown to have differing superantigenicity potentials. Immunization with identical amounts of all members of egc, and the prominent toxic agent SEB, resulted in neutralizing antisera. Two egc proteins, SEI and SElN, were found to play a predominant role within the cluster. Both displayed the highest potential to activate blood cells, and were essential to be neutralized in supernatants. The application of a supernatant of a strain bearing only egc was sufficient for a lethal outcome in a rabbit model. Again, neutralization of SEI and SElN led to the survival of all tested animals. Finally, nanogram amounts of purified rSEI and rSElN led to lethality in vivo, pointing out the importance of both as virulence determinants among egc superantigens.


Assuntos
Proteínas de Bactérias , Toxinas Bacterianas , Enterotoxinas , Staphylococcus aureus , Superantígenos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Enterotoxinas/genética , Enterotoxinas/metabolismo , Enterotoxinas/farmacologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Coelhos , Proteínas Recombinantes/farmacologia , Staphylococcus aureus/genética , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/metabolismo , Superantígenos/genética , Superantígenos/metabolismo , Superantígenos/farmacologia
17.
Ann Surg Oncol ; 23(2): 655-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26289805

RESUMO

BACKGROUND: Complement is a central part of both the innate and adaptive immune response and its activation has traditionally been considered part of the immunosurveillance response against cancer. Its pro-inflammatory role and its contribution to the development of many illnesses associated with inflammatory states implicate complement in carcinogenesis. METHODS: We evaluated the role of three protein inhibitors of complement-cobra venom factor, humanized cobra venom factor, and recombinant staphylococcus aureus superantigen-like protein 7-in the setting of a transplantable murine colon cancer model. Outcomes were evaluated by monitoring tumor growth, and flow cytometry, ELISPOT, and quantitative real-time PCR were used to determine the impact of complement inhibition on the host immune response. RESULTS: Complement inhibitors were effective at depleting complement component C3 in tumor bearing mice and this was temporally correlated with a decreased rate of tumor growth during the establishment of tumors. Treatment with cobra venom factor resulted in increased CD8(+) T cells as a percentage of tumor-infiltrating cells as well as a reduced immunosuppressive environment evidenced by decreased myeloid derived suppressor cells in splenocytes of treated mice. Complement inhibition resulted in increased expression of the chemoattractive cytokines CCL5, CXCL10, and CXCL11. DISCUSSION: Complement depletion represents a promising mode of immunotherapy in cancer by its ability to impair tumor growth by increasing the host's effective immune response to tumor and diminishing the immunosuppressive effect created by the tumor microenvironment and ultimately could be utilized as a component of combination immunotherapy.


Assuntos
Neoplasias do Colo/terapia , Ativação do Complemento/efeitos dos fármacos , Complemento C3/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Imunoterapia , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Venenos Elapídicos/farmacologia , Ensaio de Imunoadsorção Enzimática , Exotoxinas/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superantígenos/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
18.
Technol Cancer Res Treat ; 15(2): 215-26, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25759426

RESUMO

Tumor-targeted superantigens (TTSs) have been used to treat a variety of tumors in preclinical studies. The TTS utilizes the powerful T-cell activation strategy by means of staphylococcal enterotoxins (SEs) as superantigens (Sags) to target tumor cells. Monoclonal antibodies and tumor-related ligands have been used as targeting molecules of Sag. In this study, we assessed the antitumor potency of tumor-targeted superantigen (TTS) strategy to design and produce fusion protein as a new antitumor candidate. The third loop (L3) of transforming growth factor α (TGF-α) was genetically conjugated to staphylococcal enterotoxin type B (TGFαL3-SEB), and its in vitro antitumor activity against murine breast cancer cells (A431 cell line) was evaluated. We designed and prepared TGFαL3-SEB chimeric protein and evaluated superantigenic activity, binding property to cancer cells, overexpression of epidermal growth factor receptor (EGFR), and in vitro antitumor activities. Cloning of tgfαl3-seb was confirmed by colony-polymerase chain reaction, enzymatic digestion, and sequencing. The recombinant TGFαL3-SEB fusion protein with molecular weight of 31 kDa was expressed and confirmed by anti-His Western-blot analysis. The TGFαL3-SEB fusion protein attached to A431 cell line with proper affinity and induced dose-dependent cytotoxicity against EGFR-expressing cancer cells in vitro. The TGFαL3-SEB chimeric protein exhibited potent in vitro antitumor activity. Our findings indicated that TGFαL3-SEB may be a promising anticancer candidate in cancer immunotherapy, and further studies are required to explore its potential in vivo therapeutic applications.


Assuntos
Antineoplásicos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Superantígenos/farmacologia , Animais , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Enterotoxinas/genética , Receptores ErbB/metabolismo , Imunoterapia , Concentração Inibidora 50 , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Staphylococcus aureus/genética , Superantígenos/biossíntese , Fator de Crescimento Transformador alfa/genética
19.
PLoS One ; 9(11): e113018, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25402468

RESUMO

Toxic shock syndrome toxin-1 (TSST-1), a superantigen produced from Staphylococcus aureus, has been reported to bind directly to unknown receptor(s) and penetrate into non-immune cells but its function is unclear. In this study, we demonstrated that recombinant TSST-1 suppresses autophagosomal accumulation in the autophagic-induced HeLa 229 cells. This suppression is shared by a superantigenic-deficient mutant of TSST-1 but not by staphylococcal enterotoxins, suggesting that autophagic suppression of TSST-1 is superantigenic-independent. Furthermore, we showed that TSST-1-producing S. aureus suppresses autophagy in the response of infected cells. Our data provides a novel function of TSST-1 in autophagic suppression which may contribute in staphylococcal persistence in host cells.


Assuntos
Autofagia/imunologia , Toxinas Bacterianas/farmacologia , Enterotoxinas/farmacologia , Células Epiteliais/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/efeitos dos fármacos , Inanição , Superantígenos/farmacologia , Autofagia/efeitos dos fármacos , Toxinas Bacterianas/genética , Western Blotting , Citocinas/metabolismo , Enterotoxinas/genética , Células Epiteliais/patologia , Células HeLa , Humanos , Mutação/genética , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Superantígenos/genética
20.
Metabolism ; 63(6): 831-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24684823

RESUMO

OBJECTIVE: The bacteria Staphylococcus aureus is part of the normal bacterial flora and produces a repertoire of enterotoxins which can cause food poisoning and toxic shock and might contribute to the pathogenesis of inflammatory diseases. These enterotoxins directly cross-link the T cell receptor with MHC class II, activating large amounts of T cells and are therefore called superantigens. It was recently discovered that the superantigen SEA binds to the cytokine receptor gp130. As obesity and type 2 diabetes are highly associated with inflammation of the adipose tissue and gp130 has been shown to play an important role in adipocytes, we wanted to investigate the effect of SEA on adipocyte signaling and function. MATERIALS/METHODS: Binding of SEA to gp130 was examined using surface plasmon resonance in a cell free system. Effects of SEA on adipocyte signaling, insulin sensitivity and function were studied using western blotting and biological assays for lipolysis, lipogenesis and glucose uptake. RESULTS: We demonstrate that SEA binds to gp130 with a medium affinity. Furthermore, SEA induces phosphorylation of a key downstream target, STAT3, in adipocytes. SEA also inhibits insulin-induced activation of PKB and PKB downstream signaling which was associated with reduced basal and insulin induced glucose uptake, reduced lipogenesis as well as reduced ability of insulin to inhibit lipolysis. CONCLUSIONS: SEA inhibits insulin signaling as well as insulin biological responses in adipocytes supporting that bacterial infection might contribute to the development of insulin resistance and type 2 diabetes.


Assuntos
Adipócitos/metabolismo , Infecções Bacterianas/metabolismo , Receptor gp130 de Citocina/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Insulina/metabolismo , Metabolismo dos Lipídeos , Superantígenos/farmacologia , Adipócitos/efeitos dos fármacos , Oxirredutases do Álcool/metabolismo , Animais , Infecções Bacterianas/complicações , Infecções Bacterianas/microbiologia , Bioensaio , Western Blotting , Receptor gp130 de Citocina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Eletroforese em Gel de Poliacrilamida , Enterotoxinas , Escherichia coli K12 , Glucose/metabolismo , Humanos , Resistência à Insulina , Lipogênese , Lipólise , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superantígenos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...