Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 399
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Genes Cells ; 29(5): 397-416, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38454012

RESUMO

Staphylococcus aureus is a noteworthy pathogen in allergic diseases, as four staphylococcal exotoxins activate mast cells, a significant contributor to inflammation, in an IgE-independent manner. Although the adhesion of mast cells is an essential process for their immune responses, only a small number of exotoxins have been reported to affect the process. Here, we demonstrated that staphylococcal superantigen-like (SSL) 3, previously identified as a toll-like receptor 2 agonist, induced the adhesion of murine bone marrow-derived mast cells to culture substratum. SSL3-induced adhesion was mediated by fibronectin in an Arg-Gly-Asp (RGD) sequence-dependent manner, suggesting the integrins were involved in the process. Additionally, SSL3 was found to bind to an anti-adhesive surface protein CD43. SSL3 induced the adhesion of HEK293 cells expressing exogenous CD43, suggesting that CD43 is the target molecule for adhesion induced by SSL3. Evaluation of SSL3-derived mutants showed that the C-terminal region (253-326), specifically T285 and H307, are necessary to induce adhesion. SSL3 augmented the IL-13 production of mast cells in response to immunocomplex and SSL12. These findings reveal a novel function of SSL3, triggering cell adhesion and enhancing mast cell activation. This study would clarify the correlation between S. aureus and allergic diseases such as atopic dermatitis.


Assuntos
Adesão Celular , Leucossialina , Mastócitos , Staphylococcus aureus , Superantígenos , Animais , Mastócitos/metabolismo , Mastócitos/imunologia , Camundongos , Humanos , Superantígenos/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/imunologia , Células HEK293 , Leucossialina/metabolismo , Proteínas de Bactérias/metabolismo , Interleucina-13/metabolismo , Camundongos Endogâmicos C57BL
2.
J Bacteriol ; 206(3): e0044723, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38334326

RESUMO

Menstrual toxic shock syndrome (mTSS) is a rare but life-threatening disease associated with the use of high-absorbency tampons. The production of the Staphylococcus aureus toxic shock syndrome toxin-1 (TSST-1) superantigen is involved in nearly all cases of mTSS and is tightly controlled by regulators responding to the environment. In the prototypic mTSS strain S. aureus MN8, the major repressor of TSST-1 is the carbon catabolite protein A (CcpA), which responds to glucose concentrations in the vaginal tract. Healthy vaginal Lactobacillus species also depend on glucose for both growth and acidification of the vaginal environment through lactic acid production. We hypothesized that interactions between the vaginal microbiota [herein referred to as community state types (CSTs)] and S. aureus MN8 depend on environmental cues and that these interactions subsequently affect TSST-1 production. Using S. aureus MN8 ΔccpA growing in various glucose concentrations, we demonstrate that the supernatants from different CSTs grown in vaginally defined medium (VDM) could significantly decrease tst expression. When co-culturing CST species with MN8 ∆ccpA, we show that Lactobacillus jensenii completely inhibits TSST-1 production in conditions mimicking healthy menstruation or mTSS. Finally, we show that growing S. aureus in "unhealthy" or "transitional" CST supernatants results in higher interleukin 2 (IL-2) production from T cells. These findings suggest that dysbiotic CSTs may encourage TSST-1 production in the vaginal tract and further indicate that the CSTs are likely important for the protection from mTSS.IMPORTANCEIn this study, we investigate the impact of the vaginal microbiota against Staphylococcus aureus in conditions mimicking the vaginal environment at various stages of the menstrual cycle. We demonstrate that Lactobacillus jensenii can inhibit toxic shock syndrome toxin-1 (TSST-1) production, suggesting the potential for probiotic activity in treating and preventing menstrual toxic shock syndrome (mTSS). On the other side of the spectrum, "unhealthy" or "transient" bacteria such as Gardnerella vaginalis and Lactobacillus iners support more TSST-1 production by S. aureus, suggesting that community state types are important in the development of mTSS. This study sets forward a model for examining contact-independent interactions between pathogenic bacteria and the vaginal microbiota. It also demonstrates the necessity of replicating the environment when studying one as dynamic as the vagina.


Assuntos
Toxinas Bacterianas , Lactobacillus , Choque Séptico , Infecções Estafilocócicas , Feminino , Humanos , Staphylococcus aureus/metabolismo , Choque Séptico/microbiologia , Sinais (Psicologia) , Enterotoxinas/metabolismo , Superantígenos/metabolismo , Vagina/microbiologia , Bactérias/metabolismo , Infecções Estafilocócicas/microbiologia , Glucose/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34548394

RESUMO

Microorganisms have coevolved diverse mechanisms to impair host defenses. A major one, superantigens, can result in devastating effects on the immune system. While all known superantigens induce vast immune cell proliferation and come from opportunistic pathogens, recently, proteins with similar broad specificity to antibody variable (V) domain families were identified in a commensal microbiota. These proteins, identified in the human commensal Ruminococcus gnavus, are called immunoglobulin-binding protein (Ibp) A and B and have been shown to activate B cells in vitro expressing either human VH3 or murine VH5/6/7. Here, we provide molecular and functional studies revealing the basis of this Ibp/immunoglobulin (Ig) interaction. The crystal structure and biochemical assays of a truncated IbpA construct in complex with mouse VH5 antigen-binding fragment (Fab) shows a binding of Ig heavy chain framework residues to the Ibp Domain D and the C-terminal heavy chain binding domain (HCBD). We used targeted mutagenesis of contact residues and affinity measurements and performed studies of the Fab-IbpA complex to determine the stoichiometry between Ibp and VH domains, suggesting Ibp may serve to cluster full-length IgA antibodies in vivo. Furthermore, in vitro stimulation experiments indicate that binding of the Ibp HCBD alone is sufficient to activate responsive murine B cell receptors. The presence of these proteins in a commensal microbe suggest that binding a broad repertoire of immunoglobulins, particularly in the gut/microbiome environment, may provide an important function in the maintenance of host/microbiome homeostasis contrasting with the pathogenic role of structurally homologous superantigens expressed by pathogens.


Assuntos
Anticorpos Monoclonais/metabolismo , Linfócitos B/metabolismo , Clostridiales/metabolismo , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Superantígenos/metabolismo , Animais , Anticorpos Monoclonais/química , Linfócitos B/imunologia , Sítios de Ligação , Clostridiales/crescimento & desenvolvimento , Humanos , Cadeias Pesadas de Imunoglobulinas/química , Região Variável de Imunoglobulina/química , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos B/química , Superantígenos/química
4.
Angew Chem Int Ed Engl ; 63(21): e202317552, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38497459

RESUMO

Celiac disease (CeD) is an autoimmune disorder triggered by gluten proteins, affecting approximately 1 % of the global population. The 33-mer deamidated gliadin peptide (DGP) is a metabolically modified wheat-gluten superantigen for CeD. Here, we demonstrate that the 33-mer DGP spontaneously assembles into oligomers with a diameter of approximately 24 nm. The 33-mer DGP oligomers present two main secondary structural motifs-a major polyproline II helix and a minor ß-sheet structure. Importantly, in the presence of 33-mer DGP oligomers, there is a statistically significant increase in the permeability in the gut epithelial cell model Caco-2, accompanied by the redistribution of zonula occludens-1, a master tight junction protein. These findings provide novel molecular and supramolecular insights into the impact of 33-mer DGP in CeD and highlight the relevance of gliadin peptide oligomerization.


Assuntos
Doença Celíaca , Enterócitos , Gliadina , Humanos , Doença Celíaca/metabolismo , Doença Celíaca/patologia , Células CACO-2 , Gliadina/química , Gliadina/metabolismo , Enterócitos/metabolismo , Superantígenos/química , Superantígenos/metabolismo , Permeabilidade
5.
Genes Cells ; 27(9): 559-567, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35801715

RESUMO

Staphylococcal superantigen-like 12 (SSL12) is reported to evoke the degranulation in murine mast cells. The allelic variant of SSL12 in the genome of reference strain NCTC8325 induced the degranulation of murine mast cells, that of MRSA252 strain did not, nevertheless relatively high sequence similarity (82%). To identify responsible amino acid residues of SSL12 for mast cell activation, we created a series of domain swap mutants and amino acid substitution mutants between the active and inactive variants. The mutants that harbored oligonucleotide/oligosaccharide binding (OB)-fold domain of the active variant activated mast cells. The replacement at position 56 (L56F) in the OB-fold domain diminished the mast cell stimulatory activity, and the combinatorial substitutions L56F/K92E, L56F/D95S, and L56F/S100V abolished the stimulatory activities of the mutant that harbored OB-fold domain of the active variant and the intact active variant. These indicate that the responsive elements of SSL12 for mast cell activation are in the OB-fold of SSL12, and L56 would be an essential amino acid residue for the activation of mast cells. The findings would contribute to the understanding of the molecular mechanism of SSL12 for mast cell activation and the development of toxoids preventing allergic inflammations associated with Staphylococcus aureus.


Assuntos
Infecções Estafilocócicas , Superantígenos , Aminoácidos/metabolismo , Animais , Mastócitos/metabolismo , Camundongos , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/química , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Superantígenos/genética , Superantígenos/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(41): 25254-25262, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32989130

RESUMO

Multisystem Inflammatory Syndrome in Children (MIS-C) associated with COVID-19 is a newly recognized condition in children with recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These children and adult patients with severe hyperinflammation present with a constellation of symptoms that strongly resemble toxic shock syndrome, an escalation of the cytotoxic adaptive immune response triggered upon the binding of pathogenic superantigens to T cell receptors (TCRs) and/or major histocompatibility complex class II (MHCII) molecules. Here, using structure-based computational models, we demonstrate that the SARS-CoV-2 spike (S) glycoprotein exhibits a high-affinity motif for binding TCRs, and may form a ternary complex with MHCII. The binding epitope on S harbors a sequence motif unique to SARS-CoV-2 (not present in other SARS-related coronaviruses), which is highly similar in both sequence and structure to the bacterial superantigen staphylococcal enterotoxin B. This interaction between the virus and human T cells could be strengthened by a rare mutation (D839Y/N/E) from a European strain of SARS-CoV-2. Furthermore, the interfacial region includes selected residues from an intercellular adhesion molecule (ICAM)-like motif shared between the SARS viruses from the 2003 and 2019 pandemics. A neurotoxin-like sequence motif on the receptor-binding domain also exhibits a high tendency to bind TCRs. Analysis of the TCR repertoire in adult COVID-19 patients demonstrates that those with severe hyperinflammatory disease exhibit TCR skewing consistent with superantigen activation. These data suggest that SARS-CoV-2 S may act as a superantigen to trigger the development of MIS-C as well as cytokine storm in adult COVID-19 patients, with important implications for the development of therapeutic approaches.


Assuntos
Betacoronavirus/imunologia , Infecções por Coronavirus/imunologia , Pneumonia Viral/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Superantígenos/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Motivos de Aminoácidos , Betacoronavirus/química , Betacoronavirus/genética , Betacoronavirus/metabolismo , COVID-19 , Infecções por Coronavirus/genética , Infecções por Coronavirus/patologia , Enterotoxinas/química , Epitopos de Linfócito T , Humanos , Molécula 1 de Adesão Intercelular/química , Modelos Moleculares , Mutação , Neurotoxinas/química , Pandemias , Pneumonia Viral/genética , Pneumonia Viral/patologia , Ligação Proteica , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Superantígenos/química , Superantígenos/genética , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/patologia
7.
J Bacteriol ; 204(10): e0026922, 2022 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-36106854

RESUMO

Staphylococcus aureus chronically colonizes up to 30% of the human population on the skin or mucous membranes, including the nasal tract or vaginal canal. While colonization is often benign, this bacterium also has the capability to cause serious infections. Menstrual toxic shock syndrome (mTSS) is a serious toxinosis associated with improper use of tampons, which can induce an environment that is favorable to the production of the superantigen known as toxic shock syndrome toxin-1 (TSST-1). To better understand environmental signaling that influences TSST-1 production, we analyzed expression in the prototype mTSS strain S. aureus MN8. Using transcriptional and protein-based analysis in two niche-related media, we observed that TSST-1 expression was significantly higher in synthetic nasal medium (SNM) than in vaginally defined medium (VDM). One major divergence in medium composition was high glucose concentration in VDM. The glucose-dependent virulence regulator gene ccpA was deleted in MN8, and, compared with wild-type MN8, we observed increased TSST-1 expression in the ΔccpA mutant when grown in VDM, suggesting that TSST-1 is repressed by catabolite control protein A (CcpA) in the vaginal environment. We were able to relieve CcpA-mediated repression by modifying the glucose level in vaginal conditions, confirming that changes in nutritional conditions contribute to the overexpression of TSST-1 that can lead to mTSS. We also compared CcpA-mediated repression to other key regulators of tst, finding that CcpA regulation is dominant compared to other characterized regulatory mechanisms. This study underlines the importance of environmental signaling for S. aureus pathogenesis in the context of mTSS. IMPORTANCE Menstrual toxic shock syndrome (mTSS) is caused by strains of Staphylococcus aureus that overproduce a toxin known as toxic shock syndrome toxin-1 (TSST-1). This work studied how glucose levels in a model vaginal environment could influence the amount of TSST-1 that is produced by S. aureus. We found that high levels of glucose repress TSST-1 production, and this is done by a regulatory protein called catabolite control protein A (CcpA). The research also demonstrated that, compared with other regulatory proteins, the CcpA regulator appears to be the most important for maintaining low levels of TSST-1 in the vaginal environment, and this information helps to understand how changes in the vaginal environmental can lead to mTSS.


Assuntos
Choque Séptico , Infecções Estafilocócicas , Feminino , Humanos , Staphylococcus aureus/metabolismo , Proteína Estafilocócica A/metabolismo , Choque Séptico/microbiologia , Glucose/metabolismo , Superantígenos/genética , Superantígenos/metabolismo , Enterotoxinas/genética , Enterotoxinas/metabolismo , Infecções Estafilocócicas/microbiologia , Meios de Cultura
8.
Biochemistry ; 61(7): 616-624, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35285627

RESUMO

Staphylococcus aureus is an opportunistic pathogen producing many immune evasion molecules targeting various components of the host immune defense, including the Staphylococcal superantigen-like protein (SSL 1-14) family. Despite sharing similar structures with the powerful superantigens (SAgs), which cause massive T cell activation, SSLs interfere with a wide range of innate immune defenses. SSLs are divided into two subgroups, SSLs that contain a conserved carbohydrate Sialyl Lewis X [Neu5Acα2-3Galß1-4(Fucα1-3) GlcNAcß, SLeX] binding site and SSLs that lack the SLeX binding site. SSL2-6 and SSL11 possess the SLeX binding site. Our previous studies showed that SSL11 arrests cell motility by inducing cell adhesion in differentiated HL60 (dHL60) cells, while SSL7 did not bind dHL60 cells. SSL7-based chimeras were engineered by exchanging the SSL7 sequence with the corresponding SSL11 sequence and assaying for a gain of SSL11 function, namely, the induction of cell spreading and motility arrest. In addition to the SLeX-binding site, we observed that three beta-strands ß6, ß7, and ß9 and the N-terminal residues, Y16 and Y17, transitioned SSL7 to gain SSL11 activities. These studies define the structure-function properties of SSL11 that may allow SSL11 to inhibit S. aureus clearance by the host innate immune system, allowing S. aureus to maintain a carrier state in humans, an understudied aspect of S. aureus pathogenesis.


Assuntos
Infecções Estafilocócicas , Superantígenos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Humanos , Neutrófilos , Ligação Proteica , Staphylococcus aureus/metabolismo , Superantígenos/química , Superantígenos/metabolismo
9.
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
10.
Protein Expr Purif ; 197: 106098, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35513232

RESUMO

Staphylococcus aureus is a common human and animal pathogen. These bacteria have various pathogenicity factors, including enterotoxin-like proteins. SElP (staphylococcal enterotoxin-like protein P) has potential zinc ion-binding sites and is able to interact with major histocompatibility complex class II (MHCII) and T-cell receptor (TCR). A method for the expression and isolation of the enterotoxin-like protein of Staphylococcus aureus (SElP) was developed. The expression was carried out in E. coli cells, and the protein was isolated by affinity chromatography on a NiNTA column. The endotoxins were separated by affinity chromatography on Affi-Prep® polymyxin. It was shown by gel filtration that the resulting protein had a monomeric form. The protein in zinc-bound and zinc-free forms was characterized by protein melting using fluorescence method and it was shown that zinc stabilizes the spatial structure of SElP. The functional activity of SElP was investigated by the ability to interact with the histocompatibility antigen class II receptor (MHC-II) exposed on the B cell line Raji by flow cytofluorometry. The zinc-bound and zinc-free forms were shown to differ in their interaction with MHC-II. The localization of the zinc-binding site was confirmed by the introduction of the H225 and D227 mutations. The mutant protein was characterized by melting, and its propensity to form aggregates was shown.


Assuntos
Enterotoxinas , Superantígenos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Enterotoxinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Antígenos de Histocompatibilidade Classe II/química , Íons , Receptores de Antígenos de Linfócitos T , Staphylococcus aureus/metabolismo , Superantígenos/genética , Superantígenos/metabolismo , Zinco/química
11.
Microbiol Immunol ; 66(8): 394-402, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35543108

RESUMO

T cell stimulation by bacterial superantigens induces a cytokine storm. After T cell activation and inflammatory cytokine secretion, regulatory T cells (Treg) are produced to suppress the immune response. Coccomyxa sp.KJ (IPOD FERM BP-22254), a green alga, is reported to regulate immune reactions. Therefore, we examined the effects of Coccomyxa sp.KJ extract (CE) on the superantigen-induced immune response. When human peripheral blood mononuclear cells (PBMCs) were stimulated with toxic shock syndrome-1 (TSST-1) in the presence of CE, the number of activated T cells decreased moderately. Purified T cells stimulated in the presence of CE comprised more non-proliferating cells than those stimulated in the absence of CE, whereas some T cells proliferated more quickly. The levels of activation markers on the stimulated T cells increased in the presence of CE. Most of the inflammatory cytokines did not change but IL-1ß, IL-17, IL-4, and IL-13 secretion increased, whereas that of IL-2, TNF-α, and IL-18 decreased. IL-10 secretion was also decreased by CE treatment, suggesting that the immune response was not suppressed by Treg cells. CE enhanced the expression of stem cell-like memory cell markers in T cells. These results suggest that CE can regulate the fate of T cells and can help to ameliorate superantigen-induced T cell hyperactivation and immune suppression.


Assuntos
Clorófitas , Infecções Estafilocócicas , Toxinas Bacterianas , Enterotoxinas , Humanos , Leucócitos Mononucleares , Ativação Linfocitária , Staphylococcus aureus , Superantígenos/metabolismo
12.
Platelets ; 33(7): 998-1008, 2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-35073811

RESUMO

Thrombocytopenia or platelet dysfunction is a risk factor for severe infection. Staphylococcus aureus (S. aureus) releases a variety of virulence factors especially toxic shock syndrome toxin 1 (TSST-1), which may cause toxic shock syndrome. S. aureus, when carrying the tst gene, is more prone to cause toxic shock syndrome and is responsible for an especially high rate of mortality. However, the effect of TSST-1 protein on platelets is unknown. Patients with the tst gene positive S. aureus bacteremia showed more serious infection, higher mortality and lower platelet count. The tst gene positive S. aureus strains induce more platelet apoptosis and activation and corresponding up-regulation of Bak and down-regulation of Bcl-XL in addition to the activation of Caspase-3. C57BL/6 mice infected with the tst gene positive strains resulted in both a decrease in platelet count and an increase in platelet apoptosis and/or activation events and mortality. Moreover, TSST-1 protein, encoded by tst gene, caused the decrease of platelet count, the increase of platelet apoptosis and activation events and the level of inflammatory cytokines in vivo. However, TSST-1 protein was unable to induce traditional activation and apoptosis on human platelets in vitro. These results suggested that TSST-1 protein may exert indirect effects on platelet activation and apoptosis in vivo.


Assuntos
Choque Séptico , Infecções Estafilocócicas , Animais , Apoptose , Toxinas Bacterianas , Enterotoxinas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus , Superantígenos/genética , Superantígenos/metabolismo , Superantígenos/toxicidade
13.
Cell Mol Life Sci ; 78(24): 8165-8186, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34731252

RESUMO

B cell superantigens crosslink conserved domains of B cell receptors (BCRs) and cause dysregulated, polyclonal B cell activation irrespective of normal BCR-antigen complementarity. The cells typically succumb to activation-induced cell death, which can impede the adaptive immune response and favor infection. In the present study, we demonstrate that the fucose-binding lectin of Burkholderia ambifaria, BambL, bears functional resemblance to B cell superantigens. By engaging surface glycans, the bacterial lectin activated human peripheral blood B cells, which manifested in the surface expression of CD69, CD54 and CD86 but became increasingly cytotoxic at higher concentrations. The effects were sensitive to BCR pathway inhibitors and excess fucose, which corroborates a glycan-driven mode of action. Interactome analyses in a model cell line suggest BambL binds directly to glycans of the BCR and regulatory coreceptors. In vitro, BambL triggered BCR signaling and induced CD19 internalization and degradation. Owing to the lectin's six binding sites, we propose a BCR activation model in which BambL functions as a clustering hub for receptor glycans, modulates normal BCR regulation, and induces cell death through exhaustive activation.


Assuntos
Linfócitos B/metabolismo , Proteínas de Bactérias/metabolismo , Burkholderia/metabolismo , Lectinas/metabolismo , Polissacarídeos/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Superantígenos/metabolismo , Linfócitos B/imunologia , Proteínas de Bactérias/imunologia , Sítios de Ligação , Humanos , Lectinas/imunologia , Polissacarídeos/imunologia , Ligação Proteica , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais , Superantígenos/imunologia
14.
Proc Natl Acad Sci U S A ; 116(51): 25923-25931, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31772015

RESUMO

Streptococcal toxic shock syndrome (STSS) is a rapidly progressing, life-threatening, systemic reaction to invasive infection caused by group A streptococci (GAS). GAS superantigens are key mediators of STSS through their potent activation of T cells leading to a cytokine storm and consequently vascular leakage, shock, and multiorgan failure. Mucosal-associated invariant T (MAIT) cells recognize MR1-presented antigens derived from microbial riboflavin biosynthesis and mount protective innate-like immune responses against the microbes producing such metabolites. GAS lack de novo riboflavin synthesis, and the role of MAIT cells in STSS has therefore so far been overlooked. Here we have conducted a comprehensive analysis of human MAIT cell responses to GAS, aiming to understand the contribution of MAIT cells to the pathogenesis of STSS. We show that MAIT cells are strongly activated and represent the major T cell source of IFNγ and TNF in the early stages of response to GAS. MAIT cell activation is biphasic with a rapid TCR Vß2-specific, TNF-dominated response to superantigens and a later IL-12- and IL-18-dependent, IFNγ-dominated response to both bacterial cells and secreted factors. Depletion of MAIT cells from PBMC resulted in decreased total production of IFNγ, IL-1ß, IL-2, and TNFß. Peripheral blood MAIT cells in patients with STSS expressed elevated levels of the activation markers CD69, CD25, CD38, and HLA-DR during the acute compared with the convalescent phase. Our data demonstrate that MAIT cells are major contributors to the early cytokine response to GAS, and are therefore likely to contribute to the pathological cytokine storm underlying STSS.


Assuntos
Citocinas/metabolismo , Células T Invariantes Associadas à Mucosa/imunologia , Choque Séptico/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Adulto , Idoso , Citocinas/sangue , Antígenos HLA-DR/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-2/metabolismo , Linfotoxina-alfa/metabolismo , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Riboflavina/biossíntese , Streptococcus pyogenes/patogenicidade , Superantígenos/metabolismo
15.
Proc Natl Acad Sci U S A ; 115(37): E8707-E8716, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30150373

RESUMO

Antibodies classically bind antigens via their complementarity-determining regions, but an alternative mode of interaction involving V-domain framework regions has been observed for some B cell "superantigens." We report the crystal structure of an antibody employing both modes of interaction simultaneously and binding two antigen molecules. This human antibody from an allergic individual binds to the grass pollen allergen Phl p 7. Not only are two allergen molecules bound to each antibody fragment (Fab) but also each allergen molecule is bound by two Fabs: One epitope is recognized classically, the other in a superantigen-like manner. A single allergen molecule thus cross-links two identical Fabs, contrary to the one-antibody-one-epitope dogma, which dictates that a dimeric allergen at least is required for this to occur. Allergens trigger immediate hypersensitivity reactions by cross-linking receptor-bound IgE molecules on effector cells. We found that monomeric Phl p 7 induced degranulation of basophils sensitized solely with this monoclonal antibody expressed as an IgE, demonstrating that the dual specificity has functional consequences. The monomeric state of Phl p 7 and two structurally related allergens was confirmed by size-exclusion chromatography and multiangle laser light scattering, and the results were supported by degranulation studies with the related allergens, a second patient-derived allergen-specific antibody lacking the nonclassical binding site, and mutagenesis of the nonclassically recognized allergen epitope. The antibody dual reactivity and cross-linking mechanism not only have implications for understanding allergenicity and allergen potency but, importantly, also have broader relevance to antigen recognition by membrane Ig and cross-linking of the B cell receptor.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Plantas/imunologia , Proteínas de Ligação ao Cálcio/imunologia , Epitopos/imunologia , Superantígenos/imunologia , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos/imunologia , Antígenos de Plantas/química , Antígenos de Plantas/metabolismo , Basófilos/imunologia , Basófilos/fisiologia , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/metabolismo , Degranulação Celular/imunologia , Reações Cruzadas/imunologia , Cristalografia por Raios X , Epitopos/química , Epitopos/metabolismo , Humanos , Imunoglobulina E/química , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Superantígenos/química , Superantígenos/metabolismo
16.
Mol Microbiol ; 112(4): 1163-1177, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31321813

RESUMO

Toxic shock syndrome toxin-1 (TSST-1) is a superantigen (SAg) produced by Staphylococcus aureus thought to be responsible for essentially all cases of menstrual-associated toxic shock syndrome (TSS). As a potent exotoxin, it is not surprising that S. aureus has evolved multiple systems to control expression of TSST-1. Although the accessory gene regulator (Agr) system is recognized to enhance TSST-1 expression, how Agr regulates TSST-1 is unclear. Using an agr-null mutant, complementation experiments demonstrated that Agr controls TSST-1 expression through the activity of the RNAIII effector molecule. RNAIII can repress translation of the repressor of toxins (Rot) regulator, and deletion of rot increased expression of TSST-1 during the exponential phase of growth. Deletion of agr did not affect rot transcription, but did result in overexpression of the Rot protein, and Rot was also shown to bind and positively regulate the rot promoter. Overexpression of Rot dramatically repressed TSST-1, and Rot bound directly to the TSST-1 promoter. Deletion of both agr and rot in S. aureus returned TSST-1 expression to wild-type levels. This work demonstrates that Agr, although widely considered to be an inducer of TSST-1, has evolved in combination with Rot, to restrict the expression of this potent SAg.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Enterotoxinas/genética , Choque Séptico/genética , Superantígenos/genética , Transativadores/metabolismo , Proteínas de Bactérias/genética , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/metabolismo , Enterotoxinas/imunologia , Enterotoxinas/metabolismo , Exotoxinas/imunologia , Regulação Bacteriana da Expressão Gênica/genética , Genes Reguladores/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Choque Séptico/metabolismo , Infecções Estafilocócicas/genética , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Superantígenos/imunologia , Superantígenos/metabolismo , Transativadores/genética
17.
PLoS Biol ; 15(7): e2003167, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28742082

RESUMO

The bacterium Staphylococcus aureus is an important cause of the life-threatening condition toxic shock syndrome in humans. Bacterial toxins known as superantigens (SAgs) generate this illness by acting as broad activators of a substantial fraction of all T lymphocytes, bypassing the normally highly stringent T-cell receptor antigen specificity to cause a systemic inflammatory cytokine storm in the host. In a new study, Shaler et al. found that immune cells called mucosa-associated invariant T (MAIT) cells make an unexpectedly large contribution to the SAg response in a largely T-cell receptor-independent, cytokine-driven manner. Subsequent to such activation, the MAIT cells remain unresponsive to stimulation with bacterial antigen. Thus, S. aureus hijacks MAIT cells in the cytokine storm and leaves them functionally impaired. This work provides new insight into the role of MAIT cells in antibacterial immunity and opens new avenues of investigation to understand and possibly treat bacterial toxic shock and sepsis.


Assuntos
Citocinas/metabolismo , Imunidade Celular , Modelos Imunológicos , Células T Invariantes Associadas à Mucosa/imunologia , Staphylococcus aureus/imunologia , Superantígenos/metabolismo , Animais , Anergia Clonal , Humanos , Ativação Linfocitária , Células T Invariantes Associadas à Mucosa/metabolismo , Células T Invariantes Associadas à Mucosa/microbiologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia
18.
PLoS Biol ; 15(6): e2001930, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28632753

RESUMO

Superantigens (SAgs) are potent exotoxins secreted by Staphylococcus aureus and Streptococcus pyogenes. They target a large fraction of T cell pools to set in motion a "cytokine storm" with severe and sometimes life-threatening consequences typically encountered in toxic shock syndrome (TSS). Given the rapidity with which TSS develops, designing timely and truly targeted therapies for this syndrome requires identification of key mediators of the cytokine storm's initial wave. Equally important, early host responses to SAgs can be accompanied or followed by a state of immunosuppression, which in turn jeopardizes the host's ability to combat and clear infections. Unlike in mouse models, the mechanisms underlying SAg-associated immunosuppression in humans are ill-defined. In this work, we have identified a population of innate-like T cells, called mucosa-associated invariant T (MAIT) cells, as the most powerful source of pro-inflammatory cytokines after exposure to SAgs. We have utilized primary human peripheral blood and hepatic mononuclear cells, mouse MAIT hybridoma lines, HLA-DR4-transgenic mice, MAIThighHLA-DR4+ bone marrow chimeras, and humanized NOD-scid IL-2Rγnull mice to demonstrate for the first time that: i) mouse and human MAIT cells are hyperresponsive to SAgs, typified by staphylococcal enterotoxin B (SEB); ii) the human MAIT cell response to SEB is rapid and far greater in magnitude than that launched by unfractionated conventional T, invariant natural killer T (iNKT) or γδ T cells, and is characterized by production of interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2, but not IL-17A; iii) high-affinity MHC class II interaction with SAgs, but not MHC-related protein 1 (MR1) participation, is required for MAIT cell activation; iv) MAIT cell responses to SEB can occur in a T cell receptor (TCR) Vß-specific manner but are largely contributed by IL-12 and IL-18; v) as MAIT cells are primed by SAgs, they also begin to develop a molecular signature consistent with exhaustion and failure to participate in antimicrobial defense. Accordingly, they upregulate lymphocyte-activation gene 3 (LAG-3), T cell immunoglobulin and mucin-3 (TIM-3), and/or programmed cell death-1 (PD-1), and acquire an anergic phenotype that interferes with their cognate function against Klebsiella pneumoniae and Escherichia coli; vi) MAIT cell hyperactivation and anergy co-utilize a signaling pathway that is governed by p38 and MEK1/2. Collectively, our findings demonstrate a pathogenic, rather than protective, role for MAIT cells during infection. Furthermore, we propose a novel mechanism of SAg-associated immunosuppression in humans. MAIT cells may therefore provide an attractive therapeutic target for the management of both early and late phases of severe SAg-mediated illnesses.


Assuntos
Antígenos de Bactérias/toxicidade , Anergia Clonal , Modelos Imunológicos , Células T Invariantes Associadas à Mucosa/imunologia , Staphylococcus aureus/imunologia , Streptococcus pyogenes/imunologia , Superantígenos/toxicidade , Animais , Antígenos de Bactérias/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Linhagem Celular , Células Cultivadas , Anergia Clonal/efeitos dos fármacos , Cruzamentos Genéticos , Enterotoxinas/metabolismo , Enterotoxinas/toxicidade , Feminino , Humanos , Hibridomas , Imunidade Inata , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Células T Invariantes Associadas à Mucosa/citologia , Células T Invariantes Associadas à Mucosa/efeitos dos fármacos , Células T Invariantes Associadas à Mucosa/metabolismo , Organismos Livres de Patógenos Específicos , Staphylococcus aureus/metabolismo , Streptococcus pyogenes/metabolismo , Superantígenos/metabolismo , Quimeras de Transplante/sangue , Quimeras de Transplante/imunologia , Quimeras de Transplante/metabolismo
19.
J Dairy Sci ; 103(5): 4100-4108, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32197850

RESUMO

Staphylococcus aureus is one of the main causative agents of food poisoning. This bacterium is an important component of cheese microbiota and plays an important role in foodborne diseases. Another important component of the microbiota is the lactic acid bacterium, which actively participates in processes that define the physicochemical, sensorial, and microbiological features of cheese. Of the various microbiological interactions in cheese, the interaction between lactic acid bacteria and Staph. aureus is most relevant. To this end, we evaluated the viability of Staph. aureus strains and the expression of their enterotoxins in cheeses produced experimentally, using Weissella paramesenteroides GIR16L4 or Lactobacillus rhamnosus D1 or both as starter cultures. Over 7 d, we observed that the presence of lactic acid bacteria did not impair Staph. aureus growth. However, via qPCR we observed a change in the gene expression of staphylococcal enterotoxins, suggesting that molecular communication exists between Staph. aureus strains and lactic acid bacteria in cheese.


Assuntos
Toxinas Bacterianas/metabolismo , Queijo/microbiologia , Enterotoxinas/metabolismo , Lacticaseibacillus rhamnosus/crescimento & desenvolvimento , Staphylococcus aureus/crescimento & desenvolvimento , Superantígenos/metabolismo , Weissella/crescimento & desenvolvimento , Animais , Toxinas Bacterianas/genética , Queijo/análise , Enterotoxinas/genética , Microbiologia de Alimentos , Lactobacillales/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Leite , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Superantígenos/genética , Transcriptoma , Weissella/metabolismo
20.
Int J Mol Sci ; 21(21)2020 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-33113941

RESUMO

The human genome comprises 8% sequences of retroviral origin, so-called human endogenous retroviruses (HERVs). Most of these proviral sequences are defective, but some possess open reading frames. They can lead to the formation of viral transcripts, when activated by intrinsic and extrinsic factors. HERVs are thought to play a pathological role in inflammatory diseases and cancer. Since the consequences of activated proviral sequences in the human body are largely unexplored, selected envelope proteins of human endogenous retroviruses associated with inflammatory diseases, namely HERV-K18, HERV-K113, and HERV-Fc1, were investigated in the present study. A formation of glycosylated envelope proteins was demonstrated in different mammalian cell lines. Nevertheless, protein maturation seemed to be incomplete as no transport to the plasma membrane was observed. Instead, the proteins remained in the ER where they induced the expression of genes involved in unfolded protein response, such as HSPA5 and sXBP1. Furthermore, low expression levels of native envelope proteins were increased by codon optimization. Cell-free expression systems showed that both the transcriptional and translational level is affected. By generating different codon-optimized variants of HERV-K113 envelope, the influence of single rare t-RNA pools in certain cell lines was demonstrated. The mRNA secondary structure also appears to play an important role in the translation of the tested viral envelope proteins. In summary, the formation of certain HERV proteins is basically possible. However, their complete maturation and thus full biologic activity seems to depend on additional factors that might be disease-specific and await elucidation in the future.


Assuntos
Retrovirus Endógenos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Superantígenos/genética , Superantígenos/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Células A549 , Animais , Células COS , Linhagem Celular , Sistema Livre de Células , Chlorocebus aethiops , Retrovirus Endógenos/química , Retrovirus Endógenos/genética , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica , Glicosilação , Células HEK293 , Humanos , Proteínas de Membrana/química , Conformação Molecular , Conformação de Ácido Nucleico , Fases de Leitura Aberta , Biossíntese de Proteínas , RNA Mensageiro/química , Superantígenos/química , Transcrição Gênica , Proteínas do Envelope Viral/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA