Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
J Biomed Sci ; 30(1): 49, 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37381064

RESUMEN

BACKGROUND: The inflammatory response is indispensable for protective immunity, yet microbial pathogens often trigger an excessive response, 'cytokine storm', harmful to the host. Full T-cell activation requires interaction of costimulatory receptors B7-1(CD80) and B7-2(CD86) expressed on antigen-presenting cells with CD28 expressed on the T cells. We created short peptide mimetics of the homodimer interfaces of the B7 and CD28 receptors and examined their ability to attenuate B7/CD28 coligand engagement and signaling through CD28 for inflammatory cytokine induction in human immune cells, and to protect from lethal toxic shock in vivo. METHODS: Short B7 and CD28 receptor dimer interface mimetic peptides were synthesized and tested for their ability to attenuate the inflammatory cytokine response of human peripheral blood mononuclear cells, as well as for their ability to attenuate B7/CD28 intercellular receptor engagement. Mice were used to test the ability of such peptides to protect from lethal superantigen toxin challenge when administered in molar doses far below the toxin dose. RESULTS: B7 and CD28 homodimer interfaces are remote from the coligand binding sites, yet our finding is that by binding back into the receptor dimer interfaces, short dimer interface mimetic peptides inhibit intercellular B7-2/CD28 as well as the tighter B7-1/CD28 engagement, attenuating thereby pro-inflammatory signaling. B7 mimetic peptides exhibit tight selectivity for the cognate receptor in inhibiting intercellular receptor engagement with CD28, yet each diminishes signaling through CD28. In a prominent example of inflammatory cytokine storm, by attenuating formation of the B7/CD28 costimulatory axis, B7-1 and CD28 dimer interface mimetic peptides protect mice from lethal toxic shock induced by a bacterial superantigen even when administered in doses far submolar to the superantigen. CONCLUSIONS: Our results reveal that the B7 and CD28 homodimer interfaces each control B7/CD28 costimulatory receptor engagement and highlight the protective potential against cytokine storm of attenuating, yet not ablating, pro-inflammatory signaling via these receptor domains.


Asunto(s)
Antígenos CD28 , Choque Séptico , Humanos , Animales , Ratones , Leucocitos Mononucleares , Moléculas de Adhesión Celular , Síndrome de Liberación de Citoquinas , Citocinas , Polímeros , Superantígenos
2.
PLoS Genet ; 12(2): e1005840, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26886256

RESUMEN

Cancers often display gene expression profiles resembling those of undifferentiated cells. The mechanisms controlling these expression programs have yet to be identified. Exploring transcriptional enhancers throughout hematopoietic cell development and derived cancers, we uncovered a novel class of regulatory epigenetic mutations. These epimutations are particularly enriched in a group of enhancers, designated ES-specific enhancers (ESSEs) of the hematopoietic cell lineage. We found that hematopoietic ESSEs are prone to DNA methylation changes, indicative of their chromatin activity states. Strikingly, ESSE methylation is associated with gene transcriptional activity in cancer. Methylated ESSEs are hypermethylated in cancer relative to normal somatic cells and co-localized with silenced genes, whereas unmethylated ESSEs tend to be hypomethylated in cancer and associated with reactivated genes. Constitutive or hematopoietic stem cell-specific enhancers do not show these trends, suggesting selective reactivation of ESSEs in cancer. Further analyses of a hypomethylated ESSE downstream to the VEGFA gene revealed a novel regulatory circuit affecting VEGFA transcript levels across cancers and patients. We suggest that the discovered enhancer sites provide a framework for reactivation of ES genes in cancer.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Células Madre Embrionarias Humanas/metabolismo , Neoplasias/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Metilación de ADN/genética , Hematopoyesis/genética , Humanos , Neoplasias/patología , Proteínas del Grupo Polycomb/metabolismo , Reproducibilidad de los Resultados , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(42): E6437-E6446, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27708164

RESUMEN

Full T-cell activation requires interaction between the costimulatory receptors B7-2 and CD28. By binding CD28, bacterial superantigens elicit harmful inflammatory cytokine overexpression through an unknown mechanism. We show that, by engaging not only CD28 but also its coligand B7-2 directly, superantigens potently enhance the avidity between B7-2 and CD28, inducing thereby T-cell hyperactivation. Using the same 12-aa ß-strand-hinge-α-helix domain, superantigens engage both B7-2 and CD28 at their homodimer interfaces, areas remote from where these coreceptors interact, implying that inflammatory signaling can be controlled through the receptor homodimer interfaces. Short B7-2 dimer interface mimetic peptides bind diverse superantigens, prevent superantigen binding to cell-surface B7-2 or CD28, attenuate inflammatory cytokine overexpression, and protect mice from lethal superantigen challenge. Thus, superantigens induce a cytokine storm not only by mediating the interaction between MHC-II molecule and T-cell receptor but also, critically, by promoting B7-2/CD28 coreceptor engagement, forcing the principal costimulatory axis to signal excessively. Our results reveal a role for B7-2 as obligatory receptor for superantigens. B7-2 homodimer interface mimotopes prevent superantigen lethality by blocking the superantigen-host costimulatory receptor interaction.


Asunto(s)
Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Superantígenos/inmunología , Secuencia de Aminoácidos , Animales , Antígeno B7-2/química , Antígeno B7-2/genética , Línea Celular Tumoral , Citocinas/genética , Enterotoxinas/química , Enterotoxinas/inmunología , Femenino , Humanos , Ratones , Modelos Moleculares , Imitación Molecular , Péptidos/química , Péptidos/inmunología , Péptidos/metabolismo , Unión Proteica/inmunología , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes de Fusión , Transducción de Señal , Superantígenos/química , Superantígenos/metabolismo
4.
PLoS Biol ; 9(9): e1001149, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21931534

RESUMEN

Bacterial superantigens, a diverse family of toxins, induce an inflammatory cytokine storm that can lead to lethal shock. CD28 is a homodimer expressed on T cells that functions as the principal costimulatory ligand in the immune response through an interaction with its B7 coligands, yet we show here that to elicit inflammatory cytokine gene expression and toxicity, superantigens must bind directly into the dimer interface of CD28. Preventing access of the superantigen to CD28 suffices to block its lethality. Mice were protected from lethal superantigen challenge by short peptide mimetics of the CD28 dimer interface and by peptides selected to compete with the superantigen for its binding site in CD28. Superantigens use a conserved ß-strand/hinge/α-helix domain of hitherto unknown function to engage CD28. Mutation of this superantigen domain abolished inflammatory cytokine gene induction and lethality. Structural analysis showed that when a superantigen binds to the T cell receptor on the T cell and major histocompatibility class II molecule on the antigen-presenting cell, CD28 can be accommodated readily as third superantigen receptor in the quaternary complex, with the CD28 dimer interface oriented towards the ß-strand/hinge/α-helix domain in the superantigen. Our findings identify the CD28 homodimer interface as a critical receptor target for superantigens. The novel role of CD28 as receptor for a class of microbial pathogens, the superantigen toxins, broadens the scope of pathogen recognition mechanisms.


Asunto(s)
Antígenos CD28/inmunología , Citocinas/genética , Choque Séptico/inmunología , Superantígenos/inmunología , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/inmunología , Antígenos CD28/genética , Línea Celular Tumoral , Citocinas/inmunología , Enterotoxinas/inmunología , Mapeo Epitopo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Inmunidad Celular , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Unión Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Choque Séptico/genética , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Superantígenos/administración & dosificación , Resonancia por Plasmón de Superficie
6.
Genome Biol ; 24(1): 264, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38012713

RESUMEN

BACKGROUND: Common diseases manifest differentially between patients, but the genetic origin of this variation remains unclear. To explore possible involvement of gene transcriptional-variation, we produce a DNA methylation-oriented, driver-gene-wide dataset of regulatory elements in human glioblastomas and study their effect on inter-patient gene expression variation. RESULTS: In 175 of 177 analyzed gene regulatory domains, transcriptional enhancers and silencers are intermixed. Under experimental conditions, DNA methylation induces enhancers to alter their enhancing effects or convert into silencers, while silencers are affected inversely. High-resolution mapping of the association between DNA methylation and gene expression in intact genomes reveals methylation-related regulatory units (average size = 915.1 base-pairs). Upon increased methylation of these units, their target-genes either increased or decreased in expression. Gene-enhancing and silencing units constitute cis-regulatory networks of genes. Mathematical modeling of the networks highlights indicative methylation sites, which signified the effect of key regulatory units, and add up to make the overall transcriptional effect of the network. Methylation variation in these sites effectively describe inter-patient expression variation and, compared with DNA sequence-alterations, appears as a major contributor of gene-expression variation among glioblastoma patients. CONCLUSIONS: We describe complex cis-regulatory networks, which determine gene expression by summing the effects of positive and negative transcriptional inputs. In these networks, DNA methylation induces both enhancing and silencing effects, depending on the context. The revealed mechanism sheds light on the regulatory role of DNA methylation, explains inter-individual gene-expression variation, and opens the way for monitoring the driving forces behind deferential courses of cancer and other diseases.


Asunto(s)
Metilación de ADN , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Regulación de la Expresión Génica , Mutación
7.
Front Immunol ; 12: 723689, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489975

RESUMEN

The inflammatory activity of staphylococcal enterotoxin B (SEB) relies on its capacity to trigger polyclonal T-cell activation by binding both T-cell receptor (TCR) and costimulatory receptor CD28 on T cells and MHC class II and B7 molecules on antigen presenting cells (APC). Previous studies highlighted that SEB may bind TCR and CD28 molecules independently of MHC class II, yet the relative contribution of these interactions to the pro-inflammatory function of SEB remained unclear. Here, we show that binding to MHC class II is dispensable for the inflammatory activity of SEB, whereas binding to TCR, CD28 and B7 molecules is pivotal, in both human primary T cells and Jurkat T cell lines. In particular, our finding is that binding of SEB to B7 molecules suffices to trigger both TCR- and CD28-mediated inflammatory signalling. We also provide evidence that, by strengthening the interaction between CD28 and B7, SEB favours the recruitment of the TCR into the immunological synapse, thus inducing lethal inflammatory signalling.


Asunto(s)
Antígenos CD28/inmunología , Enterotoxinas/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Células Presentadoras de Antígenos/inmunología , Comunicación Celular , Células Cultivadas , Humanos , Activación de Linfocitos , Transducción de Señal , Linfocitos T/inmunología
8.
Artículo en Inglés | MEDLINE | ID: mdl-28286804

RESUMEN

Formation of the costimulatory axis between the B7-2 and CD28 coreceptors is critical for T-cell activation. Superantigens, Gram-positive bacterial virulence factors, cause toxic shock and sepsis by hyperinducing inflammatory cytokines. We report a novel role for costimulatory receptors CD28 and B7-2 as obligatory receptors for superantigens, rendering them therapeutic targets. We show that by engaging not only CD28 but also its coligand B7-2 directly, superantigens potently enhance the interaction between B7-2 and CD28, inducing thereby T-cell hyperactivation. Using a conserved twelve amino-acid domain, superantigens engage both B7-2 and CD28 at their homodimer interfaces, sites far removed from where these receptors interact, implying that inflammatory signaling can be controlled through the receptor homodimer interfaces. Short B7-2 and CD28 dimer interface mimetic peptides bind diverse superantigens, prevent superantigen binding to cell-surface B7-2 or CD28, attenuate inflammatory cytokine overexpression, and protect mice from lethal superantigen challenge. Thus, superantigens induce a cytokine storm by mediating not only the interaction between MHC-II molecule and T-cell receptor but critically, by promoting B7-2/CD28 coreceptor engagement, forcing the principal costimulatory axis to signal excessively. Our findings highlight the B7/CD28 interaction as a bottleneck in signaling for expression of inflammatory cytokines. B7-2 and CD28 homodimer interface mimetic peptides prevent superantigen lethality by blocking the superantigen-host costimulatory receptor interaction.

9.
Immunol Lett ; 82(1-2): 75-8, 2002 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-12008037

RESUMEN

Superantigens, exemplified by staphylococcal enterotoxin B (SEB), are the strongest known inducers of a cellular immune response; they elicit the production of excessive amounts of Th1 cytokines, IL-2, IFN-gamma and TNF, leading to toxic shock. We show that increasing doses of SEB cause not only a greater induction but also a more rapid cessation of IL-2 gene expression. Remarkably, exposure of human PBMC to a second dose of SEB, even at concentrations 10- or 100-fold lower than the initial inducing dose and even within 2 h after the first exposure to SEB, resulted in an immediate and essentially complete shutoff of the induced IL-2 and IFN-gamma mRNA expression. The shutoff response was observed when primary induction of IL-2 and IFN-gamma gene expression was by SEB but not when it was by phytohemaggutinin-P. Signaling by a superantigen thus results not only in a vigorous induction of Th1 cytokine genes but concomitantly induces the ability to shut off their expression upon re-exposure to superantigen. Without induction of this negative control mechanism, the cellular immune response to a superantigen would be even more pronounced.


Asunto(s)
Enterotoxinas/inmunología , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Superantígenos/inmunología , Células TH1/inmunología , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Silenciador del Gen , Humanos , Interferón gamma/genética , Interleucina-2/genética , Cinética , ARN Mensajero/biosíntesis , Activación Transcripcional
10.
Immunol Lett ; 91(2-3): 141-5, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15019282

RESUMEN

Bypassing the restricted presentation of conventional antigens, superantigens trigger an excessive cellular immune response leading to toxic shock. Antagonist peptides that inhibit the induction of human Th1 cytokine gene expression by a variety of bacterial superantigens protect mice from lethal toxic shock. We show that the surviving mice rapidly develop a broad-spectrum, protective immunity against further lethal toxin challenges with the same superantigen and even with superantigen toxins that they have not encountered before. By blocking the induction of a cellular immune response leading to toxic shock, the antagonist peptide allows the superantigen to induce a vigorous humoral immune response directed against itself, resulting in anti-toxin IgM and IgG that are broadly protective. Adoptive transfer of these antibodies to naïve mice rendered them resistant to lethal superantigen challenge. The appearance of these antibodies does not require immunization with an adjuvant and is not elicited by the antagonist peptide. Our results show that superantigens are potent immunogens when given the opportunity to induce a B cell response, in conditions where a deleterious Th1 response is prevented by the superantigen antagonist peptide.


Asunto(s)
Proteínas Bacterianas/inmunología , Enterotoxinas/inmunología , Exotoxinas/inmunología , Proteínas de la Membrana/inmunología , Péptidos/farmacología , Choque Séptico/inmunología , Choque Séptico/prevención & control , Superantígenos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Femenino , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos BALB C , Tasa de Supervivencia , Células TH1/efectos de los fármacos , Células TH1/inmunología
11.
Isr Med Assoc J ; 4(7): 520-3, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12120463

RESUMEN

BACKGROUND: Superantigens produced by Staphylococcus aureus and Streptococcus pyogenes are among the most lethal of toxins. Toxins in this family trigger an excessive cellular immune response leading to toxic shock. OBJECTIVES: To design an antagonist that is effective in vivo against a broad spectrum of superantigen toxins. METHODS: Short peptide antagonists were selected for their ability to inhibit superantigen-induced expression of human genes for cytokines that mediate shock. The ability of these peptides to protect mice against lethal toxin challenge was examined. RESULTS: Antagonist peptide protected mice against lethal challenge with staphylococcal enterotoxin B and toxic shock syndrome toxin-1, superantigens that share only 6% overall amino acid homology. Moreover, it rescued mice undergoing toxic shock. Antagonist peptides show homology to a beta-strand/hinge/alpha-helix domain that is structurally conserved among superantigens, yet remote from known binding sites for the major histocompatibility class II molecule and T cell receptor that function in toxic T cell hyperactivation. CONCLUSIONS: The lethal effect of superantigens can be blocked with a peptide antagonist that inhibits their action at the top of the toxicity cascade before activation of T cells occurs. Superantigenic toxin antagonists may serve not only as countermeasures to biologic warfare but may be useful in the treatment of staphylococcal and streptococcal toxic shock, as well as in some cases of septic shock.


Asunto(s)
Toxinas Bacterianas , Guerra Biológica , Choque Séptico/etiología , Staphylococcus aureus/inmunología , Streptococcus pyogenes/inmunología , Superantígenos/toxicidad , Animales , Enterotoxinas/toxicidad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C
12.
Toxins (Basel) ; 5(9): 1531-42, 2013 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-24022021

RESUMEN

Every adaptive immune response requires costimulation through the B7/CD28 axis, with CD28 on T-cells functioning as principal costimulatory receptor. Staphylococcal and streptococcal superantigen toxins hyperstimulate the T-cell-mediated immune response by orders of magnitude, inducing a lethal cytokine storm. We show that to elicit an inflammatory cytokine storm and lethality, superantigens must bind directly to CD28. Blocking access of the superantigen to its CD28 receptor with peptides mimicking the contact domains in either toxin or CD28 suffices to protect mice effectively from lethal shock. Our finding that CD28 is a direct receptor of superantigen toxins broadens the scope of microbial pathogen recognition mechanisms.


Asunto(s)
Antígenos CD28/inmunología , Superantígenos/inmunología , Toxinas Biológicas/inmunología , Animales , Sitios de Unión , Antígenos CD28/química , Humanos , Streptococcus pyogenes/inmunología , Superantígenos/química , Toxinas Biológicas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA