Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(47): 29811-29822, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33177235

RESUMEN

Inflammasomes have been implicated in the detection and clearance of a variety of bacterial pathogens, but little is known about whether this innate sensing mechanism has any regulatory effect on the expression of stimulatory ligands by the pathogen. During infection with Salmonella and many other pathogens, flagellin is a major activator of NLRC4 inflammasome-mediated macrophage pyroptosis and pathogen eradication. Salmonella switches to a flagellin-low phenotype as infection progresses to avoid this mechanism of clearance by the host. However, the host cues that Salmonella perceives to undergo this switch remain unclear. Here, we report an unexpected role of the NLRC4 inflammasome in promoting expression of its microbial ligand, flagellin, and identify a role for type 1 IFN signaling in switching of Salmonella to a flagellin-low phenotype. Early in infection, activation of NLRC4 by flagellin initiates pyroptosis and concomitant release of lysophospholipids which in turn enhance expression of flagellin by Salmonella thereby amplifying its ability to elicit cell death. TRIF-dependent production of type 1 IFN, however, later represses NLRC4 and the lysophospholipid biosynthetic enzyme iPLA2, causing a decline in intracellular lysophospholipids that results in down-regulation of flagellin expression by Salmonella These findings reveal a previously unrecognized immune-modulating regulatory cross-talk between endosomal TLR signaling and cytosolic NLR activation with significant implications for the establishment of infection with Salmonella.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Flagelina/metabolismo , Fosfolipasas A2 Grupo VI/metabolismo , Interferón Tipo I/metabolismo , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Flagelina/inmunología , Fosfolipasas A2 Grupo VI/antagonistas & inhibidores , Humanos , Inmunidad Innata , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Cetonas/administración & dosificación , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Lisofosfolípidos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Naftalenos/administración & dosificación , Cultivo Primario de Células , Piroptosis/inmunología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/aislamiento & purificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
2.
J Immunol ; 205(2): 438-446, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32540995

RESUMEN

Pathogenic Salmonella serovars produce clinical manifestations ranging from systemic infection typhoid to invasive nontyphoidal Salmonella disease in humans. These serovars share a high degree of homology at the genome and the proteome level. However, whether infection or immunization with one serovar provides protection against other serovars has not been well studied. We show in this study that immunization of mice with live typhoidal serovar, Salmonella Typhi, generates cross-reactive immune responses, which provide far greater resistance against challenge with nontyphoidal serovar Salmonella Enteritidis than with another nontyphoidal serovar, Salmonella Typhimurium. Splenic T cells from these immunized mice produced similar levels of IL-2 and IFN-γ upon ex vivo stimulation with Ags prepared from S Enteritidis and S Typhimurium. In contrast, Abs against S Typhi interacted with live intact S Enteritidis but did not bind intact S Typhimurium. These pathogen-reactive Abs were largely directed against oligosaccharide (O)-antigenic determinant of LPS that S Typhi shares with S Enteritidis. Abs against the O determinant, which S Typhi shares with S Typhimurium, were present in the sera of immunized mice but did not bind live intact Salmonella because of surface inaccessibility of this determinant. Similar accessibility-regulated interaction was seen with Abs generated against S Typhimurium and S Enteritidis. Our results suggest that the ability of protective Abs elicited with one Salmonella serovar to engage with and consequently provide protection against another Salmonella serovar is determined by the accessibility of shared O Ags. These findings have significant and broader implications for immunity and vaccine development against pathogenic Salmonellae.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Epítopos/inmunología , Antígenos O/inmunología , Vacunas contra la Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Reacciones Cruzadas , Resistencia a la Enfermedad , Epítopos/genética , Humanos , Inmunidad Heteróloga , Inmunidad Humoral , Interferón gamma , Interleucina-2/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Antígenos O/genética , Salmonella typhimurium/genética , Serogrupo
3.
J Leukoc Biol ; 109(4): 821-831, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32717772

RESUMEN

TLRs recognize conserved pathogen associated molecular patterns and generate innate immune responses. Several circulating and cell membrane associated proteins have been shown to collaborate with TLRs in sensing microbial ligands and promoting inflammatory responses. Here, we show that serum and serum-borne lipids including lysophosphatidylcholine (LPC) amplify inflammatory responses from intestinal epithelial cells and mononuclear phagocytes primed with microbial TLR ligands. Treatment with the inhibitors of G protein-coupled receptor (GPCR) signaling, suramin, or pertussis toxin (PT), the inhibitor of JNK-MAPK, or knockdown of LPC response-regulating GPCR, G2A, decreases the augmentation brought about by serum or LPC in TLR-induced inflammatory response. In vivo administration of PT or anti-G2A antibody reduces TLR2-activated cytokine secretion. The ability of host lipids to costimulate TLR-generated cellular responses represents a novel pathway for the amplification of innate immunity and inflammation.


Asunto(s)
Inflamación/sangre , Lípidos/sangre , Receptores Toll-Like/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lisofosfatidilcolinas/farmacología , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Bio Protoc ; 11(21): e4205, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34859120

RESUMEN

The ability to identify the role of a particular gene within a system is dependent on control of the expression of that gene. In this protocol, we describe a method for stable, conditional expression of Nod-Like receptors (NLRs) in THP-1 cells using a lentiviral expression system. This system combines all the necessary components for tetracycline-inducible gene expression in a single lentivector with constitutive co-expression of a selection marker, which is an efficient means for controlling gene expression using a single viral infection of cells. This is done in a third generation lentiviral expression platform that improves the safety of lentiviruses and allows for greater gene expression than previous lentiviral platforms. The lentiviral expression plasmid is first engineered to contain the gene of interest driven by a TRE (tetracycline response element) promoter in a simple gateway cloning step and is then co-transfected into HEK293T cells, along with packaging and envelope plasmids to generate the virus. The virus is used to infect a cell type of interest at a low MOI so that the majority of the transduced cells contain a single viral integration. Infected cells are grown under selection, and viral integration is validated by qPCR. Gene expression in stably transduced cells is induced with doxycycline and validated by qPCR, immunoblot, and flow cytometry. This flexible lentiviral expression platform may be used for stable and robust induction of a gene of interest in a range of cells for multiple applications. Graphic abstract: Schematic overview of lentiviral transduction of THP-1 cells.

5.
Chem Sci ; 11(11): 3054-3067, 2020 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-34122810

RESUMEN

Antibiotic resistant infections are projected to cause over 10 million deaths by 2050, yet the development of new antibiotics has slowed. This points to an urgent need for methodologies for the rapid development of antibiotics against emerging drug resistant pathogens. We report on a generalizable combined computational and synthetic approach, called antibody-recruiting protein-catalyzed capture agents (AR-PCCs), to address this challenge. We applied the combinatorial protein catalyzed capture agent (PCC) technology to identify macrocyclic peptide ligands against highly conserved surface protein epitopes of carbapenem-resistant Klebsiella pneumoniae, an opportunistic Gram-negative pathogen with drug resistant strains. Multi-omic data combined with bioinformatic analyses identified epitopes of the highly expressed MrkA surface protein of K. pneumoniae for targeting in PCC screens. The top-performing ligand exhibited high-affinity (EC50 ∼50 nM) to full-length MrkA, and selectively bound to MrkA-expressing K. pneumoniae, but not to other pathogenic bacterial species. AR-PCCs that bear a hapten moiety promoted antibody recruitment to K. pneumoniae, leading to enhanced phagocytosis and phagocytic killing by macrophages. The rapid development of this highly targeted antibiotic implies that the integrated computational and synthetic toolkit described here can be used for the accelerated production of antibiotics against drug resistant bacteria.

6.
Sci Signal ; 13(661)2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293463

RESUMEN

Small, genetically determined differences in transcription [expression quantitative trait loci (eQTLs)] are implicated in complex diseases through unknown molecular mechanisms. Here, we showed that a small, persistent increase in the abundance of the innate pathogen sensor NOD1 precipitated large changes in the transcriptional state of monocytes. A ~1.2- to 1.3-fold increase in NOD1 protein abundance resulting from loss of regulation by the microRNA cluster miR-15b/16 lowered the threshold for ligand-induced activation of the transcription factor NF-κB and the MAPK p38. An additional sustained increase in NOD1 abundance to 1.5-fold over basal amounts bypassed this low ligand concentration requirement, resulting in robust ligand-independent induction of proinflammatory genes and oncogenes. These findings reveal that tight regulation of NOD1 abundance prevents this sensor from exceeding a physiological switching checkpoint that promotes persistent inflammation and oncogene expression. Furthermore, our data provide insight into how a quantitatively small change in protein abundance can produce marked changes in cell state that can serve as the initiator of disease.


Asunto(s)
Regulación de la Expresión Génica , Monocitos/metabolismo , Proteína Adaptadora de Señalización NOD1/biosíntesis , Proteínas Oncogénicas/biosíntesis , Transducción de Señal , Transcripción Genética , Humanos , Inflamación/metabolismo , Células THP-1
7.
Innate Immun ; 22(3): 238-44, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26888964

RESUMEN

TLRs have a fundamental role in immunity. We have recently reported that stimulation of TLR2 and TLR5 in freshly isolated and activated human T cells with microbial ligands without concomitant activation through the TCR brings about secretion of neutrophil chemoattractant, CXCL8, and effector cytokine, IFN-γ, respectively. However, the mechanism of TLR signaling in T cells has not been worked out. Here, we show that the Src family kinases, p56(lck)(Lck) and p59(fyn)(Fyn), which are essential for activation of T cells through the TCR, are also critical for signal transduction through TLRs in human T cells. The secretion of CXCL8 following stimulation of the model human T cell line, Jurkat, with the TLR5 ligand, flagellin, was reduced in presence of the Src-kinase inhibitor, PP2 and specific inhibitors of Lck and Fyn. These inhibitors suppressed generation of activated JNK and p38, which were both required for TLR-induced CXCL8 production. The Lck-deficient derivative of Jurkat, JCam1.6, responded poorly to TLR2, TLR5 and TLR7 agonists, and did not generate active signaling intermediates. Lck and Fyn inhibitors also reduced TLR5-induced IFN-γ secretion from the activated T cell phenotype-representing T cell line, HuT78, without modulating JNK and p38 activation. These results reveal that TCR and TLRs share key proximal signaling regulators in T cells.


Asunto(s)
Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/fisiología , Receptor Toll-Like 5/metabolismo , Flagelina/inmunología , Humanos , Inmunidad Innata , Interferón gamma/metabolismo , Interleucina-8/metabolismo , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Mutación/genética , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/microbiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 7/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Innate Immun ; 21(7): 778-83, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26303218

RESUMEN

Vi capsular polysaccharide is currently in use as a vaccine against human typhoid caused by Salmonella Typhi. The vaccine efficacy correlates with IgG anti-Vi Abs. We have recently reported that Vi can generate inflammatory responses through activation of the TLR2/TLR1 complex. In the present study, we show that immunization with Vi produces IgM as well as IgG Abs in wild type mice. This ability is not compromised in mice deficient in T cells. However, immunization of mice lacking the TLR adaptor protein, MyD88, with Vi elicits only IgM Abs. These results suggest that MyD88-dependent pro-inflammatory ability of the Vi vaccine might be vital in generating IgG Abs with this T-independent Ag.


Asunto(s)
Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Fiebre Tifoidea/inmunología , Vacunas Tifoides-Paratifoides/administración & dosificación , Animales , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inmunización , Cambio de Clase de Inmunoglobulina/genética , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Polisacáridos Bacterianos/inmunología , Transducción de Señal
9.
J Leukoc Biol ; 93(4): 521-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23345392

RESUMEN

T cells produce a number of cytokines and chemokines upon stimulation with TLR agonists in the presence or absence of TCR signals. Here, we show that secretion of neutrophil chemoattractant CXCL8 from human T cell line Jurkat in response to stimulation with TLR agonists is reduced when cell stimulation is carried out in presence of serum. Serum does not, however, inhibit TCR-activated secretion of CXCL8 nor does it down-regulate TLR-costimulated IL-2 secretion from activated T cells. The molecule that can mimic the ability to bring about suppression in CXCL8 from TLR-activated T cells is serum-borne bioactive lipid, S1P. Serum and S1P-mediated inhibition require intracellular calcium. S1P also suppresses CXCL8 secretion from peripheral blood-derived human T cells activated ex vivo with various TLR ligands. Our findings reveal a previously unrecognized role for S1P in regulating TLR-induced CXCL8 secretion from human T cells.


Asunto(s)
Interleucina-8/metabolismo , Lisofosfolípidos/farmacología , Esfingosina/análogos & derivados , Linfocitos T/efectos de los fármacos , Receptores Toll-Like/genética , Calcio/metabolismo , Clorhidrato de Fingolimod , Flagelina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Células Jurkat , Lipopéptidos/farmacología , Activación de Linfocitos/efectos de los fármacos , Imitación Molecular , Oxadiazoles/farmacología , Glicoles de Propileno/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Suero/química , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Linfocitos T/citología , Linfocitos T/inmunología , Tiofenos/farmacología , Receptores Toll-Like/inmunología , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA