Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cytotherapy ; 25(8): 821-836, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37055321

RESUMEN

BACKGROUND AIMS: Extracellular vesicles (EVs) harvested from conditioned media of human mesenchymal stromal cells (MSCs) suppress acute inflammation in various disease models and promote regeneration of damaged tissues. After successful treatment of a patient with acute steroid-refractory graft-versus-host disease (GVHD) using EVs prepared from conditioned media of human bone marrow-derived MSCs, this study focused on improving the MSC-EV production for clinical application. METHODS: Independent MSC-EV preparations all produced according to a standardized procedure revealed broad immunomodulatory differences. Only a proportion of the MSC-EV products applied effectively modulated immune responses in a multi-donor mixed lymphocyte reaction (mdMLR) assay. To explore the relevance of such differences in vivo, at first a mouse GVHD model was optimized. RESULTS: The functional testing of selected MSC-EV preparations demonstrated that MSC-EV preparations revealing immunomodulatory capabilities in the mdMLR assay also effectively suppress GVHD symptoms in this model. In contrast, MSC-EV preparations, lacking such in vitro activities, also failed to modulate GVHD symptoms in vivo. Searching for differences of the active and inactive MSC-EV preparations, no concrete proteins or miRNAs were identified that could serve as surrogate markers. CONCLUSIONS: Standardized MSC-EV production strategies may not be sufficient to warrant manufacturing of MSC-EV products with reproducible qualities. Consequently, given this functional heterogeneity, every individual MSC-EV preparation considered for the clinical application should be evaluated for its therapeutic potency before administration to patients. Here, upon comparing immunomodulating capabilities of independent MSC-EV preparations in vivo and in vitro, we found that the mdMLR assay was qualified for such analyses.


Asunto(s)
Vesículas Extracelulares , Enfermedad Injerto contra Huésped , Células Madre Mesenquimatosas , MicroARNs , Humanos , Animales , Ratones , Medios de Cultivo Condicionados/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad Injerto contra Huésped/terapia , Células Madre Mesenquimatosas/metabolismo
2.
J Immunol ; 203(11): 2872-2886, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31636238

RESUMEN

TLR2 serves as a costimulatory molecule on activated T cells. However, it is unknown how the functionality and antiviral activity of CD8+ T cells are modulated by direct TLR2 signaling. In this study, we looked at the TLR2-mediated enhancement of TCR-driven CD8+ T cell activation in vitro and in woodchuck hepatitis virus transgenic mice. In vitro stimulation of CD8+ T cells purified from C57BL/6 mice showed that TLR2 agonist Pam3CSK4 directly enhanced the TCR-dependent CD8+ T cell activation. Transcriptome analysis revealed that TLR2 signaling increased expression of bioenergy metabolism-related genes in CD8+ T cells, such as IRF4, leading to improved glycolysis and glutaminolysis. This was associated with the upregulation of genes related to immune regulation and functions such as T-bet and IFN-γ. Glycolysis and glutaminolysis were in turn essential for the TLR2-mediated enhancement of T cell activation. Administration of TLR2 agonist Pam3CSK4 promoted the expansion and functionality of vaccine-primed, Ag-specific CD8+ T cells in both wild type and transgenic mice and improved viral suppression. Thus, TLR2 could promote CD8+ T cell immunity through regulating the energy metabolism.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Lipopéptidos/administración & dosificación , Lipopéptidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/inmunología
3.
EMBO Rep ; 18(12): 2144-2159, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29097394

RESUMEN

Immunity to mycobacteria involves the formation of granulomas, characterized by a unique macrophage (MΦ) species, so-called multinucleated giant cells (MGC). It remains unresolved whether MGC are beneficial to the host, that is, by prevention of bacterial spread, or whether they promote mycobacterial persistence. Here, we show that the prototypical antimycobacterial molecule nitric oxide (NO), which is produced by MGC in excessive amounts, is a double-edged sword. Next to its antibacterial capacity, NO propagates the transformation of MΦ into MGC, which are relatively permissive for mycobacterial persistence. The mechanism underlying MGC formation involves NO-induced DNA damage and impairment of p53 function. Moreover, MGC have an unsurpassed potential to engulf mycobacteria-infected apoptotic cells, which adds a further burden to their antimycobacterial capacity. Accordingly, mycobacteria take paradoxical advantage of antimicrobial cellular efforts by driving effector MΦ into a permissive MGC state.


Asunto(s)
Células Gigantes/microbiología , Macrófagos/fisiología , Mycobacterium/metabolismo , Óxido Nítrico/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Daño del ADN , Genes p53/fisiología , Células Gigantes/metabolismo , Humanos , Macrófagos/microbiología , Ratones , Mycobacterium/inmunología , Óxido Nítrico/biosíntesis
4.
J Immunol ; 196(6): 2733-41, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26873993

RESUMEN

Streptococci are common human colonizers with a species-specific mucocutaneous distribution. At the same time, they are among the most important and most virulent invasive bacterial pathogens. Thus, site-specific cellular innate immunity, which is predominantly executed by resident and invading myeloid cells, has to be adapted with respect to streptococcal sensing, handling, and response. In this article, we show that TLR13 is the critical mouse macrophage (MΦ) receptor in the response to group B Streptococcus, both in bone marrow-derived MΦs and in mature tissue MΦs, such as those residing in the lamina propria of the colon and the dermis, as well as in microglia. In contrast, TLR13 and its chaperone UNC-93B are dispensable for a potent cytokine response of blood monocytes to group B Streptococcus, although monocytes serve as the key progenitors of intestinal and dermal MΦs. Furthermore, a specific role for TLR13 with respect to MΦ function is supported by the response to staphylococci, where TLR13 and UNC-93B limit the cytokine response in bone marrow-derived MΦs and microglia, but not in dermal MΦs. In summary, TLR13 is a critical and site-specific receptor in the single MΦ response to ß-hemolytic streptococci.


Asunto(s)
Macrófagos/fisiología , Proteínas de Transporte de Membrana/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Receptores Toll-Like/metabolismo , Animales , Colon/patología , Citocinas/metabolismo , Hemólisis , Interacciones Huésped-Patógeno , Inmunidad Mucosa/genética , Inmunidad Mucosa/inmunología , Macrófagos/microbiología , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Especificidad de Órganos , Piel/patología , Receptores Toll-Like/genética
5.
J Allergy Clin Immunol ; 139(2): 667-678.e5, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27544739

RESUMEN

BACKGROUND: Bacterial cowshed isolates are allergy protective in mice; however, the underlying mechanisms are largely unknown. We examined the ability of Lactococcus lactis G121 to prevent allergic inflammatory reactions. OBJECTIVE: We sought to identify the ligands and pattern recognition receptors through which L lactis G121 confers allergy protection. METHODS: L lactis G121-induced cytokine release and surface expression of costimulatory molecules by untreated or inhibitor-treated (bafilomycin and cytochalasin D) human monocyte-derived dendritic cells (moDCs), bone marrow-derived mouse dendritic cells (BMDCs), and moDC/naive CD4+ T-cell cocultures were analyzed by using ELISA and flow cytometry. The pathology of ovalbumin-induced acute allergic airway inflammation after adoptive transfer of BMDCs was examined by means of microscopy. RESULTS: L lactis G121-treated murine BMDCs and human moDCs released TH1-polarizing cytokines and induced TH1 T cells. Inhibiting phagocytosis and endosomal acidification in BMDCs or moDCs impaired the release of TH1-polarizing cytokines, costimulatory molecule expression, and T-cell activation on L lactis G121 challenge. In vivo allergy protection mediated by L lactis G121 was dependent on endosomal acidification in dendritic cells (DCs). Toll-like receptor (Tlr) 13-/- BMDCs showed a weak response to L lactis G121 and were unresponsive to its RNA. The TH1-polarizing activity of L lactis G121-treated human DCs was blocked by TLR8-specific inhibitors, mediated by L lactis G121 RNA, and synergistically enhanced by activation of nucleotide-binding oligomerization domain-containing protein (NOD) 2. CONCLUSION: Bacterial RNA is the main driver of L lactis G121-mediated protection against experimentally induced allergy and requires both bacterial uptake by DCs and endosomal acidification. In mice L lactis G121 RNA signals through TLR13; however, the most likely intracellular receptor in human subjects is TLR8.


Asunto(s)
Antígenos Bacterianos/inmunología , Células Dendríticas/inmunología , Endosomas/metabolismo , Lactococcus lactis/inmunología , Pulmón/inmunología , Hipersensibilidad a la Leche/inmunología , ARN Bacteriano/inmunología , Animales , Bovinos , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Hipersensibilidad a la Leche/prevención & control , Proteína Adaptadora de Señalización NOD2/metabolismo , Células TH1/inmunología , Receptor Toll-Like 8/antagonistas & inhibidores , Receptores Toll-Like/genética
6.
EMBO Rep ; 16(12): 1656-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26545385

RESUMEN

Toll-like receptor (TLR) 13 and TLR2 are the major sensors of Gram-positive bacteria in mice. TLR13 recognizes Sa19, a specific 23S ribosomal (r) RNA-derived fragment and bacterial modification of Sa19 ablates binding to TLR13, and to antibiotics such as erythromycin. Similarly, RNase A-treated Staphylococcus aureus activate human peripheral blood mononuclear cells (PBMCs) only via TLR2, implying single-stranded (ss) RNA as major stimulant. Here, we identify human TLR8 as functional TLR13 equivalent that promiscuously senses ssRNA. Accordingly, Sa19 and mitochondrial (mt) 16S rRNA sequence-derived oligoribonucleotides (ORNs) stimulate PBMCs in a MyD88-dependent manner. These ORNs, as well as S. aureus-, Escherichia coli-, and mt-RNA, also activate differentiated human monocytoid THP-1 cells, provided they express TLR8. Moreover, Unc93b1(-/-)- and Tlr8(-/-)-THP-1 cells are refractory, while endogenous and ectopically expressed TLR8 confers responsiveness in a UR/URR RNA ligand consensus motif-dependent manner. If TLR8 function is inhibited by suppression of lysosomal function, antibiotic treatment efficiently blocks bacteria-driven inflammatory responses in infected human whole blood cultures. Sepsis therapy might thus benefit from interfering with TLR8 function.


Asunto(s)
Escherichia coli/genética , Escherichia coli/inmunología , ARN Bacteriano/química , ARN Bacteriano/inmunología , ARN/química , ARN/inmunología , Receptor Toll-Like 8/inmunología , Animales , Línea Celular Tumoral , Humanos , Leucocitos Mononucleares/inmunología , Ratones , Oligorribonucleótidos , ARN/genética , ARN Bacteriano/genética , ARN Mitocondrial , ARN Ribosómico 16S , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Receptor Toll-Like 8/química , Receptor Toll-Like 8/genética
7.
Cell Physiol Biochem ; 39(4): 1271-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606466

RESUMEN

BACKGROUND/AIMS: Viral infections represent a global health problem with the need for new viral therapies and better understanding of the immune response during infection. The most immediate and potent anti-viral defense mechanism is the production of type I interferon (IFN-I) which are activated rapidly following recognition of viral infection by host pathogen recognition receptors (PRR). The mechanisms of innate cellular signaling downstream of PRR activation remain to be fully understood. In the present study, we demonstrate that CASP2 and RIPK1 domain-containing adaptor with death domain (CRADD/RAIDD) is a critical component in type I IFN production. METHODS: The role of RAIDD during IFN-I production was investigated using western blot, shRNA mediated lentiviral knockdown, immunoprecipitation and IFN-I driven dual luciferase assay. RESULTS: Immunoprecipitation analysis revealed the molecular interaction of RAIDD with interferon regulatory factor 7 (IRF7) and its phosphorylating kinase IKKε. Using an IFN-4α driven dual luciferase analysis in RAIDD deficient cells, type I IFN activation by IKKε and IRF7 was dramatically reduced. Furthermore, deletion of either the caspase recruitment domain (CARD) or death domain (DD) of RAIDD inhibited IKKε and IRF7 mediated interferon-4α activation. CONCLUSION: We have identified that the adaptor molecule RAIDD coordinates IKKε and IRF7 interaction to ensure efficient expression of type I interferon.


Asunto(s)
Proteína Adaptadora de Señalización CRADD/genética , Quinasa I-kappa B/genética , Factor 7 Regulador del Interferón/genética , Receptor Toll-Like 3/genética , Animales , Proteína Adaptadora de Señalización CRADD/inmunología , Dominio de Reclutamiento y Activación de Caspasas , Regulación de la Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Quinasa I-kappa B/inmunología , Factor 7 Regulador del Interferón/inmunología , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Lentivirus/genética , Lentivirus/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Plásmidos/química , Plásmidos/metabolismo , Poli I-C/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal , Receptor Toll-Like 3/inmunología
8.
J Virol ; 88(18): 10840-50, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008920

RESUMEN

UNLABELLED: Modified vaccinia virus Ankara (MVA) serves as a versatile platform in vaccine development. This highly attenuated orthopoxvirus, which cannot replicate in mammalian cells, triggers strong innate immune responses, including cell migration. Previously, we have shown that induction of chemokine (C-C motif) ligand 2 (CCL2) by MVA is necessary for the recruitment of monocytes and T cells, but not neutrophils, to the lung. Here, we identified neutrophil-attracting chemokines produced by MVA-infected primary murine lung fibroblasts and murine bone marrow-derived macrophages. We demonstrate that MVA, but not vaccinia virus (VACV) strain WR, induces chemokine expression, which is independent of Toll-like receptor 2 (TLR2) signaling. Additionally, we show that both chemokine (C-C motif) receptor 1 (CCR1) and chemokine (C-X-C motif) receptor 2 (CXCR2) are involved in MVA-induced neutrophil chemotaxis in vitro. Finally, intranasal infection of Ccr1(-/-) mice with MVA, as well as application of the CCR1 antagonist J-113863, revealed a role for CCR1 in leukocyte recruitment, including neutrophils, into the lung. IMPORTANCE: Rapid attraction of leukocytes to the site of inoculation is unique to MVA in comparison to other VACV strains. The findings here extend current knowledge about the regulation of MVA-induced leukocyte migration, particularly regarding neutrophils, which could potentially be exploited to improve other VACV strains currently in development as oncolytic viruses and viral vectors. Additionally, the data presented here indicate that the inflammatory response may vary depending on the cell type infected by MVA, highlighting the importance of the site of vaccine application. Moreover, the rapid recruitment of neutrophils and other leukocytes can directly contribute to the induction of adaptive immune responses elicited by MVA inoculation. Thus, a better understanding of leukocyte migration upon MVA infection is particularly relevant for further development and use of MVA-based vaccines and vectors.


Asunto(s)
Neutrófilos/inmunología , Receptores CCR1/inmunología , Infecciones del Sistema Respiratorio/inmunología , Virus Vaccinia/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Pulmón/inmunología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR1/genética , Infecciones del Sistema Respiratorio/virología , Receptor Toll-Like 2/inmunología , Vaccinia , Virus Vaccinia/genética
10.
J Immunol ; 189(1): 328-36, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22634614

RESUMEN

Recognition of foreign nucleic acids is important for the induction of an innate immune response against invading pathogens. Although the pathways involved in sensing bacterial DNA and viral RNA are now well established, only limited knowledge is available on mechanisms underlying recognition of bacterial RNA. It has been reported that intracellular delivery of Escherichia coli RNA activates the Nlrp3 inflammasome, but whether this is a general property of bacterial RNA remains unclear as are the pathways involved in pro-IL-1ß induction and caspase-1 activation by bacterial RNA. In this study, we report that bacterial RNA from both Gram-positive and Gram-negative bacteria induces activation of caspase-1 and secretion of IL-1ß by murine dendritic cells and bone-marrow derived macrophages. Stimulation was independent of the presence of 5'-triphosphate termini and occurred with whole RNA preparations from bacteria but not from eukaryotes. Induction of pro-IL-1ß as well as the priming for caspase-1 activation by bacterial RNA was dependent on UNC93B, an endoplasmic reticulum protein essential for delivery of TLRs to the endosome, whereas the established nucleic acid sensing endosomal TLRs 3, 7, and 9 were dispensable. Additionally, caspase-1 activation and IL-1ß production by transfected bacterial RNA were absent in MyD88-deficient cells but independent of TRIF. Thus, our data indicate the presence of a yet unidentified intracellular nucleic acid receptor involved in bacterial RNA-induced inflammasome activation and release of IL-1ß.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Caspasa 1/metabolismo , Interleucina-1beta/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas de Transporte de Membrana/fisiología , ARN Bacteriano/fisiología , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 9/fisiología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Animales , Línea Celular , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Activación Enzimática/genética , Macrófagos/enzimología , Macrófagos/metabolismo , Macrófagos/microbiología , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 9/deficiencia
11.
Nanomedicine ; 10(8): 1787-98, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25014891

RESUMEN

Retroviral infections e.g. HIV still represent a unique burden in the field of vaccine research. A common challenge in vaccine design is to find formulations that create appropriate immune responses to protect against and/or control the given pathogen. Nanoparticles have been considered to be ideal vaccination vehicles that mimic invading pathogens. In this study, we present biodegradable calcium phosphate (CaP) nanoparticles, functionalized with CpG and retroviral T cell epitopes of Friend virus (FV) as excellent vaccine delivery system. CaP nanoparticles strongly increased antigen delivery to antigen-presenting cells to elicit a highly efficient T cell-mediated immune response against retroviral FV infection. Moreover, single-shot immunization of chronically FV-infected mice with functionalized CaP nanoparticles efficiently reactivated effector T cells which led to a significant decrease in viral loads. Thus, our findings clearly indicate that a nanoparticle-based peptide immunization is a promising approach to improve antiretroviral vaccination. FROM THE CLINICAL EDITOR: In this study, biodegradable calcium phosphate nanoparticles were used as a vaccine delivery system after functionalization with CpG and Friend virus-derived T-cell epitopes. This vaccination strategy resulted in increased T-cell mediated immune response even in chronically infected mice, providing a promising approach to the development of clinically useful antiretroviral vaccination strategies.


Asunto(s)
Inmunidad Celular/inmunología , Nanopartículas/química , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Citometría de Flujo , Ratones
12.
JCI Insight ; 9(6)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38358825

RESUMEN

Despite effective antibiotic therapy, brain-destructive inflammation often cannot be avoided in pneumococcal meningitis. The causative signals are mediated predominantly through TLR-recruited myeloid differentiation primary response adaptor 88 (MyD88), as indicated by a dramatic pneumococcal meningitis phenotype of Myd88-/- mice. Because lipoproteins and single-stranded RNA are crucial for recognition of Gram-positive bacteria such as Streptococcus pneumoniae by the host immune system, we comparatively analyzed the disease courses of Myd88-/- and Tlr2-/- Tlr13-/- mice. Their phenotypic resemblance indicated TLR2 and -13 as master sensors of S. pneumoniae in the cerebrospinal fluid. A neutralizing anti-TLR2 antibody (T2.5) and chloroquine (CQ) - the latter applied here as an inhibitor of murine TLR13 and its human ortholog TLR8 - abrogated activation of murine and human primary immune cells exposed to antibiotic-treated S. pneumoniae. The inhibitory effect of the T2.5/CQ cocktail was stronger than that of dexamethasone, the current standard adjunctive drug for pneumococcal meningitis. Accordingly, TLR2/TLR13 blockade concomitant with ceftriaxone application significantly improved the clinical course of pneumococcal meningitis compared with treatment with ceftriaxone alone or in combination with dexamethasone. Our study indicates the importance of murine TLR13 and human TLR8, besides TLR2, in pneumococcal meningitis pathology, and suggests their blockade as a promising antibiotic therapy adjunct.


Asunto(s)
Meningitis Neumocócica , Ratones , Humanos , Animales , Meningitis Neumocócica/tratamiento farmacológico , Meningitis Neumocócica/complicaciones , Meningitis Neumocócica/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Receptor Toll-Like 2/metabolismo , Ceftriaxona/farmacología , Ceftriaxona/uso terapéutico , Factor 88 de Diferenciación Mieloide , Receptor Toll-Like 8 , Streptococcus pneumoniae , Encéfalo/metabolismo , Dexametasona/farmacología
13.
J Immunol ; 186(8): 4845-52, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398605

RESUMEN

Filarial parasites have to trespass many barriers to successfully settle within their mammalian host, which is equipped with mechanical borders and complex weaponry of an evolved immune system. However, little is known about mechanisms of early local events in filarial infections. In this study, bone marrow-derived dendritic cells not only upregulated activation markers CD40 and CD80 upon in vitro stimulation with filarial extracts, but also secreted CCL17, a chemokine known to be produced upon microbial challenge. Mice deficient for CCL17 had an up to 4-fold higher worm burden compared with controls by day 10 of infection with the murine filaria Litomosoides sigmodontis. Also, numbers of mast cells (MCs) invading the skin and degranulation were significantly increased, which was associated with enhanced vascular permeability and larval establishment. This phenotype was reverted by inhibition of MC degranulation with disodium cromoglycate or by blockade of histamine. In addition, we showed that CCL17-mediated vascular permeability was dependent on the presence of Wolbachia endosymbionts and TLR2. Our findings reveal that CCL17 controls filarial larval entry by limiting MC-dependent vascular permeability.


Asunto(s)
Quimiocina CCL17/inmunología , Filariasis/inmunología , Filarioidea/inmunología , Mastocitos/inmunología , Animales , Antígenos Helmínticos/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Permeabilidad Capilar/inmunología , Degranulación de la Célula/inmunología , Células Cultivadas , Quimiocina CCL17/genética , Quimiocina CCL17/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Filariasis/genética , Filariasis/parasitología , Filarioidea/microbiología , Filarioidea/fisiología , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interacciones Huésped-Parásitos/inmunología , Larva/inmunología , Larva/microbiología , Larva/fisiología , Pulmón/inmunología , Pulmón/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Mastocitos/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Microscopía Confocal , Piel/inmunología , Piel/metabolismo , Factores de Tiempo , Wolbachia/inmunología
14.
Cell Mol Biol Lett ; 18(3): 433-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23893288

RESUMEN

Toll-like receptor 9 (TLR9) is a component of the innate immune system, which recognizes the DNA of both pathogens and hosts. Thus, it can drive autoimmune diseases. Intracellular antibodies expressed inside the ER block transitory protein functions by inhibiting the translocation of the protein from the ER to its subcellular destination. Here, we describe the construction and characterization of an anti-TLR9 ER intrabody (αT9ib). The respective single-chain Fv comprises the variable domains of the heavy and light chain of a monoclonal antibody (mAb; 5G5) towards human and murine TLR9. Co-expression of αT9ib and mouse TLR9 in HEK293 cells resulted in co-localization of both molecules with the ER marker calnexin. Co-immunoprecipitation of mouse TLR9 with αT9ib indicated that αT9ib interacts with its cognate antigen. The expression of αT9ib inhibited NF-κB-driven reporter gene activation upon CpG DNA challenge but not the activation of TLR3 or TLR4. Consequently, TLR9-driven TNFα production was inhibited in RAW264.7 macrophages upon transfection with the αT9ib expression plasmid. The αT9ib-encoding open reading frame was integrated into an adenoviral cosmid vector to produce the recombinant adenovirus (AdV)-αT9ib. Transduction with AdVαT9ib specifically inhibited TLR9-driven cellular TNFα release. These data strongly indicate that αT9ib is a very promising experimental tool to block TLR9 signaling.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo/inmunología , Anticuerpos de Cadena Única/inmunología , Receptor Toll-Like 9/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/inmunología , Secuencia de Bases , Calnexina/inmunología , Calnexina/metabolismo , Línea Celular , Clonación Molecular , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Citometría de Flujo , Células HEK293 , Humanos , Immunoblotting , Luciferasas/genética , Luciferasas/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Unión Proteica/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Front Immunol ; 14: 1277033, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37869001

RESUMEN

Introduction: Early sepsis is a life-threatening immune dysregulation believed to feature a "cytokine storm" due to activation of pattern recognition receptors by pathogen and danger associated molecular patterns. However, treatments with single toll-like receptor (TLR) blockers have shown no clinical benefit. We speculated that sepsis patients at the time of diagnosis are heterogeneous in relation to their cytokine production and its potential inhibition by a triple cocktail of TLR blockers. Accordingly, we analyzed inflammatory cytokine production in whole blood assays from early sepsis patients and determined the effects of triple TLR-blockade. Methods: Whole blood of 51 intensive care patients sampled within 24h of meeting Sepsis-3 criteria was incubated for 6h without or with specific TLR2, 4, and 7/8 stimuli or suspensions of heat-killed S. aureus or E. coli bacteria as pan-TLR challenges, and also with a combination of monoclonal antibodies against TLR2 and 4 and chloroquine (endosomal TLR inhibition), subsequent to dose optimization. Concentrations of tumor necrosis factor (TNF), Interleukin(IL)-6, IL-8, IL-10, IL-1α and IL-1ß were measured (multiplex ELISA) before and after incubation. Samples from 11 sex and age-matched healthy volunteers served as controls and for dose-finding studies. Results: Only a fraction of sepsis patient samples revealed ongoing cytokine production ex vivo despite sampling within 24 h of first meeting Sepsis-3 criteria. In dose finding studies, inhibition of TLR2, 4 and endosomal TLRs reliably suppressed cytokine production to specific TLR agonists and added bacteria. However, inflammatory cytokine production ex vivo was only suppressed in the high cytokine producing samples but not in the majority. The suppressive response to TLR-blockade correlated both with intraassay inflammatory cytokine production (r=0.29-0.68; p<0.0001-0.04) and cytokine baseline concentrations (r=0.55; p<0.0001). Discussion: Upon meeting Sepsis-3 criteria for less than 24 h, a mere quarter of patient samples exhibits a strong inflammatory phenotype, as characterized by increased baseline inflammatory cytokine concentrations and a stark TLR-dependent increase upon further ex vivo incubation. Thus, early sepsis patient cohorts as defined by Sepsis-3 criteria are very heterogeneous in regard to inflammation. Accordingly, proper ex vivo assays may be useful in septic individuals before embarking on immunomodulatory treatments.


Asunto(s)
Sepsis , Receptor Toll-Like 2 , Humanos , Receptor Toll-Like 2/genética , Escherichia coli , Staphylococcus aureus , Receptores Toll-Like , Citocinas , Sepsis/tratamiento farmacológico
16.
Cell Microbiol ; 13(11): 1668-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21790939

RESUMEN

Defence mechanisms against intracellular bacterial pathogens are incompletely understood. Our study characterizes a type I IFN-dependent cell-autonomous defence pathway directed against Legionella pneumophila, an intracellular model organism and frequent cause of pneumonia. We show that macrophages infected with L. pneumophila produced IFNß in a STING- and IRF3- dependent manner. Paracrine type I IFNs stimulated upregulation of IFN-stimulated genes and a cell-autonomous defence pathway acting on replicating and non-replicating Legionella within their specialized vacuole. Our infection experiments in mice lacking receptors for type I and/or II IFNs show that type I IFNs contribute to expression of IFN-stimulated genes and to bacterial clearance as well as resistance in L. pneumophila pneumonia in addition to type II IFN. Overall, our study shows that paracrine type I IFNs mediate defence against L. pneumophila, and demonstrates a protective role of type I IFNs in in vivo infections with intracellular bacteria.


Asunto(s)
Interferón Tipo I/inmunología , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/patología , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , Ratones , Vacuolas/inmunología , Vacuolas/microbiología
17.
J Immunol ; 185(10): 6179-89, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20943997

RESUMEN

The induction of type I IFN is the most immediate host response to viral infections. Type I IFN has a direct antiviral activity mediated by antiviral enzymes, but it also modulates the function of cells of the adaptive immune system. Many viruses can suppress type I IFN production, and in retroviral infections, the initial type I IFN is weak. Thus, one strategy of immunotherapy in viral infection is the exogenous induction of type I IFN during acute viral infection by TLR ligands. Along these lines, the TLR3/MDA5 ligand polyinosinic-polycytidylic acid [poly(I:C)] has already been used to treat viral infections. However, the immunological mechanisms underlying this successful therapy have not been defined until now. In this study, the Friend retrovirus (FV) mouse model was used to investigate the mode of action of poly(I:C) in antiretroviral immunotherapy. Postexposure, poly(I:C) treatment of FV-infected mice resulted in a significant reduction in viral loads and protection from virus-induced leukemia. This effect was IFN dependent because type I IFN receptor-deficient mice could not be protected by poly(I:C). The poly(I:C)-induced IFN response resulted in the expression of antiviral enzymes, which suppressed FV replication. Also, the virus-specific T cell response was augmented. Interestingly, it did not enhance the number of virus-specific CD4(+) and CD8(+) T cells, but rather the functional properties of these cells, such as cytokine production and cytotoxic activity. The results demonstrate a direct antiviral and immunomodulatory effect of poly(I:C) and, therefore, suggests its potential for clinical treatment of retroviral infections.


Asunto(s)
Antivirales/farmacología , Interferón Tipo I/biosíntesis , Poli I-C/farmacología , Infecciones por Retroviridae/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Infecciones Tumorales por Virus/tratamiento farmacológico , Animales , Femenino , Virus de la Leucemia Murina de Friend , Interferón Tipo I/inmunología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Infecciones Tumorales por Virus/inmunología , Carga Viral/efectos de los fármacos
18.
PLoS One ; 17(8): e0273247, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35981050

RESUMEN

RATIONALE: The immune profile of sepsis patients is incompletely understood and hyperinflammation and hypoinflammation may occur concurrently or sequentially. Immune checkpoint inhibition (ICI) may counter hypoinflammation but effects are uncertain. We tested the reactivity of septic whole blood to bacteria, Toll-like receptor (TLR) ligands and to ICI. METHODS: Whole blood assays of 61 patients' samples within 24h of meeting sepsis-3 criteria and 12 age and sex-matched healthy volunteers. Measurements included pattern/danger-associated molecular pattern (P/DAMP), cytokine concentrations at baseline and in response to TLR 2, 4, and 7/8 ligands, heat-inactivated Staphylococcus aureus or Escherichia coli, E.coli lipopolysaccharide (LPS), concentration of soluble and cellular immune checkpoint molecules, and cytokine concentrations in response to ICI directed against programmed-death receptor 1 (PD1), PD1-ligand 1, or cytotoxic T-lymphocyte antigen 4, both in the absence and presence of LPS. MAIN RESULTS: In sepsis, concentrations of P/DAMPs and inflammatory cytokines were increased and the latter increased further upon incubation ex vivo. However, cytokine responses to TLR 2, 4, and 7/8 ligands, heat-inactivated S. aureus or E. coli, and E. coli LPS were all depressed. Depression of the response to LPS was associated with increased in-hospital mortality. Despite increased PD-1 expression on monocytes and T-cells, and monocyte CTLA-4 expression, however, addition of corresponding checkpoint inhibitors to assays failed to increase inflammatory cytokine concentrations in the absence and presence of LPS. CONCLUSION: Patients first meeting Sepsis-3 criteria reveal 1) depressed responses to multiple TLR-ligands, bacteria, and bacterial LPS, despite concomitant inflammation, but 2) no response to immune checkpoint inhibition.


Asunto(s)
Sepsis , Receptor Toll-Like 2 , Citocinas/metabolismo , Escherichia coli/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico , Ligandos , Lipopolisacáridos , Monocitos/metabolismo , Sepsis/metabolismo , Staphylococcus aureus/metabolismo , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/metabolismo
19.
Basic Res Cardiol ; 106(1): 89-98, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20967453

RESUMEN

Restoration of myocardial blood flow after ischemia triggers an inflammatory response involving toll-like receptors (TLRs). TLR2(-/-)-mice show short-term advantages upon reperfusion injury as compared with WT controls. Accordingly, it has been shown that transient TLR2-blockade prior to reperfusion is associated with improved left-ventricular performance after myocardial scar formation. We present here adverse myocardial remodeling due to a chronic lack of TLR2 expression. Myocardial ischemia/reperfusion (MI/R) was surgically induced in C3HeN-mice by ligation of the left anterior descending coronary artery for 20 min, followed by 24 h or 28 days of reperfusion. TLR2(-/-)-mice and TLR2-Ab treated (T2.5) WT-mice displayed a reduction of infarct size, plasma troponin T concentrations, and leukocyte infiltration as compared with untreated controls after 24 h of reperfusion. After 28 days, however, magnetic resonance imaging revealed a marked left ventricular dilation in TLR2(-/-)-animals, which was associated with pronounced matrix remodeling characterized by reduced collagen and decorin density in the infarct scar. Our data show adverse effects on myocardial remodeling in TLR2(-/-)-mice. Although interception with TLR2 signaling is a promising concept for the prevention of reperfusion injury after myocardial ischemia, these data give cause for serious concern with respect to the time-point and duration of the potential treatment.


Asunto(s)
Matriz Extracelular/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/patología , Receptor Toll-Like 2/metabolismo , Cicatrización de Heridas , Animales , Biglicano/metabolismo , Colágeno/metabolismo , Decorina/metabolismo , Hipertrofia Ventricular Izquierda/etiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Miocarditis/etiología , Miocarditis/metabolismo , Miocarditis/patología , ARN Mensajero/metabolismo , Receptor Toll-Like 2/inmunología , Remodelación Ventricular
20.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33538775

RESUMEN

Human memory B cells (MBCs) are generated and diversified in secondary lymphoid tissues throughout the organism. A paired immunoglobulin (Ig)-gene repertoire analysis of peripheral blood (PB) and splenic MBCs from infant, adult, and elderly humans revealed that throughout life, circulating MBCs are comprehensively archived in the spleen. Archive MBC clones are systematically preserved and uncoupled from class-switching. Clonality in the spleen increases steadily, but boosts at midlife, thereby outcompeting small clones. The splenic marginal zone (sMZ) represents a primed MBC compartment, generated from a stochastic exchange within the archive memory pool. This is supported by functional assays, showing that PB and splenic CD21+ MBCs acquire transient CD21high expression upon NOTCH2-stimulation. Our study provides insight that the human MBC system in PB and spleen is composed of three interwoven compartments: the dynamic relationship of circulating, archive, and its subset of primed (sMZ) memory changes with age, thereby contributing to immune aging.


Asunto(s)
Envejecimiento/inmunología , Linfocitos B/inmunología , Memoria Inmunológica , Bazo/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biopsia , Donantes de Sangre , Línea Celular , Niño , Preescolar , Técnicas de Cocultivo , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Persona de Mediana Edad , Fenotipo , Receptores de Complemento 3d/metabolismo , Bazo/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA