Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gut ; 68(7): 1190-1199, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30279238

RESUMEN

OBJECTIVE: Loss of the Crohn's disease predisposing NOD2 gene results in an intestinal microenvironment conducive for colonisation by attaching-and-effacing enteropathogens. However, it remains elusive whether it relies on the intracellular recruitment of the serine-threonine kinase RIPK2 by NOD2, a step that is required for its activation of the transcription factor NF-κB. DESIGN: Colonisation resistance was evaluated in wild type and mutant mice, as well as in ex-germ-free (ex-GF) mice which were colonised either with faeces from Ripk2-deficient mice or with bacteria with similar preferences for carbohydrates to those acquired by the pathogen. The severity of the mucosal pathology was quantified at several time points postinfection by using a previously established scoring. The community resilience in response to infection was evaluated by 16S ribosomal RNA gene sequence analysis. The control of pathogen virulence was evaluated by monitoring the secretion of Citrobacter-specific antibody response in the faeces. RESULTS: Primary infection was similarly outcompeted in ex-GF Ripk2-deficient and control mice, demonstrating that the susceptibility to infection resulting from RIPK2 deficiency cannot be solely attributed to specific microbiota community structures. In contrast, delayed clearance of Citrobacter rodentium and exacerbated histopathology were preceded by a weakened propensity of intestinal macrophages to afford innate lymphoid cell activation. This tissue protection unexpectedly required the regenerating family member 3ß by instigating interleukin (IL) 17A-mediated neutrophil recruitment to the intestine and subsequent phosphorylation of signal transducer and activator of transcription 3. CONCLUSIONS: These results unveil a previously unrecognised mechanism that efficiently protects from colonisation by diarrhoeagenic bacteria early in infection.


Asunto(s)
Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , Infecciones por Enterobacteriaceae/prevención & control , Interleucina-17/fisiología , Infiltración Neutrófila/fisiología , Proteína Adaptadora de Señalización NOD2/fisiología , Animales , Proteínas Adaptadoras de Señalización CARD/fisiología , Citrobacter rodentium , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/patología , Mucosa Intestinal/patología , Ratones , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Transducción de Señal
2.
J Immunol ; 198(3): 1297-1307, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011933

RESUMEN

During macrophage activation, expression of IL-1R-associated kinase (IRAK)-M is induced to suppress TLR-mediated responses and is a hallmark of endotoxin tolerance. Endotoxin tolerance requires tight regulation of genes occurring at the transcriptional and epigenetic levels. To identify novel regulators of IRAK-M, we used RAW 264.7 macrophages and performed a targeted RNA interference screen of genes encoding chromatin-modifying enzymes, signaling molecules, and transcription factors involved in macrophage activation. Among these, the transcription factor CCAAT/enhancer binding protein (C/EBP)ß, known to be involved in macrophage inactivation, was necessary for the induction of IRAK-M expression. Chromatin immunoprecipitation showed that C/EBPß was recruited to the IRAK-M promoter following LPS stimulation and was indispensable for IRAK-M transcriptional activation. Among histone 3-modifying enzymes, our screen showed that knockdown of the histone 3 lysine 27 (H3K27) methyltransferase and part of the polycomb recessive complex 2, enhancer of Zeste 2, resulted in IRAK-M overexpression. In contrast, knockdown of the H3K27 demethylase ubiquitously transcribed tetratricopeptide repeat X chromosome suppressed the induction of IRAK-M in response to LPS stimulation. Accordingly, we demonstrated that H3K27 on the IRAK-M promoter is trimethylated in unstimulated cells and that this silencing epigenetic mark is removed upon LPS stimulation. Our data propose a mechanism for IRAK-M transcriptional regulation according to which, in the naive state, polycomb recessive complex 2 repressed the IRAK-M promoter, allowing low levels of expression; following LPS stimulation, the IRAK-M promoter is derepressed, and transcription is induced to allow its expression.


Asunto(s)
Epigénesis Genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Macrófagos/metabolismo , Transcripción Genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Células Cultivadas , Remoción de Radical Alquila , Lipopolisacáridos/farmacología , Ratones , FN-kappa B/fisiología , Regiones Promotoras Genéticas
3.
Gut ; 67(10): 1836-1844, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28790160

RESUMEN

OBJECTIVE: In association with innate and adaptive immunity, the microbiota controls the colonisation resistance against intestinal pathogens. Caspase recruitment domain 9 (CARD9), a key innate immunity gene, is required to shape a normal gut microbiota. Card9-/- mice are more susceptible to the enteric mouse pathogen Citrobacter rodentium that mimics human infections with enteropathogenic and enterohaemorrhagic Escherichia coli. Here, we examined how CARD9 controls C. rodentium infection susceptibility through microbiota-dependent and microbiota-independent mechanisms. DESIGN: C. rodentium infection was assessed in conventional and germ-free (GF) wild-type (WT) and Card9-/- mice. To explore the impact of Card9-/-microbiota in infection susceptibility, GF WT mice were colonised with WT (WT→GF) or Card9-/- (Card9-/- →GF) microbiota before C. rodentium infection. Microbiota composition was determined by 16S rDNA gene sequencing. Inflammation severity was determined by histology score and lipocalin level. Microbiota-host immune system interactions were assessed by quantitative PCR analysis. RESULTS: CARD9 controls pathogen virulence in a microbiota-independent manner by supporting a specific humoral response. Higher susceptibility to C. rodentium-induced colitis was observed in Card9-/- →GF mice. The microbiota of Card9-/- mice failed to outcompete the monosaccharide-consuming C. rodentium, worsening the infection severity. A polysaccharide-enriched diet counteracted the ecological advantage of C. rodentium and the defective pathogen-specific antibody response in Card9-/- mice. CONCLUSIONS: CARD9 modulates the susceptibility to intestinal infection by controlling the pathogen virulence in a microbiota-dependent and microbiota-independent manner. Genetic susceptibility to intestinal pathogens can be overridden by diet intervention that restores humoural immunity and a competing microbiota.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Colitis , Microbioma Gastrointestinal/fisiología , Polisacáridos , Inmunidad Adaptativa/fisiología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Citrobacter rodentium/efectos de los fármacos , Citrobacter rodentium/patogenicidad , Colitis/inmunología , Colitis/microbiología , Dietoterapia/métodos , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/fisiología , Ratones , Polisacáridos/efectos adversos , Polisacáridos/metabolismo , Virulencia/fisiología
4.
Immunity ; 31(2): 220-31, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19699171

RESUMEN

MicroRNAs regulated by lipopolysaccharide (LPS) target genes that contribute to the inflammatory phenotype. Here, we showed that the protein kinase Akt1, which is activated by LPS, positively regulated miRNAs let-7e and miR-181c but negatively regulated miR-155 and miR-125b. In silico analyses and transfection studies revealed that let-7e repressed Toll-like receptor 4 (TLR4), whereas miR-155 repressed SOCS1, two proteins critical for LPS-driven TLR signaling, which regulate endotoxin sensitivity and tolerance. As a result, Akt1(-/-) macrophages exhibited increased responsiveness to LPS in culture and Akt1(-/-) mice did not develop endotoxin tolerance in vivo. Overexpression of let-7e and suppression of miR-155 in Akt1(-/-) macrophages restored sensitivity and tolerance to LPS in culture and in animals. These results indicate that Akt1 regulates the response of macrophages to LPS by controlling miRNA expression.


Asunto(s)
Lipopolisacáridos/inmunología , Macrófagos/inmunología , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Tolerancia Inmunológica/inmunología , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Receptor Toll-Like 4/inmunología
5.
J Vis Exp ; (200)2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37870303

RESUMEN

The field of Adoptive Cell Therapy (ACT) has been revolutionized by the development of genetically modified cells, specifically Chimeric Antigen Receptor (CAR)-T cells. These modified cells have shown remarkable clinical responses in patients with hematologic malignancies. However, the high cost of producing these therapies and conducting extensive quality control assessments has limited their accessibility to a broader range of patients. To address this issue, many academic institutions are exploring the feasibility of in-house manufacturing of genetically modified cells, while adhering to guidelines set by national and international regulatory agencies. Manufacturing genetically modified T cell products on a large scale presents several challenges, particularly in terms of the institution's production capabilities and the need to meet infusion quantity requirements. One major challenge involves producing large-scale viral vectors under Good Manufacturing Practice (GMP) guidelines, which is often outsourced to external companies. Additionally, simplifying the T cell transduction process can help minimize variability between production batches, reduce costs, and facilitate personnel training. In this study, we outline a streamlined process for lentiviral transduction of primary human T cells with a fluorescent marker as the gene of interest. The entire process adheres to GMP-compliant standards and is implemented within our academic institution.


Asunto(s)
Inmunoterapia Adoptiva , Linfocitos T , Humanos , Inmunoterapia Adoptiva/métodos , Tratamiento Basado en Trasplante de Células y Tejidos
6.
Front Med (Lausanne) ; 10: 1166871, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37275377

RESUMEN

Regulatory T-cell (Treg) immunotherapy has emerged as a promising and highly effective strategy to combat graft-versus-host disease (GvHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Both naturally occurring Treg and induced Treg populations have been successfully evaluated in trials illustrating the feasibility, safety, and efficacy required for clinical translation. Using a non-mobilized leukapheresis, we have developed a good manufacturing practice (GMP)-compatible induced Treg product, termed iG-Tregs, that is enriched in cells expressing the potent immunosuppressive human leucocyte antigen-G molecule (HLA-G+). To assess the safety and the maximum tolerable dose (MTD) of iG-Tregs, we conduct a phase I-II, two-center, interventional, dose escalation (3 + 3 design), open-label study in adult patients undergoing allo-HCT from an HLA-matched sibling donor, which serves also as the donor for iG-Treg manufacturing. Herein, we present the clinical protocol with a detailed description of the study rationale and design as well as thoroughly explain every step from patient screening, product manufacturing, infusion, and participant follow-up to data collection, management, and analysis (registered EUDRACT-2021-006367-26).

7.
J Immunol ; 182(10): 6444-51, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414798

RESUMEN

High levels of plasma adiponectin are associated with low levels of inflammatory markers and cardioprotection. The mechanism via which adiponectin exerts its anti-inflammatory effect is yet unknown. In the present study, we demonstrate that globular adiponectin (gAd) induces the expression of the inactive isoform of IL-1R-associated kinases (IRAK), IRAK-M. Homologous deletion of IRAK-M in IRAK-M(-/-) mice abolished the tolerogenic properties of gAd because pretreatment of IRAK-M(-/-) macrophages with gAd did not suppress LPS-induced proinflammatory cytokine production. GAd activated the MAPKs MEK1/2 and ERK1/2 in macrophages via their upstream regulator Tpl2. Activation of ERK1/2 via Tpl2 appeared necessary for the induction of IRAK-M because gAd did not induce IRAK-M in Tpl2(-/-) macrophages or in macrophages pretreated with the MEK1/2 inhibitor UO126. In addition, activation of PI3K and Akt1 also appeared necessary for the induction of IRAK-M by gAd, because treatment of Akt1(-/-) macrophages or pretreatment of macrophages with the PI3K inhibitor wortmannin abolished gAd-induced IRAK-M expression. Analysis of IRAK-M expression in human peripheral blood cells confirmed that serum adiponectin was negatively associated with IRAK-M and responsiveness to LPS. In conclusion, our data demonstrate that IRAK-M is a major mediator of gAd-induced endotoxin tolerance in primary macrophages, expression of which depends on the activation of Tpl2/ERK and PI3K/Akt1 signaling pathways.


Asunto(s)
Adiponectina/inmunología , Endotoxinas/inmunología , Tolerancia Inmunológica , Quinasas Asociadas a Receptores de Interleucina-1/biosíntesis , Macrófagos/inmunología , Adiponectina/metabolismo , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/inmunología , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología
8.
Proc Natl Acad Sci U S A ; 105(8): 2987-92, 2008 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18287049

RESUMEN

The protein kinase encoded by the Tpl2 protooncogene plays an obligatory role in the transduction of Toll-like receptor and death receptor signals in macrophages, B cells, mouse embryo fibroblasts, and epithelial cells in culture and promotes inflammatory responses in animals. To address its role in T cell activation, we crossed the T cell receptor (TCR) transgene 2C, which recognizes class I MHC presented peptides, into the Tpl2(-/-) genetic background. Surprisingly, the TCR2C(tg/tg)/Tpl2(-/-) mice developed T cell lymphomas with a latency of 4-6 months. The tumor cells were consistently TCR2C(+)CD8(+)CD4(-), suggesting that they were derived either from chronically stimulated mature T cells or from immature single positive (ISP) cells. Further studies showed that the population of CD8(+) ISP cells was not expanded in the thymus of TCR2C(tg/tg)/Tpl2(-/-) mice, making the latter hypothesis unlikely. Mature peripheral T cells of Tpl2(-/-) mice were defective in ERK activation and exhibited enhanced proliferation after TCR stimulation. The same cells were defective in the induction of CTLA4, a negative regulator of the T cell response, which is induced by TCR signals via ERK. These findings suggest that Tpl2 functions normally in a feedback loop that switches off the T cell response to TCR stimulation. As a result, Tpl2, a potent oncogene, functions as a tumor suppressor gene in chronically stimulated T cells.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Western Blotting , Antígeno CTLA-4 , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Citometría de Flujo , Inmunohistoquímica , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/inmunología , Linfocitos T/metabolismo , Transgenes/genética
9.
Pediatr Res ; 66(2): 179-84, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19390489

RESUMEN

During infections, pathogens bind to toll-like receptor (TLR)4 and CD14 receptors and induce cytokine release, leading to inflammation. Here, we investigated TLR4 and CD14 expression on peripheral blood leukocytes (PBLs) and their roles in lipopolysaccharide (LPS)-induced cytokine and chemokine release. Full-term and preterm neonates and adults were studied. PBLs were pretreated with anti-TLR4- and anti-CD14-blocking antibodies and stimulated with LPS. Cytokine and chemokine levels were measured in supernatants. TLR4, CD14 expression, and LPS-induced CXCL8 release were higher in neonates, possibly contributing to aberrant inflammation. TLR4 blockade resulted in approximately 3-fold greater suppression of LPS-induced CXCL8 release in preterm neonates (38%) than in adults (14%). CD14 blockade (approximately 80%) in neonates induced approximately 3-fold greater inhibition of CXCL8 release, compared with anti-TLR4 (approximately 30%). Anti-TLR4 partly (50-60%) inhibited IL-10 and TNF-alpha, whereas anti-CD14 completely suppressed their release. Our findings reveal that neonates depend more on TLR4 for CXCL8 release. Furthermore, neonatal LPS-induced CXCL8 release, apart from TLR4/CD14-mediated signaling, is regulated by LPS interactions with other TLRs and/or immune receptors. IL-10 and TNF-alpha release depends on LPS binding not only to CD14/TLR4 but also to CD14 associated with another TLR. Our findings reveal the contribution of TLR4 and CD14 in neonatal cytokine and chemokine release and could aid in design of antagonists to prevent harmful inflammation.


Asunto(s)
Recién Nacido/inmunología , Inflamación/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/inmunología , Receptor Toll-Like 4/inmunología , Adulto , Animales , Línea Celular , Citocinas/inmunología , Humanos , Interleucina-10/inmunología , Interleucina-8/inmunología , Leucocitos Mononucleares/inmunología , Factor de Necrosis Tumoral alfa/inmunología
10.
Mol Immunol ; 45(10): 2970-80, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18336909

RESUMEN

Toll-like receptor 4 (TLR4) recognizes and initiates signals from Gram-negative bacterial lipopolysaccharide (LPS) triggering the inflammatory response. Expression levels of TLR4 on macrophages partly regulate the magnitude of the response to LPS. Vasoactive Intestinal Peptide (VIP) is known to block inflammatory responses by inhibiting pro-inflammatory cytokine production from activated macrophages. In the present report we demonstrate that VIP directly suppressed TLR4 expression on naïve primary mouse macrophages utilizing signalling cascades that control TLR4 transcription. VIP-induced suppression of TLR4 occurred at the transcriptional level by decreasing PU.1 DNA binding. Mutation of the proximal PU.1 but not the AP-1-binding site on the TLR4 promoter abrogated VIP-induced suppression of TLR4 transcription. Moreover, inhibition of PI3K by wortmannin or homologous deletion of the Akt1 isoform, a pathway known to act as a negative regulator of macrophage activation, alleviated the suppressive action of VIP on TLR4 expression. To evaluate the biological significance of VIP effect on TLR4 expression, Raw264.7 macrophages were pre-treated with VIP for 24h and then exposed to LPS. Pre-treatment with VIP rendered macrophages hypo-responsive to LPS resulting in reduced pro-inflammatory cytokine production. Moreover, in vivo administration of VIP in C57BL/6 mice resulted in lower IL-6 production upon treatment with LPS. Overall, the results indicate that VIP promotes endotoxin tolerance by downregulating TLR4 expression via Akt1.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/enzimología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Toll-Like 4/inmunología , Péptido Intestinal Vasoactivo/farmacología , Animales , Línea Celular , Citocinas/biosíntesis , ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Mediadores de Inflamación , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Toll-Like 4/genética , Transactivadores/metabolismo , Transcripción Genética/efectos de los fármacos
11.
Ann N Y Acad Sci ; 1083: 185-95, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17148740

RESUMEN

Several recently published reports, including ours, suggest that adiponectin is a strong proinflammatory agent. Indeed, exposure of human placenta and adipose tissue to adiponectin induces the production of interleukin-1beta (IL-1beta), IL-6, tumor necrosis factor alpha (TNF-alpha), and prostaglandin E2 (PGE2). We have previously shown that adiponectin is a powerful inducer of proinflammatory cytokines production by macrophages. The reported anti-inflammatory effect of adiponectin may be due to the induction of macrophage tolerance to further adiponectin exposure or to other proinflammatory stimuli including the Toll-like receptor (TLR) 3 ligand polyI:C and the TLR4 ligand lipopolysaccharide (LPS). We now present additional data supporting the hypothesis that adiponectin is a strong proinflammatory adipokine. More specifically, we demonstrate that adiponectin induces IL-1beta and IL-8 from THP-1 macrophage cell line. The effect of adiponectin is not restricted to differentiated THP-1 macrophages but it is evident at lower levels in undifferentiated THP-1 monocytes promoting TNF-alpha, IL-6, and IL-8 production. Thus, its high levels in the circulation of lean subjects render their macrophages resistant to several proinflammatory stimuli including its own thus acting in effect as an anti-inflammatory agent. Lowering of its high levels, as a consequence of increased body mass index (BMI), renders macrophages sensitive to any proinflammatory insult.


Asunto(s)
Adiponectina/fisiología , Citocinas/biosíntesis , Síndrome Metabólico/inmunología , Adiponectina/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Inflamación/inmunología , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Interleucina-8/biosíntesis , Macrófagos/metabolismo , Monocitos/metabolismo , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
12.
Biochem Biophys Res Commun ; 335(4): 1254-63, 2005 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-16115611

RESUMEN

Adiponectin exerts anti-inflammatory effects via macrophages, suppressing the production of pro-inflammatory cytokines in response to bacterial lipopolysaccharide (LPS). Here, we provide experimental evidence that the "anti-inflammatory" effect of adiponectin may be due to an induction of macrophage tolerance: globular adiponectin (gAd) is a powerful inducer of TNF-alpha and IL-6 secretion in primary human peripheral macrophages, in the THP-1 human macrophage cell line, and in primary mouse peritoneal macrophages. Pre-exposure of macrophages to 10 microg/ml gAd rendered them tolerant to further gAd exposure or to other pro-inflammatory stimuli such as TLR3 ligand polyI:C and TLR4 ligand LPS, while pre-exposure to 1 microg/ml of and re-exposure to 10 microg/ml gAd unmasked its pro-inflammatory properties. GAd induced NF-kappaB activation and tolerance to further gAd or LPS exposure. Our data suggest that adiponectin constant presence in the circulation in high levels (in lean subjects) renders macrophages resistant to pro-inflammatory stimuli, including its own.


Asunto(s)
Citocinas/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Interleucina-6/inmunología , Macrófagos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adiponectina , Línea Celular , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/inducido químicamente , Lipopolisacáridos , Macrófagos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA