Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Plants (Basel) ; 12(6)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36987036

RESUMEN

Arnica montana is well known for its anti-inflammatory properties. While the anti-inflammatory activity of Arnica flowers (Arnicae flos) has been extensively studied, that of the whole plant (Arnicae planta tota) is less characterized. We compared the ability of Arnicae planta tota and Arnicae flos extracts to inhibit the pro-inflammatory NF-κB-eicosanoid pathway, using several in vitro and in vivo assays. We showed that Arnicae planta tota inhibited NF-κB reporter activation, with an IC50 of 15.4 µg/mL (vs. 52.5 µg/mL for Arnicae flos). Arnicae planta tota also inhibited LPS-induced expression of ALOX5 and PTGS2 genes in human differentiated macrophages. ALOX5 and PTGS2 encode the 5-lipoxygenase (5-LO) and cyclooxygenase-2 (COX-2) enzymes that initialize the conversion of arachidonic acid into leukotrienes and prostaglandins, respectively. Arnicae planta tota inhibited 5-LO and COX-2 enzymatic activity in vitro and in human primary peripheral blood cells, with lower IC50 compared to Arnicae flos. Finally, Arnicae planta tota applied topically reduced carrageenan-induced mouse paw oedema more efficiently than Arnicae flos. Altogether, Arnicae planta tota displayed a superior anti-inflammatory activity compared to Arnicae flos, suggesting that Arnicae-planta-tota-containing products might be more effective in alleviating the manifestations of acute inflammation than those based on Arnicae flos alone.

2.
Immunobiology ; 218(7): 1005-11, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23332217

RESUMEN

Our previous studies indicated that mouse beta defensin 14 (mBD14, Defb14), a newly identified member of the beta-defensin super family, interacts with the chemokine receptors CCR2 and CCR6. In this study we report that pre-stimulation of primary mouse macrophages with mBD14 results in a synergistic, enhanced expression of pro-inflammatory cytokines and chemokines induced by TLR ligand re-stimulation. Experiments using specific inhibitors of G(i)-protein-coupled receptor signaling provide evidence that this effect seems to be mediated by a G(i)-protein-coupled receptor expressed on bone marrow derived macrophages. However, using primary macrophages derived from CCR6- and CCR2-deficient mice clearly demonstrated that the enhanced pro-inflammatory cytokine and chemokine expression is independent of the chemokine receptors CCR6 and CCR2. Additionally, signaling pathway analysis indicated that mBD14 is capable of inducing MAPK ERK1/2 phosphorylation and the induction of CD86 and F4/80 expression in bone marrow-derived macrophages after mBD14 stimulation. Collectively, our data indicate that ß-defensins activate primary macrophages and enhance pro-inflammatory responses by using G(i)PCRs in order to support inflammatory reactions induced by TLR ligands.


Asunto(s)
Citocinas/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , beta-Defensinas/farmacología , Animales , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Citocinas/biosíntesis , Citocinas/genética , Sinergismo Farmacológico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Macrófagos/inmunología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Fosforilación , Cultivo Primario de Células , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores CCR6/deficiencia , Receptores CCR6/genética , Receptores CCR6/inmunología , Proteínas Recombinantes/farmacología , Transducción de Señal , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
3.
Oncoimmunology ; 1(7): 1159-1160, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23170263

RESUMEN

For many years, ß-defensins were best known for their antimicrobial activity. However, ß-defensins also exert immunomodulatory functions, such as the chemotactic recruitment of immune cells via chemokine receptors. We demonstrated that mouse ß-defensin 14 recruits CCR6(+) B cells into fibrosarcomas, resulting in enhanced angiogenesis and tumor development.

4.
J Immunol ; 188(10): 4931-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22504651

RESUMEN

ß-Defensins are known for their antimicrobial activity and belong to the molecular barrier of the innate immune system against invading pathogens. In addition, it has been shown that some members of the ß-defensin superfamily have the capacity to promote local innate inflammatory and systemic adaptive immune responses, mediated in part by the interaction with CCR6. We found that mouse ß-defensin 14 (mBD14, Defb14), a newly identified member of the mouse ß-defensin superfamily, is expressed in mouse fibrosarcoma tumor tissue. Tumor cells overexpressing mBD14 demonstrated enhanced solid tumor growth in syngeneic C57BL/6 mice concomitant with increased vascularization of these tumors. Furthermore, mBD14-overexpressing tumors demonstrated increased expression of proangiogenic MIP-2 (CXCL2) ex vivo. In contrast, vascular endothelial growth factor expression was not affected. Cellular analysis of tumor-infiltrating leukocytes revealed a significant increase of CCR6(+) B220(+) lymphocytes in solid tumors derived from mBD14-overexpressing tumor cells. Enhanced tumor growth of mBD14-overexpressing fibrosarcomas was abolished in CCR6-deficient mice, which was paralleled by decreased infiltration of CCR6(+) B220(+) lymphocytes, indicating the requirement of CCR6 expression on host cells. Previously, the interaction of activated, LTαß(+), lymphocytes with lymphotoxin ß-receptor-expressing fibrosarcoma tumor cells has been identified as a new CXCL2-dependent proangiogenic pathway. Coexpression of a soluble lymphotoxin ß-receptor:Ig fusion protein, an inhibitor of CXCL2-dependent angiogenesis, in mBD14-overexpressing fibrosarcoma tumor cells abolished enhanced solid tumor growth. Thus, we conclude that mBD14 expression by tumor-infiltrating host cells results in the chemoattraction of CCR6(+) B220(+) lymphocytes, which in turn initiates a proangiogenic pathway leading to enhanced angiogenesis and organized tumor tissue development.


Asunto(s)
Fibrosarcoma/inmunología , Fibrosarcoma/patología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Receptores CCR6/fisiología , beta-Defensinas/fisiología , Animales , Femenino , Fibrosarcoma/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Receptores CCR6/biosíntesis , Células Tumorales Cultivadas , beta-Defensinas/biosíntesis
5.
Pediatr Surg Int ; 28(5): 507-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22426493

RESUMEN

PURPOSE: There is evidence that open as well as minimally invasive abdominal surgery impair post-operative innate and acquired immune function. To compare the impact of these approaches as well as the one of different peritoneal gas exposures on immune function, we investigated cellular as well as cytokine-based immune parameters in mesenteric lymph nodes and the spleen postoperatively. METHODS: Mice (n = 26) were randomly assigned to the 4 study groups: (1) sham controls undergoing anesthesia alone, (2) laparotomy, and (3) air, or (4) carbon dioxide pneumoperitoneum. Mice were sacrificed 48 h after the intervention, and their spleens and mesenteric lymph nodes were harvested. Cytokine production (TNF-α, IL-6, IL-10, and IFN-γ), splenic T cell subpopulations (cytotoxic T cells, T helper cells, and regulatory T cells) were analyzed. RESULTS: TNF-α production of splenocytes 16 h after ex vivo lipopolysaccharides (LPS) stimulation was significantly increased in the laparotomy group compared to all other groups. In contrast, TNF-α production of lymph node cells and IL-6 production of splenocytes after ex vivo LPS stimulation did not differ significantly between the groups. The numbers of regulatory T cells (Treg) in the spleen differed between groups. A significant reduction in Treg cell frequency was detected in the CO(2) insufflation group compared to the laparotomy and the air insufflation group. CONCLUSION: Our findings demonstrate a distinct difference in immune effector functions and cellular composition of the spleen with regard to splenic TNF-α production and increased numbers of Treg cells in the spleen. These findings are in line with a higher peritoneal inflammatory status consequent to peritoneal air rather than CO(2) exposure. Treg turned out to be key modulators of postoperative dysfunction of acquired immunity.


Asunto(s)
Citocinas/inmunología , Laparoscopía , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Mesenterio/citología , Mesenterio/inmunología , Bazo/citología , Bazo/inmunología , Linfocitos T/inmunología , Análisis de Varianza , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Laparotomía , Ratones , Neumoperitoneo Artificial , Distribución Aleatoria
6.
Mol Immunol ; 51(2): 128-35, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22437076

RESUMEN

Our previous studies indicated that LTßR activation mainly by T cell derived LTα1ß2 is crucial for the control and down-regulation of intestinal inflammation. In order to dissect the cellular and molecular role of LTßR activation in the experimental model of DSS-induced intestinal inflammation, we have generated cell type-specific LTßR-deficient mice with specific ablation of LTßR expression on macrophages/neutrophils (LTßR((flox/flox))×LysM-Cre). These mice develop an exacerbated intestinal inflammation in our experimental model indicating that LTßR expression on macrophages/neutrophils is responsible for the control and down-regulation of the inflammatory reaction. These results were verified by adoptive transfer experiments of BMDM from wild-type and LTßR-deficient mice. Furthermore, transfer of activated CD4+ T cells derived from wild-type mice, but not from LTßR ligand-deficient mice attenuated the signs of intestinal inflammation. Finally, we demonstrate that LTßR activation on BMDM results in induction of TRIM30α, a negative regulator of NFκB activation. Concordantly, ablation of LTßR signaling results in the inability to induce TRIM30α expression concomitant with an increased expression of pro-inflammatory cytokines in our experimental model. Taken together, our data demonstrate that LTßR activation on macrophages by CD4+ T cell derived LTαß controls the pro-inflammatory response by activation of a TRIM30α-dependent signaling pathway, crucial for the down-regulation of the inflammatory response in this experimental model.


Asunto(s)
Colitis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptor beta de Linfotoxina/metabolismo , Macrófagos/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Técnicas de Cocultivo , Colitis/inducido químicamente , Colitis/inmunología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Péptidos y Proteínas de Señalización Intracelular/inmunología , Receptor beta de Linfotoxina/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Immunol ; 188(7): 3426-33, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22357629

RESUMEN

Our previous studies indicated that lymphotoxin ß receptor (LTßR) activation controls and downregulates inflammatory reactions. In this study, we report that LTßR activation on primary mouse macrophages results in induction of tripartite motif containing (TRIM) 30α, which negatively regulates NF-κB activation induced by TLR signaling. LTßR activation results in a downregulation of proinflammatory cytokine and mediator expression upon TLR restimulation, demonstrating that LTßR signaling is involved in the induction of TLR cross-tolerance. Specific knockdown experiments using TRIM30α-specific small interfering RNA abolished the LTßR-dependent induction of TRIM30α and LTßR-mediated TLR cross-tolerance. Concordantly, LTßR activation on bone marrow-derived macrophages induced cross-tolerance to TLR4 and TLR9 ligands in vitro. Furthermore, we have generated cell type-specific LTßR-deficient mice with ablation of LTßR expression on macrophages/neutrophils (LTßR(flox/flox) × LysM-Cre). In bone marrow-derived macrophages derived from these mice LTßR-induced cross-tolerance to TLR4 and TLR9 ligands was impaired. Additionally, mice with a conditional ablation of LTßR expression on macrophages (LTßR(flox/flox) × LysM-Cre) are resistant to LTßR-induced TLR4 tolerance in vivo. Collectively, our data indicate that LTßR activation on macrophages by T cell-derived lymphotoxin α(1)ß(2) controls proinflammatory responses by activation of a TRIM30α-controlled, counterregulatory signaling pathway to protect against exacerbating inflammatory reactions.


Asunto(s)
Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Receptor beta de Linfotoxina/inmunología , Macrófagos/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 9/inmunología , Animales , Línea Celular/inmunología , Citocinas/biosíntesis , Citocinas/genética , Femenino , Regulación de la Expresión Génica/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Receptor beta de Linfotoxina/deficiencia , Receptor beta de Linfotoxina/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 9/agonistas
8.
Int J Cancer ; 128(6): 1363-70, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473944

RESUMEN

Lymphotoxin beta receptor (LTßR) activation on mouse fibrosarcoma cells (BFS-1) results in enhanced solid tumor growth paralleled by increased angiogenesis induced by the expression of pro-angiogenic CXCL2. In our study, we demonstrate that both functional ligands of the LTßR, namely LTα(1) ß(2) and LIGHT, are involved in the activation of LTßR in solid fibrosarcomas. To identify whether the lymphocyte population is involved in the activation of LTßR in these fibrosarcoma tumors, we used conditional LTß-deficient mice that specifically lack LTß expression either on T cells (T-LTß(-/-)) or on B cells (B-LTß(-/-)). Solid tumor growth was reduced in both mouse strains when compared to tumor growth in wild-type mice, indicating the participation of both T and B host lymphocytes in the activation of LTßR in these tumors. Tumor growth was also reduced in LIGHT-deficient mice, suggesting a contribution of this ligand to the activation of LTßR in BFS-1 fibrosarcomas. LTßR signaling can involve IκBα and/or NFκB-inducing kinase (NIK) for subsequent NFκB activation in different types of cells. Expression of a dominant negative form of IκBα or of a dominant negative mutant of NIK resulted in decreased activation of NFκB signaling and reduced expression of pro-angiogenic CXCL2 in vitro. Moreover, expression of dominant negative form of NIK or an IκBα repressor in these fibrosarcoma cells resulted in reduced solid tumor growth in vivo, suggesting that both IκBα and NIK are involved in pro-angiogenic signaling after LTßR activation. Our data support the idea that the ablation of LTßR signaling should be considered for cancer treatment.


Asunto(s)
Fibrosarcoma/patología , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/fisiología , FN-kappa B/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Linfocitos B/metabolismo , Linfocitos B/patología , Western Blotting , Proliferación Celular , Células Cultivadas , Quimiocina CXCL2/metabolismo , Femenino , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/farmacología
9.
J Immunol ; 184(12): 6688-94, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483750

RESUMEN

Beta-defensins play a dual role during immune response. Their direct antimicrobial properties contribute to the local innate immune response by combating microbial invasions. Furthermore, previous studies revealed the capacity of certain beta-defensin family members to chemoattract immature dendritic cells and CD45RO+ CD4+ T cells through chemokine receptor CCR6. However, because beta-defensins also chemoattract macrophages and monocytes, which do not express CCR6, efforts have been made to identify other receptors for these polypeptides. In this study, we demonstrate the capacity of human beta-defensin (hBD)2 and 3 and their mouse orthologs, beta-defensin 4 and 14, to interact with CCR2, a chemokine receptor expressed on monocytes, macrophages, and neutrophils. These beta-defensins, fused to the Fc region of human IgG1, showed binding to CCR2-transfected HEK293 cells, as revealed by flow cytometry. The beta-defensin fusion proteins also induced CCR2-specific chemotaxis of transfected HEK293 cells, human peripheral blood monocytes, and mouse peritoneal exudate cells in a dose-dependent manner. Preincubation of human monocytes with CCL2/MCP-1, the chemokine ligand for CCR2, abolished migration induced by beta-defensins. Conversely, preincubation with hBD2:Ig or hBD3:Ig inhibited MCP-1 induced migration. Peritoneal exudate cells from CCR2-deficient mice failed to migrate toward these fusion proteins. In conclusion, the beta-defensins used in this study contribute to the innate and adaptive immune response in their role as chemoattractants. Our data indicate that hBD2 and hBD3, together with their mouse orthologs (beta-defensin 4 and 14), are chemotactic for a broad spectrum of leukocytes in a CCR6- and CCR2-dependent manner.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Receptores CCR2/inmunología , beta-Defensinas/inmunología , Animales , Separación Celular , Citometría de Flujo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores CCR2/metabolismo , Receptores CCR6/inmunología , Receptores CCR6/metabolismo , beta-Defensinas/metabolismo
10.
J Biol Chem ; 285(10): 7028-34, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20068036

RESUMEN

Beta-defensins are small antimicrobial polypeptides that are mainly expressed by epithelial cells and play an important role in the antimicrobial innate immune response. In addition to the direct microbicidal effects of these polypeptides, members of the beta-defensin super family have the capacity to promote local innate inflammatory and systemic adaptive immune responses, which are in part mediated by the CC-chemokine receptor CCR6. Here we report the expression of recombinant mBD4 and its human orthologue hBD2 fused to the constant domain of human IgG(1) to obtain correct folding and to increase stability and solubility using the Drosophila S2 expression system. Purified recombinant mBD4:Ig and hBD2:Ig fusion proteins retained potent antimicrobial activity against Gram-negative and Gram-positive bacteria. Furthermore, these beta-defensin fusion proteins showed specific binding to CCR6-expressing cells as revealed by flow cytometry. Interestingly, although hBD2:Ig bound to both human and mouse CCR6-expressing cells, mBD4:Ig did only bind to mCCR6-expressing cells but not to hCCR6-expressing cells. Both beta-defensin fusion proteins demonstrated chemotactic activity for cells expressing the mouse CC-chemokine receptor CCR6. The chemokine ligand CCL20 competed with the beta-defensin fusion proteins for specific binding to CCR6 as analyzed by fluorescence-activated cell sorter analysis. Both beta-defensin fusion proteins demonstrated chemotactic activity for cells expressing the mouse CCR6 receptor, but mBD4:Ig did not induce chemotactic activity of cells expressing human CCR6. This result supports our finding that mBD4 does not interact with human CCR6-expressing cells. Further evidence for specific interaction of the beta-defensin fusion proteins with CCR6-expressing cells is demonstrated by the observation that CCL20 and beta-defensin fusion proteins desensitize each other in inducing chemotactic activity. In addition both mBD4:Ig and hBD2:Ig demonstrated CCR6-independent chemotaxis of freshly isolated mouse resident peritoneal cells and human peripheral blood mononuclear cells, indicating the interaction with another chemotaxis-inducing receptor. Thus, the beta-defensin fusion proteins used in this study retained their biological activity and are a feasible tool to identify and analyze specific beta-defensin receptor interactions.


Asunto(s)
Quimiotaxis/fisiología , Receptores CCR6/metabolismo , beta-Defensinas/metabolismo , Secuencia de Aminoácidos , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Línea Celular , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Escherichia coli/efectos de los fármacos , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Receptores CCR6/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , beta-Defensinas/genética , beta-Defensinas/farmacología
11.
J Biol Chem ; 283(9): 5414-9, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18167348

RESUMEN

beta-Defensins are small antimicrobial polypeptides that are mainly expressed by epithelial cells and play an important role in the antimicrobial innate immune response. In addition to the direct microbicidal effects of these polypeptides, it became evident that certain members of the beta-defensin super family have the capacity to promote local innate inflammatory and systemic adaptive immune responses by interacting with the CC-chemokine receptor CCR6. We have identified mouse beta-defensin 14 (mBD14, Defb14) as an orthologue of human beta-defensin 3 (hBD3 or DEFB103). Based on primary structural analysis, mBD14 demonstrates greater (68%) homology to its human orthologue, containing three conserved cystein linkages, characteristic for the beta-defensin super family. mBD14 is expressed in a wide variety of tissues including spleen, colon, and tissues of the upper and lower respiratory tract. Interestingly, we also detected mBD14 expression in immature CD11c+ bone marrow-derived dendritic cells. The expression of mBD14 can be induced by Toll-like receptor agonists such as lipopolysaccharide and poly(I:C) and by pro-inflammatory stimuli e.g. tumor necrosis factor and interferon-gamma. Furthermore, expression of mBD14 seems to be regulated by activation of the intracellular pattern recognition receptor NOD2/CARD15 as revealed by reporter gene analysis. We prepared a recombinant mBD14-Ig fusion protein that retained potent antimicrobial activity against several Escherichia coli strains but not against various Gram-positive Staphylococcus aureus strains. hBD3 and also the newly identified mBD14 were chemotactic for cells expressing the mouse CC-chemokine receptor CCR6. In addition, both hBD3 and mBD14 were chemotactic for freshly isolated mouse resident peritoneal cells. Thus, mBD14, based on structural and functional similarities, appears to be an orthologue of hBD3.


Asunto(s)
Regulación de la Expresión Génica/fisiología , beta-Defensinas/metabolismo , Animales , Línea Celular , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Escherichia coli/crecimiento & desarrollo , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/fisiología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inductores de Interferón/farmacología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Ratones , Proteína Adaptadora de Señalización NOD2/inmunología , Proteína Adaptadora de Señalización NOD2/metabolismo , Especificidad de Órganos/fisiología , Poli I-C/farmacología , Receptores CCR6/agonistas , Receptores CCR6/inmunología , Receptores CCR6/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Homología de Secuencia de Aminoácido , Staphylococcus aureus/crecimiento & desarrollo , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , beta-Defensinas/genética , beta-Defensinas/inmunología , beta-Defensinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...