Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Dev Comp Immunol ; 49(2): 259-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25498541

RESUMEN

Soluble colony stimulating factor-1 receptor (sCSF-1R) is a novel bony fish protein that contributes to the regulation of macrophage proliferation. We recently showed that this soluble receptor is highly upregulated by teleost macrophages in the presence of apoptotic cells. Further, recombinant sCSF-1R inhibited leukocyte infiltration into a challenge site in vivo. Herein, we characterized the mechanisms underlying these changes as a platform to better understand the evolutionary origins of the CSF-1 immune-regulatory axis and inflammation control in teleosts. Using an in vivo model of self-resolving peritonitis, we show that sCSF-1R downregulates chemokine expression and inhibits neutrophil chemotaxis. Soluble CSF-1R also inhibited gene expression of several pro-inflammatory cytokines and promoted the expression of an anti-inflammatory mediator, IL-10. Finally, the phenotype of infiltrating neutrophils changed significantly in the presence of sCSF-1R. Both a reduced capacity for phagocytosis and pathogen killing were observed. Overall, our results implicate sCSF-1R as an important regulator of neutrophil responses in teleosts. It remains unclear whether this represents an inflammation regulatory factor that is unique to this animal group or one that may be evolutionarily conserved and continues to contribute to the regulation of antimicrobial processes at inflammatory sites in higher vertebrates.


Asunto(s)
Citocinas/biosíntesis , Carpa Dorada/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Aeromonas/inmunología , Animales , Apoptosis/inmunología , Inhibición de Migración Celular/inmunología , Células Cultivadas , Quimiotaxis/inmunología , Proteínas de Peces/inmunología , Inmunomodulación/inmunología , Interleucina-10/biosíntesis , Macrófagos/inmunología , Infiltración Neutrófila/inmunología , Peritonitis/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología
2.
J Immunol ; 193(3): 1416-26, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981451

RESUMEN

Airway smooth muscle cell (ASMC) migration is an important mechanism postulated to play a role in airway remodeling in asthma. CXCL1 chemokine has been linked to tissue growth and metastasis. In this study, we present a detailed examination of the inhibitory effect of CXCL1 on human primary ASMC migration and the role of the decoy receptor, Duffy AgR for chemokines (DARC), in this inhibition. Western blots and pathway inhibitors showed that this phenomenon was mediated by activation of the ERK-1/2 MAPK pathway, but not p38 MAPK or PI3K, suggesting a biased selection in the signaling mechanism. Despite being known as a nonsignaling receptor, small interference RNA knockdown of DARC showed that ERK-1/2 MAPK activation was significantly dependent on DARC functionality, which, in turn, was dependent on the presence of heat shock protein 90 subunit α. Interestingly, DARC- or heat shock protein 90 subunit α-deficient ASMCs responded to CXCL1 stimulation by enhancing p38 MAPK activation and ASMC migration through the CXCR2 receptor. In conclusion, we demonstrated DARC's ability to facilitate CXCL1 inhibition of ASMC migration through modulation of the ERK-1/2 MAPK-signaling pathway.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Inhibición de Migración Celular/inmunología , Quimiocina CXCL1/fisiología , Sistema del Grupo Sanguíneo Duffy/fisiología , Receptores de Superficie Celular/fisiología , Receptores de Interleucina-8B/fisiología , Biomarcadores/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/fisiología , Sistema del Grupo Sanguíneo Duffy/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/inmunología , Cultivo Primario de Células , Receptores de Superficie Celular/metabolismo , Receptores de Interleucina-8B/metabolismo
3.
J Leukoc Biol ; 94(6): 1317-23, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23975892

RESUMEN

IPF is a chronic, progressive pulmonary disease, leading to respiratory failure. In search of mechanisms of IPF, we used the bleomycin-induced lung-injury model in mice, which causes acute inflammation that may progress to chronic lung inflammation and fibrosis. Here, we asked whether CXCL6/GCP-2, a member of the CXC chemokine superfamily, may be involved in IPF development. First, we reported an increase of CXCL6 levels in BALF from patients with IPF, as well as in the lung of mice, 24 h after bleomycin administration. To investigate whether CXCL6 played a role in experimental bleomycin-induced pulmonary fibrosis, we treated mice with an anti-mCXCL6 mAb that has been shown to inhibit neutrophil chemotaxis in vitro. CXCL6 antibody blockade attenuated acute inflammation with a reduced pulmonary neutrophil influx, IL-1ß, CXCL1, and TIMP-1 production. In the later phase (14 days after bleomycin exposure), lymphocyte recruitment and fibrosis markers, such as collagen and TIMP-1, were diminished, as well as collagen deposition and fibrotic lesion the lung. Therefore, the data suggest that CXCL6 contributes to experimental pulmonary fibrosis, and CXCL6 inhibition might be used to reduce lung toxicity associated with bleomycin treatment.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Neutralizantes/farmacología , Bleomicina/efectos adversos , Quimiocina CXCL6/antagonistas & inhibidores , Neumonía/inmunología , Fibrosis Pulmonar/inmunología , Animales , Antibióticos Antineoplásicos/farmacología , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Neutralizantes/inmunología , Bleomicina/farmacología , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Quimiocina CXCL6/inmunología , Quimiocina CXCL6/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología
4.
J Immunol ; 191(1): 424-33, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23729444

RESUMEN

Glucocorticoid-induced leucine zipper (GILZ) is an anti-inflammatory protein first identified in T lymphocytes. We recently observed that GILZ is highly expressed in synovial endothelial cells in rheumatoid arthritis. However, the function of GILZ in endothelial cells is unknown. To investigate the actions of GILZ in this cell type, we induced GILZ expression in HUVECs via transient transfection. GILZ overexpression significantly reduced the capacity of TNF-stimulated HUVECs to support leukocyte rolling, adhesion, and transmigration. These effects were associated with decreased expression of E-selectin, ICAM-1, CCL2, CXCL8, and IL-6. Experiments in a human microvascular endothelial cell line demonstrated that TNF-inducible NF-κB activity was significantly inhibited by overexpression of GILZ. Exogenous GILZ inhibited TNF-induced NF-κB p65 DNA binding, although this occurred in the absence of an effect on p65 nuclear translocation, indicating that the mechanism of action of exogenous GILZ in endothelial cells differs from that reported in other cell types. GILZ overexpression also inhibited TNF-induced activation of p38, ERK, and JNK MAPKs, as well as increased expression of the MAPK inhibitory phosphatase, MKP-1. In contrast, silencing endogenous GILZ in glucocorticoid-treated HUVECs did not alter their capacity to support leukocyte interactions. These data demonstrate that exogenous GILZ exerts inhibitory effects on endothelial cell adhesive function via a novel pathway involving modulation of NF-κB p65 DNA binding and MAPK activity. Induction of GILZ expression in endothelial cells may represent a novel therapeutic modality with the potential to inhibit inflammatory leukocyte recruitment.


Asunto(s)
Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/genética , Migración Transendotelial y Transepitelial/inmunología , Adhesión Celular/genética , Adhesión Celular/inmunología , Comunicación Celular/inmunología , Línea Celular , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Endotelio Vascular/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Microcirculación/genética , Microcirculación/inmunología , Cultivo Primario de Células , Distribución Aleatoria , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología , Migración Transendotelial y Transepitelial/genética
5.
J Immunol ; 191(1): 395-406, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23733875

RESUMEN

Cyclooxygenase (Cox)-2 dependent PGs modulate several functions in many pathophysiological processes, including migration of immune cells. In this study, we addressed the role of Cox-2 in macrophage migration by using in vivo and in vitro models. Upon thioglycolate challenge, CD11b(+) F4/80(+) macrophages showed a diminished ability to migrate to the peritoneal cavity in cox-2(-/-) mice. In vivo migration of cox-2(-/-) macrophages from the peritoneal cavity to lymph nodes, as well as cell adhesion to the mesothelium, was reduced in response to LPS. In vitro migration of cox-2(-/-) macrophages toward MCP-1, RANTES, MIP-1α, or MIP-1ß, as well as cell adhesion to ICAM-1 or fibronectin, was impaired. Defects in cell migration were not due to changes in chemokine receptor expression. Remarkably, cox-2(-/-) macrophages showed a deficiency in focal adhesion formation, with reduced phosphorylation of paxillin (Tyr(188)). Interestingly, expression of the p110γ catalytic subunit of PI3K was severely reduced in the absence of Cox-2, leading to defective Akt phosphorylation, as well as cdc42 and Rac-1 activation. Our results indicate that the paxillin/p110γ-PI3K/Cdc42/Rac1 axis is defective in cox-2(-/-) macrophages, which results in impaired cell adhesion and migration.


Asunto(s)
Inhibición de Migración Celular/inmunología , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Ciclooxigenasa 2/deficiencia , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Fosfatidilinositol 3-Quinasas/deficiencia , Transducción de Señal/inmunología , Animales , Adhesión Celular/genética , Adhesión Celular/inmunología , Inhibición de Migración Celular/genética , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/fisiología , Macrófagos Peritoneales/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/genética , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP rac1/deficiencia
6.
PLoS One ; 7(11): e47578, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226199

RESUMEN

In multiple sclerosis (MS), treatment with the monoclonal antibody natalizumab effectively reduces the formation of acute lesions in the central nervous system (CNS). Natalizumab binds the integrin very late antigen (VLA)-4, expressed on the surface of immune cells, and inhibits VLA-4 dependent transmigration of circulating immune-cells across the vascular endothelium into the CNS. Recent studies suggested that natalizumab treated MS patients have an increased T-cell pool in the blood compartment which may be selectively enriched in activated T-cells. Proposed causes are sequestration of activated T-cells due to reduced extravasation of activated and pro-inflammatory T-cells or due to induction of VLA-4 mediated co-stimulatory signals by natalizumab. In this study we examined how natalizumab treatment altered the distribution of effector and memory T-cell subsets in the blood compartment and if T-cells in general or myelin-reactive T-cells in particular showed signs of increased immune activation. Furthermore we examined the effects of natalizumab on CD4(+) T-cell responses to myelin in vitro. Natalizumab-treated MS patients had significantly increased numbers of effector-memory T-cells in the blood. In T-cells from natalizumab-treated MS patients, the expression of TNF-α mRNA was increased whereas the expression of fourteen other effector cytokines or transcription factors was unchanged. Natalizumab-treated MS patients had significantly decreased expression of the co-stimulatory molecule CD134 on CD4(+)CD26(HIGH) T-cells, in blood, and natalizumab decreased the expression of CD134 on MBP-reactive CD26(HIGH)CD4(+) T-cells in vitro. Otherwise CD4(+) T-cells from natalizumab-treated and untreated MS patients showed similar responses to MBP. In conclusion natalizumab treatment selectively increased the effector memory T-cell pool but not the activation state of T-cells in the blood compartment. Myelin-reactive T-cells were not selectively increased in natalizumab treated MS.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales Humanizados/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Memoria Inmunológica/efectos de los fármacos , Integrina alfa4beta1/genética , Integrina alfa4beta1/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/inmunología , Natalizumab , Receptores OX40/genética , Receptores OX40/inmunología , Transducción de Señal/efectos de los fármacos , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Factor de Necrosis Tumoral alfa/farmacología
7.
J Immunol ; 189(12): 5886-95, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23129756

RESUMEN

Chemokine receptor cross-desensitization provides an important mechanism to regulate immune cell recruitment at sites of inflammation. We previously reported that the mycobacterial cell wall glycophospholipid mannose-capped lipoarabinomannan (ManLAM) could induce human peripheral blood T cell chemotaxis. Therefore, we examined the ability of ManLAM to desensitize T cells to other chemoattractants as a potential mechanism for impaired T cell homing and delayed lung recruitment during mycobacterial infection. We found that ManLAM pretreatment inhibited in vitro migration of naive human or mouse T cells to the lymph node egress signal sphingosine-1-phosphate (S1P). Intratracheal administration of ManLAM in mice resulted in significant increases in T cells, primarily CCR5(+) (Th1) cells, in lung-draining lymph nodes. To investigate the selective CCR5 effect, mouse T cells were differentiated into Th1 or Th2 populations in vitro, and their ability to migrate to S1P with or without ManLAM pretreatment was analyzed. ManLAM pretreatment of Th1 populations inhibited S1P-induced migration but had no effect on Th2 cell S1P-directed migration, suggesting a differential effect by S1P on the two subsets. The PI3K/AKT inhibitor Ly294002 inhibited S1P-directed migration by Th1 cells, whereas the ERK inhibitor U0126 inhibited Th2 cell S1P-directed migration. These observations demonstrate that S1P-induced migratory responses in Th1 and Th2 lymphocytes occurs via different signaling pathways and suggests further that the production of ManLAM during Mycobacterium tuberculosis infection may function to sequester Th1 cells in lung-draining lymph nodes, thereby delaying their recruitment to the lung.


Asunto(s)
Inhibición de Migración Celular/inmunología , Lipopolisacáridos/fisiología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Animales , Antígenos Bacterianos/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Femenino , Humanos , Activación de Linfocitos/inmunología , Manosa/fisiología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/química , Células TH1/citología , Células TH1/metabolismo
8.
J Immunol ; 189(12): 5612-21, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23136203

RESUMEN

Vaccine adjuvant-induced inflammation augments vaccine immunity in part by recruiting APCs to vaccine draining lymph nodes (LNs). However, the role of one APC subtype, inflammatory monocytes, in regulating vaccine immunity in healthy animals has not been fully examined in detail. Therefore, vaccine-mediated monocyte recruitment and subsequent immune responses were investigated using murine vaccination models and in vitro assays. Recruitment of inflammatory monocytes to vaccine draining LNs was rapid and mediated primarily by local production of MCP-1, as revealed by studies in MCP-1(-/-) mice. Interrupting monocyte recruitment to LNs by either transient monocyte depletion or monocyte migration blockade led to marked amplification of both cellular and humoral immune responses to vaccination. These results were most consistent with the idea that rapidly mobilized inflammatory monocytes were actually suppressing vaccine responses. The suppressive nature of vaccine-elicited monocytes was confirmed using in vitro cocultures of murine monocytes and T cells. Furthermore, it was determined that inflammatory monocytes suppressed T cell responses by sequestering cysteine, as cysteine supplementation in vitro and in vivo appreciably augmented vaccine responses. These findings indicated, therefore, that vaccination-elicited inflammation, although necessary for effective immunity, also generated potent counter-regulatory immune responses that were mediated primarily by inflammatory monocytes. Therefore, interrupting monocyte-mediated vaccine counterregulatory responses may serve as an effective new strategy for broadly amplifying vaccine immunity.


Asunto(s)
Vacunas contra el Cáncer/antagonistas & inhibidores , Vacunas contra el Cáncer/inmunología , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Monocitos/patología , Vacunas de ADN/antagonistas & inhibidores , Vacunas de ADN/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Cationes , Línea Celular Tumoral , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Cisteína/administración & dosificación , Tolerancia Inmunológica/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Monocitos/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/deficiencia , Receptores CCR2/genética , Vacunas de ADN/administración & dosificación
9.
J Immunol ; 189(1): 411-24, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661092

RESUMEN

Mononuclear cell migration into the vascular subendothelium constitutes an early event of the atherogenic process. Because the effect of retinoid X receptor (RXR)α on arterial mononuclear leukocyte recruitment is poorly understood, this study investigated whether RXR agonists can affect this response and the underlying mechanisms involved. Decreased RXRα expression was detected after 4 h stimulation of human umbilical arterial endothelial cells with TNF-α. Interestingly, under physiological flow conditions, TNF-α-induced endothelial adhesion of human mononuclear cells was concentration-dependently inhibited by preincubation of the human umbilical arterial endothelial cells with RXR agonists such as bexarotene or 9-cis-retinoid acid. RXR agonists also prevented TNF-α-induced VCAM-1 and ICAM-1 expression, as well as endothelial growth-related oncogene-α and MCP-1 release. Suppression of RXRα expression with a small interfering RNA abrogated these responses. Furthermore, inhibition of MAPKs and NF-κB pathways were involved in these events. RXR agonist-induced antileukocyte adhesive effects seemed to be mediated via RXRα/peroxisome proliferator-activated receptor (PPAR)γ interaction, since endothelial PPARγ silencing abolished their inhibitory responses. Furthermore, RXR agonists increased RXR/PPARγ interaction, and combinations of suboptimal concentrations of both nuclear receptor ligands inhibited TNF-α-induced mononuclear leukocyte arrest by 60-65%. In vivo, bexarotene dose-dependently inhibited TNF-α-induced leukocyte adhesion to the murine cremasteric arterioles and decreased VCAM-1 and ICAM-1 expression. Therefore, these results reveal that RXR agonists can inhibit the initial inflammatory response that precedes the atherogenic process by targeting different steps of the mononuclear recruitment cascade. Thus, RXR agonists may constitute a new therapeutic tool in the control of the inflammatory process associated with cardiovascular disease.


Asunto(s)
Inhibición de Migración Celular/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , PPAR gamma/metabolismo , Receptor alfa X Retinoide/agonistas , Bexaroteno , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Línea Celular , Inhibición de Migración Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Endotelio Vascular/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/sangre , Leucocitos Mononucleares/efectos de los fármacos , Microcirculación/efectos de los fármacos , Microcirculación/inmunología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inmunología , PPAR gamma/fisiología , Receptor alfa X Retinoide/biosíntesis , Receptor alfa X Retinoide/fisiología , Tetrahidronaftalenos/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Arterias Umbilicales/efectos de los fármacos , Arterias Umbilicales/inmunología , Arterias Umbilicales/patología , Molécula 1 de Adhesión Celular Vascular/sangre
10.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706085

RESUMEN

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Asunto(s)
Inhibición de Migración Celular/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Macrófagos/inmunología , Macrófagos/patología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Inhibición de Migración Celular/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Tioglicolatos/toxicidad
11.
J Immunol ; 189(1): 337-46, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22664869

RESUMEN

CCR4 on T cells is suggested to mediate skin homing in mice. Our objective was to determine the interaction of CCR4, E-selectin ligand (ESL), and α(4)ß(1) on memory and activated T cells in recruitment to dermal inflammation. mAbs to rat CCR4 were developed. CCR4 was on 5-21% of memory CD4 cells, and 20% were also ESL(+). Anti-TCR-activated CD4 and CD8 cells were 40-55% CCR4(+), and ∼75% of both CCR4(+) and CCR4(-) cells were ESL(+). CCR4(+) memory CD4 cells migrated 4- to 7-fold more to dermal inflammation induced by IFN-γ, TNF, TLR agonists, and delayed-type hypersensitivity than CCR4(-) cells. CCR4(+) activated CD4 cells migrated only 5-50% more than CCR4(-) cells to these sites. E-selectin blockade inhibited ∼60% of CCR4(+) activated CD4 cell migration but was less effective on memory cells where α(4)ß(1) was more important. Anti-α(4)ß(1) also inhibited CCR4(-) activated CD4 cells more than CCR4(+) cells. Anti-E-selectin reduced activated CD8 more than CD4 cell migration. These findings modify our understanding of CCR4, ESL, α(4)ß(1), and dermal tropism. There is no strict relationship between CCR4 and ESL for skin homing of CD4 cells, because the activation state and inflammatory stimulus are critical determinants. Dermal homing memory CD4 cells express CCR4 and depend more on α(4)ß(1) than ESL. Activated CD4 cells do not require CCR4, but CCR4(+) cells are more dependent on ESL than on α(4)ß(1), and CCR4(-) cells preferentially use α(4)ß(1). The differentiation from activated to memory CD4 cells increases the dependence on CCR4 for skin homing and decreases the requirement for ESL.


Asunto(s)
Movimiento Celular/inmunología , Selectina E/fisiología , Memoria Inmunológica , Integrina alfa4beta1/fisiología , Activación de Linfocitos/inmunología , Receptores CCR4/fisiología , Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Células CHO , Inhibición de Migración Celular/inmunología , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Selectina E/biosíntesis , Selectina E/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Integrina alfa4beta1/antagonistas & inhibidores , Masculino , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/fisiología , Ratas , Ratas Endogámicas Lew , Receptores CCR4/biosíntesis , Receptores CCR4/deficiencia , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Sialoglicoproteínas/biosíntesis , Piel/patología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
12.
J Immunol ; 187(12): 6402-9, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22095714

RESUMEN

In addition to its antibacterial activity, the cathelicidin-derived LL-37 peptide induces multiple immunomodulatory effects on host cells. Atomic force microscopy, F-actin staining with phalloidin, passage of FITC-conjugated dextran through a monolayer of lung epithelial cells, and assessment of bacterial outgrowth from cells subjected to Pseudomonas aeruginosa infection were used to determine LL-37's effect on epithelial cell mechanical properties, permeability, and bacteria uptake. A concentration-dependent increase in stiffness and F-actin content in the cortical region of A549 cells and primary human lung epithelial cells was observed after treatment with LL-37 (0.5-5 µM), sphingosine 1-phosphate (1 µM), or LPS (1 µg/ml) or infection with PAO1 bacteria. Other cationic peptides, such as RK-31, KR-20, or WLBU2, and the antibacterial cationic steroid CSA-13 did not reproduce the effect of LL-37. A549 cell pretreatment with WRW4, an antagonist of the transmembrane formyl peptide receptor-like 1 protein attenuated LL-37's ability to increase cell stiffness. The LL-37-mediated increase in cell stiffness was accompanied by a decrease in permeability and P. aeruginosa uptake by a confluent monolayer of polarized normal human bronchial epithelial cells. These results suggested that the antibacterial effect of LL-37 involves an LL-37-dependent increase in cell stiffness that prevents epithelial invasion by bacteria.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Catelicidinas/fisiología , Permeabilidad de la Membrana Celular/inmunología , Inhibición de Migración Celular/inmunología , Pulmón/inmunología , Pseudomonas aeruginosa/patogenicidad , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Secuencia de Aminoácidos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Inhibición de Migración Celular/efectos de los fármacos , Células Cultivadas , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Datos de Secuencia Molecular , Pseudomonas aeruginosa/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos
13.
J Immunol ; 187(12): 6327-34, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22102721

RESUMEN

Fascin, as a substrate of protein kinase C (PKC), is a well-known cytoskeletal regulatory protein required for cell migration, invasion, and adhesion in normal and cancer cells. In an effort to identify the role of fascin in PKC-mediated cellular signaling, its expression was suppressed by stable transfection of specific short hairpin RNAs (shRNAs) in mouse monocytic leukemia RAW264.7 cells. Suppression of fascin expression resulted in impaired cellular migration and invasion through extracellular matrix proteins. Unexpectedly, the specific shRNA transfectants exhibited a marked reduction in LPS-induced expression of TNF-α and IL-6 by blocking the translation of their mRNAs. Transient transfection assay using a luciferase expression construct containing the 3' untranslated region of TNF-α or IL-6 mRNA revealed a significant reduction in both LPS- and PMA- (the direct activator of PKC) induced reporter activity in cells transfected with fascin-specific shRNA, indicating that fascin-mediated translational regulation targeted 3' untranslated region. Furthermore, LPS-induced translational activation of reporter expression was blocked by a pharmacological inhibitor of PKC, and the dominant-negative form of PKCα attenuated LPS-induced translational activation. The same type of regulation was also observed in the human monocytic leukemia cell line THP-1 and in mouse peritoneal macrophages. These data demonstrate the involvement of fascin in the PKC-mediated translational regulation of TNF-α and IL-6 expression during the LPS response.


Asunto(s)
Proteínas Portadoras/fisiología , Interleucina-6/genética , Lipopolisacáridos/farmacología , Proteínas de Microfilamentos/fisiología , Biosíntesis de Proteínas/inmunología , Proteína Quinasa C/fisiología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/genética , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/biosíntesis , Línea Celular , Línea Celular Tumoral , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/fisiología , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Ratones Endogámicos ICR , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/biosíntesis , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
14.
Exp Dermatol ; 20(10): 855-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21824198

RESUMEN

Psoriasis is a chronic IL-23/Th17 pathway-associated skin disease. An increased expression of lesional CCL20 can recruit CCR6+ Th17, and lesional cytokine milieu persistently activates keratinocytes to produce CCL20. Lipid-lowering drugs, statins, are known to possess immune-modulating functions. In this study, we explored an inhibitory effect of statins on CCL20/CCR6 interaction. We demonstrated that IL-1ß, TNF-α, and IL-17A significantly increased CCL20 production from HaCaT cells. However, these increments were markedly inhibited by fluvastatin and simvastatin, but not by pravastatin. In the chemotaxis migration assay, pretreatment with fluvastatin and simvastatin inhibited the migration of human CD4+ T cells towards CCL20. However, the level of CCR6 surface expression in memory CD4+ T cells was not affected. Our results suggest that not all, but specific types of statins may be of benefit in alleviating psoriasis partially via interrupting CCL20/CCR6 chemotactic interaction, the mechanism which may eventually lessen the infiltration of Th17 cells.


Asunto(s)
Quimiocina CCL20/metabolismo , Quimiotaxis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Psoriasis/tratamiento farmacológico , Receptores CCR6/metabolismo , Línea Celular , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Quimiotaxis/inmunología , Humanos , Interleucina-17/farmacología , Interleucina-1beta/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Psoriasis/inmunología , Factor de Necrosis Tumoral alfa/farmacología
15.
J Immunol ; 186(7): 4088-97, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21339367

RESUMEN

B cell chemotaxis occurs in response to specific chemokine gradients and is critical for homeostasis and immune response. The molecular regulation of B cell membrane-actin interactions during migration is poorly understood. In this study, we report a role for ezrin, a member of the membrane-cytoskeleton cross-linking ezrin-radixin-moesin proteins, in the regulation of the earliest steps of B cell polarization and chemotaxis. We visualized chemokine-induced changes in murine B cell morphology using scanning electron microscopy and spatiotemporal dynamics of ezrin in B cells using epifluorescence and total internal reflection microscopy. Upon chemokine stimulation, ezrin is transiently dephosphorylated to assume an inactive conformation and localizes to the lamellipodia. B cells expressing a phosphomimetic conformationally active mutant of ezrin or those in which ezrin dephosphorylation was pharmacologically inhibited displayed impaired microvillar dynamics, morphological polarization, and chemotaxis. Our data suggest a 2-fold involvement of ezrin in B cell migration, whereby it first undergoes chemokine-induced dephosphorylation to facilitate membrane flexibility, followed by relocalization to the actin-rich lamellipodia for dynamic forward protrusion of the cells.


Asunto(s)
Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Inhibición de Migración Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Citoesqueleto/química , Citoesqueleto/inmunología , Actinas/metabolismo , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Animales , Subgrupos de Linfocitos B/química , Línea Celular Tumoral , Inhibición de Migración Celular/genética , Quimiotaxis de Leucocito/genética , Proteínas del Citoesqueleto/biosíntesis , Citoesqueleto/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Mutagénesis Sitio-Dirigida , Fosforilación/genética , Fosforilación/inmunología , Conformación Proteica , Seudópodos/genética , Seudópodos/inmunología , Seudópodos/metabolismo , Fase de Descanso del Ciclo Celular/genética , Fase de Descanso del Ciclo Celular/inmunología , Treonina/química , Treonina/genética
16.
J Immunol ; 186(5): 3197-205, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21282514

RESUMEN

CXCL5, a member of the CXC family of chemokines, contributes to neutrophil recruitment during lung inflammation, but its regulation is poorly understood. Because the T cell-derived cytokine IL-17A enhances host defense by triggering production of chemokines, particularly in combination with TNF-α, we hypothesized that IL-17A would enhance TNF-α-induced expression of CXCL5. Intratracheal coadministration of IL-17A and TNF-α in mice induced production of CXCL1, CXCL2, and CXCL5, which was associated with increased neutrophil influx in the lung at 8 and 24 h. The synergistic effects of TNF-α and IL17A were greatly attenuated in Cxcl5(-/-) mice at 24 h, but not 8 h, after exposure, a time when CXCL5 expression was at its peak in wild-type mice. Bone marrow chimeras produced using Cxcl5(-/-) donors and recipients demonstrated that lung-resident cells were the source of CXCL5. Using differentiated alveolar epithelial type II (ATII) cells derived from human fetal lung, we found that IL-17A enhanced TNF-α-induced CXCL5 transcription and stabilized TNF-α-induced CXCL5 transcripts. Whereas expression of CXCL5 required activation of NF-κB, IL-17A did not increase TNF-α-induced NF-κB activation. Apical costimulation of IL-17A and TNF-α provoked apical secretion of CXCL5 by human ATII cells in a transwell system, whereas basolateral costimulation led to both apical and basolateral secretion of CXCL5. The observation that human ATII cells secrete CXCL5 in a polarized fashion may represent a mechanism to recruit neutrophils in host defense in a fashion that discriminates the site of initial injury.


Asunto(s)
Quimiocina CXCL5/biosíntesis , Interleucina-17/fisiología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Células Cultivadas , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Quimiocina CXCL5/deficiencia , Quimiocina CXCL5/metabolismo , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-17/administración & dosificación , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Alveolos Pulmonares/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Immunopharmacol Immunotoxicol ; 33(1): 1-10, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20373855

RESUMEN

Current evidence indicates that periodontal disease is frequently due to inappropriate levels of gingival granulocyte functions. Reason of this failure may be the toxic effects of a number of local or systemic exogenous factors, capable of spreading through the gingival crevice environment, and strongly conditioning the granulocyte activities. The wide list includes bacteria and granulotoxic products, hedonistic drugs (mainly tobacco), and chemotherapeutic agents (especially antimicrobials used for preventing or reducing the accumulation of dental plaque). Almost always, their presence induces a time- and/or dose-dependent toxicity.


Asunto(s)
Inhibición de Migración Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Neutrófilos/inmunología , Enfermedades Periodontales/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Inhibición de Migración Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Neutrófilos/efectos de los fármacos , Enfermedades Periodontales/etiología , Enfermedades Periodontales/microbiología , Preparaciones Farmacéuticas/administración & dosificación , Fumar/efectos adversos , Fumar/inmunología , Factores de Tiempo
18.
J Immunol ; 185(3): 1450-9, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20622113

RESUMEN

Dense accumulations of T cells are often found in peritumoral areas, which reduce the efficiency of contact-dependent lysis of tumor cells. We demonstrate in this study that the extracellular matrix (ECM) produced by tumors can directly regulate T cell migration. The transmigration rate of several T cells including peripheral blood primary T cell, Jurkat, and Molt-4 measured for glioma cells or glioma ECM was consistently low. Jurkat cells showed reduced amoeba-like shape formation and delayed ERK activation when they were in contact with monolayers or ECM of glioma cells as compared with those in contact with HepG2 and MCF-7 cells. Phospho-ERK was located at the leading edge of migrating Jurkat cells. Glioma cells, but not MCF-7 and HepG2 cells, expressed tenascin-C. Knocking down the tenascin-C gene using the short hairpin RNA strategy converted glioma cells to a transmigration-permissive phenotype for Jurkat cells regarding ERK activation, transmigration, and amoeba-like shape formation. In addition, exogenous tenascin-C protein reduced the amoeba-like shape formation and transmigration of Jurkat cells through MCF-7 and HepG2 cell monolayers. A high level of tenascin-C was visualized immunohistochemically in glioma tumor tissues. CD3(+) T cells were detected in the boundary tumor area and stained strongly positive for tenascin-C. In summary, glioma cells can actively paralyze T cell migration by the expression of tenascin-C, representing a novel immune suppressive mechanism achieved through tumor ECM.


Asunto(s)
Inhibición de Migración Celular/inmunología , Polaridad Celular/inmunología , Matriz Extracelular/inmunología , Glioblastoma/inmunología , Tolerancia Inmunológica , Subgrupos de Linfocitos T/inmunología , Tenascina/fisiología , Línea Celular Tumoral , Movimiento Celular/inmunología , Células Cultivadas , Activación Enzimática/genética , Activación Enzimática/inmunología , Matriz Extracelular/enzimología , Matriz Extracelular/patología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Glioblastoma/enzimología , Glioblastoma/patología , Células Hep G2 , Humanos , Tolerancia Inmunológica/genética , Células Jurkat , Microscopía Confocal , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/patología , Tenascina/deficiencia , Tenascina/genética
19.
J Neuroimmunol ; 224(1-2): 45-50, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20537410

RESUMEN

Chemokines are critical mediators of immune cell entry into the central nervous system (CNS), as occurs in neuroinflammatory disease such as multiple sclerosis. Chemokines are also implicated in the immune response to viral infections. Many viruses encode proteins that mimic or block chemokine actions, in order to evade host immune responses. The murine gammaherpesvirus-68 encodes a chemokine-binding protein called M3, which has unique biochemical features that enable it to bind to and inhibit an unusually broad range of chemokines. We applied a replication-defective adenoviral vector encoding M3 (AdM3) directly to the CNS to evaluate the capacity of this protein to inhibit neuroinflammation using the experimental autoimmune encephalomyelitis (EAE) model. Treatment with the AdM3 vector significantly reduced the clinical severity of EAE, attenuated CNS histopathology, and reduced numbers of immune cells infiltrating the CNS. These results suggest that M3 may represent a novel therapeutic approach to neuroinflammatory disease.


Asunto(s)
Quimiocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Rhadinovirus/inmunología , Proteínas Virales/uso terapéutico , Adenoviridae/genética , Animales , Línea Celular , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Quimiocinas/antagonistas & inhibidores , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/virología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica/inmunología , Rhadinovirus/genética , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Proteínas Virales/genética
20.
Biochem Pharmacol ; 80(6): 858-66, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20488163

RESUMEN

Kv1.3 plays a crucial role in the activation and proliferation of T-lymphocytes and macrophages. While Kv1.3 is responsible for the voltage-dependent potassium current in T-cells, in macrophages this K(+) current is generated by the association of Kv1.3 and Kv1.5. Patients with autoimmune diseases show a high number of effector memory T cells that are characterized by a high expression of Kv1.3 and Kv1.3 antagonists ameliorate autoimmune disorders in vivo. Diclofenac is a non-steroidal anti-inflammatory drug (NSAID) used in patients who suffer from painful autoimmune diseases such as rheumatoid arthritis. In this study, we show that diclofenac impairs immune response via a mechanism that involves Kv1.3. While diclofenac inhibited Kv1.3 expression in activated macrophages and T-lymphocytes, Kv1.5 remained unaffected. Diclofenac also decreased iNOS levels in Raw 264.7 cells, impairing their activation in response to lipopolysaccharide (LPS). LPS-induced macrophage migration and IL-2 production in stimulated Jurkat T-cells were also blocked by pharmacological doses of diclofenac. These effects were mimicked by Margatoxin, a specific Kv1.3 inhibitor, and Charybdotoxin, which blocks both Kv1.3 and Ca(2+)-activated K(+) channels (K(Ca)3.1). Because Kv1.3 is a very good target for autoimmune therapies, the effects of diclofenac on Kv1.3 are of high pharmacological relevance.


Asunto(s)
Diclofenaco/farmacología , Sistemas de Liberación de Medicamentos/métodos , Factores Inmunológicos/farmacología , Canal de Potasio Kv1.3/metabolismo , Leucocitos/inmunología , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Células Cultivadas , Humanos , Células Jurkat , Canal de Potasio Kv1.3/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/patología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA