Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Int Immunol ; 28(9): 463-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26989091

RESUMEN

Recruiting pathogenic T cells to the central nervous system (CNS) is a critical step during the development of experimental autoimmune encephalomyelitis (EAE). Here, we report that the absence of autophagy and microtubule-associated protein 1A/1B-light chain 3-associated phagocytosis significantly delayed the onset of EAE in Atg7 conditional knockout (Atg7 CKO) mice in myeloid cells. T-helper cell-cell priming appeared to be normal in the Atg7 CKO mice, but the mice showed significant accumulation of Th17 cells in the lung. The data suggested that the stalling of Th17 cells in the lung en route to the CNS caused the delay. The lung of Atg7 CKO mice, in which we previously demonstrated spontaneous mild inflammation, showed high expression of CCL20, a chemokine that attracts Th17 cells. We have also shown that LPS intranasal instillation delayed EAE onset, suggesting that pulmonary inflammation has an impact on EAE development. Based on our data, therapeutic immunomodulation targeted to the lung, rather than systemically, might be a possible future option to treat multiple sclerosis.


Asunto(s)
Inhibición de Migración Celular/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Neumonía/inmunología , Células Th17/inmunología , Animales , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/inmunología , Inhibición de Migración Celular/genética , Sistema Nervioso Central/patología , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Neumonía/genética , Neumonía/patología
2.
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
3.
Postgrad Med ; 126(5): 167-75, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25295661

RESUMEN

Bacteremia due to Staphylococcus aureus is one of the major causes of morbidity and mortality in India, but studies targeting the source of Staphylococcus aureus bacteremia are lacking. S. aureus has a vivid armamentarium consisting of toxins, adhesins, and other virulence factors by virtue of which it can cause varied types of infections, sometimes of a serious nature. This review highlights the possible causes of S. aureus bacteremia, and discusses the necessity of tracing its source and eliminating it with proper antibiotic therapy to avoid recurrences or relapses.


Asunto(s)
Bacteriemia/fisiopatología , Infecciones Estafilocócicas/fisiopatología , Antibacterianos/uso terapéutico , Bacteriemia/diagnóstico , Bacteriemia/tratamiento farmacológico , Biopelículas/crecimiento & desarrollo , Adhesión Celular/fisiología , Inhibición de Migración Celular/inmunología , Farmacorresistencia Bacteriana , Humanos , India/epidemiología , Factores de Riesgo , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/tratamiento farmacológico
4.
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
5.
Clin Exp Immunol ; 175(3): 359-72, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24032475

RESUMEN

Multiple sclerosis (MS) and chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) represent chronic, autoimmune demyelinating disorders of the central and peripheral nervous system. Although both disorders share some fundamental pathogenic elements, treatments do not provide uniform effects across both disorders. We aim at providing an overview of current and future disease-modifying strategies in these disorders to demonstrate communalities and distinctions. Intravenous immunoglobulins (IVIG) have demonstrated short- and long-term beneficial effects in CIDP but are not effective in MS. Dimethyl fumarate (BG-12), teriflunomide and laquinimod are orally administered immunomodulatory drugs that are already approved or likely to be approved in the near future for the basic therapy of patients with relapsing-remitting MS (RRMS) due to positive results in Phase III clinical trials. However, clinical trials with these drugs in CIDP have not (yet) been initiated. Natalizumab and fingolimod are approved for the treatment of RRMS, and trials to evaluate their safety and efficacy in CIDP are now planned. Alemtuzumab, ocrelizumab and daclizumab respresent monoclonal antibodies in advanced stages of clinical development for their use in RRMS patients. Attempts to study the safety and efficacy of alemtuzumab and B cell-depleting anti-CD20 antibodies, i.e. rituximab, ocrelizumab or ofatumumab, in CIDP patients are currently under way. We provide an overview of the mechanism of action and clinical data available on disease-modifying immunotherapy options for MS and CIDP. Enhanced understanding of the relative effects of therapies in these two disorders may aid rational treatment selection and the development of innovative treatment approaches in the future.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/tratamiento farmacológico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Humanos , Factores Inmunológicos/administración & dosificación , Inmunoterapia , Integrina alfa4/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo
6.
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
7.
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
8.
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
9.
J Immunol ; 190(8): 4305-14, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509351

RESUMEN

Integrins are adhesion molecules critical for the recruitment of leukocytes from blood into peripheral tissues. However, whether integrins are also involved in leukocyte exit from peripheral tissues via afferent lymphatics to the draining lymph node remains poorly understood. In this article, we show that adhesion by the collagen IV-binding integrin α1ß1 unexpectedly inhibited macrophage exit from inflamed skin. We monitored macrophages exiting mouse footpads using a newly developed in situ pulse labeling technique. Blockade of α1ß1 integrin or genetic deletion (Itga1(-/-)) increased macrophage exit efficiency. Chemotaxis assays through collagen IV showed more efficient migration of Itga1(-/-) macrophages relative to wild type. Given that macrophages are key orchestrators of inflammation, α1ß1 integrin adhesion may represent a mechanism for regulating inflammatory responses by controlling macrophage exit or persistence in inflamed tissues.


Asunto(s)
Inhibición de Migración Celular/inmunología , Mediadores de Inflamación/fisiología , Integrina alfa1beta1/fisiología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Adhesión Celular/genética , Adhesión Celular/inmunología , Inhibición de Migración Celular/genética , Pie , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Cadenas alfa de Integrinas/biosíntesis , Cadenas alfa de Integrinas/deficiencia , Cadenas alfa de Integrinas/genética , Integrina alfa1beta1/biosíntesis , Integrina alfa1beta1/deficiencia , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/fisiología
10.
J Immunol ; 190(8): 4360-70, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23475220

RESUMEN

Glucocorticoids (GCs) repress lymphocyte function by controlling gene expression. In this study, we investigated Ag-specific effector T cells and provide evidence that GCs also modulate these cells' cytoskeletal architecture by nongenomic mechanisms. Following GC treatment, effector T cells rapidly lose their polarized morphology, which impedes both their migratory capacity and their interaction with APCs. The cytoskeleton rearrangements are preceded by an activation of ezrin-radixin-moesin proteins, which transiently increases the cellular rigidity but seems to occur independently of altered tyrosine phosphorylation. Phospholipase C activity is critically involved in mediating these nongenomic effects, because its inhibition prevents both T cell depolarization and ezrin-radixin-moesin phosphorylation after GC exposure. GC administration in vivo induced similar morphological changes in effector T cells as observed in vitro, suggesting that the above process plays a role in modulating inflammatory diseases. Taken together, our findings identify a novel mechanism through which GCs rapidly repress T cell function independently of gene transcription.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Inhibición de Migración Celular/inmunología , Proteínas del Citoesqueleto/fisiología , Dexametasona/farmacología , Proteínas de la Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Inhibición de Migración Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Células Cultivadas , Ratas , Ratas Endogámicas Lew , Ratas Transgénicas , Subgrupos de Linfocitos T/ultraestructura
11.
J Immunol ; 190(6): 2659-69, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23378428

RESUMEN

The Tec family tyrosine kinase, Itk, regulates signaling downstream of the TCR. The absence of Itk in CD4(+) T cells results in impaired Th2 responses along with defects in maturation, cytokine production, and survival of iNKT cells. Paradoxically, Itk(-/-) mice have spontaneously elevated serum IgE levels, resulting from an expansion of the Vγ1.1(+)Vδ6.3(+) subset of γδ T cells, known as γδ NKT cells. Comparisons between γδ NKT cells and αß iNKT cells showed convergence in the pattern of cell surface marker expression, cytokine profiles, and gene expression, suggesting that these two subsets of NKT cells undergo similar differentiation programs. Hepatic γδ NKT cells have an invariant TCR and are derived predominantly from fetal progenitors that expand in the thymus during the first weeks of life. The adult thymus contains these invariant γδ NKT cells plus a heterogeneous population of Vγ1.1(+)Vδ6.3(+) T cells with diverse CDR3 sequences. This latter population, normally excluded from the liver, escapes the thymus and homes to the liver when Itk is absent. In addition, Itk(-/-) γδ NKT cells persistently express high levels of Zbtb16 (PLZF) and Il4, genes that are normally downregulated in the most mature subsets of NKT cells. These data indicate that Itk signaling is required to prevent the expansion of γδ NKT cells in the adult thymus, to block their emigration, and to promote terminal NKT cell maturation.


Asunto(s)
Diferenciación Celular/inmunología , Senescencia Celular/inmunología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Proteínas Tirosina Quinasas/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/biosíntesis , Timo/enzimología , Timo/inmunología , Animales , Inhibición de Migración Celular/inmunología , Movimiento Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/citología , Timo/citología
12.
J Immunol ; 190(5): 1961-73, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23365083

RESUMEN

The immune system is characterized by the preferential migration of lymphocytes through specific tissues (i.e., tissue tropism). Tissue tropism is mediated, in part, by the α(4) integrins expressed by T lymphocytes. The α(4)ß(1) integrin mediates migration of memory T lymphocytes into the CNS, whereas the α(4)ß(7) integrin mediates migration preferentially into gastrointestinal tissue. This paradigm was established primarily from investigations in rodents; thus, the objective of this investigation was to determine if blocking the α(4)ß(7) integrin exclusively would affect migration of T lymphocytes into the CNS of primates. The effects of the dual α(4)ß(1) and α(4)ß(7) antagonist natalizumab were compared with those of the α(4)ß(7) antagonist vedolizumab on experimental autoimmune encephalomyelitis in the rhesus monkey. Animals received an initial i.v. bolus of placebo, natalizumab (30 mg/kg), or vedolizumab (30 mg/kg) before intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein and then Ab once weekly thereafter. Natalizumab prevented CNS inflammation and demyelination significantly (p < 0.05), compared with time-matched placebo control animals, whereas vedolizumab did not inhibit these effects, despite saturating the α(4)ß(7) integrin in each animal for the duration of the investigation. These results demonstrate that blocking α(4)ß(7) exclusively does not inhibit immune surveillance of the CNS in primates.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4beta1/antagonistas & inhibidores , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Inyecciones Intravenosas , Integrina alfa4beta1/inmunología , Macaca mulatta , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Natalizumab , Especificidad de Órganos , Placebos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología
13.
Neurobiol Aging ; 34(3): 943-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22819137

RESUMEN

Age-related macular degeneration is an outer retinal disease that involves aging and immune dysfunction. In the aging retina, microglia aggregate in the outer retina and acquire intracellular autofluorescent lipofuscin deposits. In this study, we investigated whether accumulation of A2E, a key bisretinoid constituent of ocular lipofuscin, alters the physiology of retinal microglia in pathologically relevant ways. Our findings show that sublethal accumulations of intracellular A2E in cultured retinal microglia increased microglial activation and decreased microglial neuroprotection of photoreceptors. Increased A2E accumulation also lowered microglial expression of chemokine receptors and suppressed microglial chemotaxis, suggesting that lipofuscin accumulation may potentiate subretinal microglial accumulation. Significantly, A2E accumulation altered microglial complement regulation by increasing complement factor B and decreasing complement factor H expression, favoring increased complement activation and deposition in the outer retina. Taken together, our findings highlight the role of microglia in the local control of complement activation in the retina and present the age-related accumulation of ocular lipofuscin in subretinal microglia as a cellular mechanism capable of driving outer retinal immune dysregulation in age-related macular degeneration pathogenesis.


Asunto(s)
Envejecimiento , Activación de Complemento , Lipofuscina , Degeneración Macular , Microglía , Compuestos de Piridinio/metabolismo , Retina , Retinoides/metabolismo , Anciano , Anciano de 80 o más Años , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Inhibición de Migración Celular/inmunología , Células Cultivadas , Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Humanos , Lipofuscina/inmunología , Lipofuscina/metabolismo , Degeneración Macular/inmunología , Degeneración Macular/metabolismo , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/inmunología , Microglía/metabolismo , Persona de Mediana Edad , Receptores de Quimiocina/genética , Retina/citología , Retina/inmunología , Retina/metabolismo
14.
Immunol Lett ; 149(1-2): 19-29, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23183094

RESUMEN

Eosinophils are major effectors cells implicated in a number of chronic inflammatory diseases in humans, particularly bronchial asthma and allergic rhinitis. The human chemokine receptor C-C receptor 3 (hCCR3) provides a mechanism for the recruitment of eosinophils into tissue and thus has recently become an attractive biological target for therapeutic intervention. In order to develop peptides antagonists of hCCR3-hCCL11 (human eotaxin) interactions, a random bacteriophage hexapeptide library was used to map structural features of hCCR3 by determining the epitopes of neutralizing anti-hCCR3 mAb 7B11. This mAb t is selective for hCCR3 and exhibit potent antagonist activity in receptor binding and functional assays. After three rounds of biopanning, four mAb7B11-binding peptides were identified from a 6-mer linear peptide library. The phage bearing the peptides showed specific binding to immobilized mAb 7B11 with over 94% of phages bound being competitively inhibited by free synthetic peptides. In FACScan analysis all selected phage peptides were able to strongly inhibit the binding of mAb 7B11 to hCCR3-transfected preB-300-19 murine cells. Furthermore, synthetic peptides of the corresponding phage epitopes were effective in blocking the antibody-hCCR3 interactions and to inhibit the binding of hCCL11 to hCCR3 transfectants. Chemically synthesized peptides CKGERF, FERKGK, SSMKVK and RHVSSQ, effectively competed for (125)I-hCCL11 binding to hCCR3 with IC(50) ranging from 3.5 to 9.7µM. Calcium release and chemotaxis of hCCR3 transfectants or human eosinophils were inhibited by all peptides in a dose-dependent manner. Furthermore, they showed inhibitory effects on chemotaxis of human eosinophils induced by hCCL11, hCCL5, hCCL7, hCCL8, and hCCL24. Specificities of all selected peptides were assessed with hCXCR1, hCXCR2, hCXCR3, and hCCR5 receptors. Peptides CKGERF and FERKGK showed inhibitory effects on eosinophil chemotaxis in a murine model of mCCL11-induced peritoneal eosinophilia. The development of peptides inhibiting the interactions between hCCR3 and its chemokine ligands will facilitate the development of small peptides antagonists with the hope of ameliorating chronic inflammatory diseases in humans.


Asunto(s)
Epítopos/inmunología , Biblioteca de Péptidos , Receptores CCR3/inmunología , Animales , Línea Celular , Inhibición de Migración Celular/efectos de los fármacos , Inhibición de Migración Celular/inmunología , Quimiocinas/genética , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Eosinofilia/tratamiento farmacológico , Eosinofilia/genética , Eosinofilia/inmunología , Eosinofilia/patología , Eosinófilos/inmunología , Eosinófilos/patología , Epítopos/genética , Epítopos/farmacología , Humanos , Ratones , Unión Proteica/genética , Unión Proteica/inmunología , Receptores CCR3/genética
15.
J Immunol ; 190(1): 27-35, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23209325

RESUMEN

Epidermal Langerhans cells (LCs) of the skin represent the prototype migratory dendritic cell (DC) subtype. In the skin, they take up Ag, migrate to the draining lymph nodes, and contribute to Ag transport and immunity. Different depletion models for LCs have revealed contrasting roles and contributions of this cell type. To target the migratory properties of DCs, we generated mice lacking the Rho-family GTPase Cdc42 specifically in DCs. In these animals, the initial seeding of the epidermis with LCs is functional, resulting in slightly reduced Langerhans cell numbers. However, Cdc42-deficient LCs fail to leave the skin in steady state as well as upon stimulation, as they do not enter the skin-draining afferent lymph vessels. Similarly, also other Cdc42-deficient migratory DC subsets fail to home properly to the corresponding draining lymph nodes. We used this novel mouse model, in which LCs are locked out, to demonstrate that these cells contribute substantially to priming of Ag-specific CD4 and CD8 T cell responses upon epicutaneous immunization, but could not detect a role in the induction of contact hypersensitivity to various doses of hapten.


Asunto(s)
Inhibición de Migración Celular/inmunología , Movimiento Celular/inmunología , Células de Langerhans/inmunología , Proteína de Unión al GTP cdc42/fisiología , Animales , Inhibición de Migración Celular/genética , Movimiento Celular/genética , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Células Dendríticas/patología , Dermatitis por Contacto/genética , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Modelos Animales de Enfermedad , Epidermis/enzimología , Epidermis/inmunología , Epidermis/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Células de Langerhans/enzimología , Células de Langerhans/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación/genética , Quimera por Radiación/inmunología , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP cdc42/genética
16.
J Immunol ; 190(1): 411-7, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23203922

RESUMEN

Previous studies have suggested that CD47, an essential cell-surface protein, plays an important role in polymorphonuclear neutrophil (PMN) transmigration across tissue cells and extracellular matrix. In the current study, the role of CD47 in PMN transmigration and infiltration into tissues was further evaluated by investigating the function of CD47(-/-) PMN and inflammatory conditions induced in CD47(-/-) mice. Using in vitro time-course assays, we found that CD47(-/-) PMN exhibited no impediment, but slightly enhanced response to and transmigration toward, the chemoattractant fMLF. In vivo analysis in CD47(-/-) mice by inducing acute peritonitis and aggressive colitis observed consistent results, indicating that both PMN and monocytes effectively infiltrated inflammatory sites despite the absence of CD47 on these leukocytes or the surrounding tissue cells. Although PMN transmigration was not delayed in CD47(-/-) mice, fewer PMN were found in the intestine at the postacute/chronic stage of chronic colitis induced with sustained low-dose dextran sulfate sodium. Further analysis suggested that the paucity of PMN accumulation was attributable to attenuated granulopoiesis secondary to assessed lower levels of IL-17. Administration of exogenous IL-17A markedly increased PMN availability and rapidly rendered severe colitis in CD47(-/-) mice under dextran sulfate sodium treatment.


Asunto(s)
Antígeno CD47/genética , Quimiotaxis de Leucocito/inmunología , Colitis/inmunología , Granulocitos/inmunología , Mielopoyesis/inmunología , Neutrófilos/inmunología , Enfermedad Aguda , Animales , Antígeno CD47/fisiología , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Quimiotaxis de Leucocito/genética , Enfermedad Crónica , Colitis/genética , Colitis/patología , Femenino , Granulocitos/metabolismo , Granulocitos/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Mielopoyesis/genética , Neutrófilos/citología , Neutrófilos/patología
17.
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
18.
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
19.
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
20.
J Immunol ; 189(9): 4592-601, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23028056

RESUMEN

VCAM-1 plays a key role in leukocyte trafficking during inflammatory responses. However, molecular mechanisms underlying this function have not been clearly elucidated. In this study, using phage display technology, we developed a rabbit/human chimeric VCAM-1 Ab, termed VCAM-1 domain 6 (VCAM-1-D6), which specifically recognizes aa 511-599 within the sixth Ig-like domain. We report that the VCAM-1-D6 Ab blocked U937 cell transmigration across activated HUVECs but did not alter adhesion of U937 cells to the HUVECs. We also demonstrate that VCAM-1-D6 does not alter TNF-α-stimulated endothelial cell chemokine or cytokine production. Furthermore, through in vivo efficacy testing using a mouse islet allograft model, we demonstrate that VCAM-1-D6 significantly alleviates allograft rejection by blocking leukocyte infiltration to the grafted islets. Taken together, our results suggest that the VCAM-1-D6 Ab may block VCAM-1-mediated inflammation and could be a useful tool in treating inflammatory diseases.


Asunto(s)
Anticuerpos Bloqueadores/fisiología , Adhesión Celular/inmunología , Inhibición de Migración Celular/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Inmunoglobulina G/fisiología , Leucocitos/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Anticuerpos Bloqueadores/genética , Adhesión Celular/genética , Inhibición de Migración Celular/genética , Endotelio Vascular/química , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/fisiología , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/genética , Leucocitos/citología , Ratones , Estructura Terciaria de Proteína/genética , Conejos , Células U937 , Molécula 1 de Adhesión Celular Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA