Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
BMC Cancer ; 15: 391, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25958384

RESUMEN

BACKGROUND: NBPF1 (Neuroblastoma Breakpoint Family, member 1) was originally identified in a neuroblastoma patient on the basis of its disruption by a chromosomal translocation t(1;17)(p36.2;q11.2). Considering this genetic defect and the frequent genomic alterations of the NBPF1 locus in several cancer types, we hypothesized that NBPF1 is a tumor suppressor. Decreased expression of NBPF1 in neuroblastoma cell lines with loss of 1p36 heterozygosity and the marked decrease of anchorage-independent clonal growth of DLD1 colorectal carcinoma cells with induced NBPF1 expression further suggest that NBPF1 functions as tumor suppressor. However, little is known about the mechanisms involved. METHODS: Expression of NBPF was analyzed in human skin and human cervix by immunohistochemistry. The effects of NBPF1 on the cell cycle were evaluated by flow cytometry. We investigated by real-time quantitative RT-PCR the expression profile of a panel of genes important in cell cycle regulation. Protein levels of CDKN1A-encoded p21(CIP1/WAF1) were determined by western blotting and the importance of p53 was shown by immunofluorescence and by a loss-of-function approach. LC-MS/MS analysis was used to investigate the proteome of DLD1 colon cancer cells with induced NBPF1 expression. Possible biological interactions between the differentially regulated proteins were investigated with the Ingenuity Pathway Analysis tool. RESULTS: We show that NBPF is expressed in the non-proliferative suprabasal layers of squamous stratified epithelia of human skin and cervix. Forced expression of NBPF1 in HEK293T cells resulted in a G1 cell cycle arrest that was accompanied by upregulation of the cyclin-dependent kinase inhibitor p21(CIP1/WAF1) in a p53-dependent manner. Additionally, forced expression of NBPF1 in two p53-mutant neuroblastoma cell lines also resulted in a G1 cell cycle arrest and CDKN1A upregulation. However, CDKN1A upregulation by NBPF1 was not observed in the DLD1 cells, which demonstrates that NBPF1 exerts cell-specific effects. In addition, proteome analysis of NBPF1-overexpressing DLD1 cells identified 32 differentially expressed proteins, of which several are implicated in carcinogenesis. CONCLUSIONS: We demonstrated that NBPF1 exerts different tumor suppressive effects, depending on the cell line analyzed, and provide new clues into the molecular mechanism of the enigmatic NBPF proteins.


Asunto(s)
Proteínas Portadoras/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Neuroblastoma/genética , Proteínas Supresoras de Tumor/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Epitelio/metabolismo , Epitelio/patología , Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Familia de Multigenes , Neuroblastoma/metabolismo , Proteoma , Proteómica , Transducción de Señal , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
2.
Am J Respir Crit Care Med ; 189(3): 282-91, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24456467

RESUMEN

RATIONALE: Sepsis is one of the leading causes of death around the world. The failure of clinical trials to treat sepsis demonstrates that the molecular mechanisms are multiple and are still insufficiently understood. OBJECTIVES: To clarify the long disputed hierarchical contribution of several central inflammatory mediators (IL-1ß, IL-18, caspase [CASP] 7, CASP1, and CASP11) in septic shock and to explore their therapeutic potential. METHODS: LPS- and tumor necrosis factor (TNF)-induced lethal shock, and cecal ligation and puncture (CLP) were performed in genetically or pharmacologically targeted mice. Body temperature and survival were monitored closely, and plasma was analyzed for several markers of cellular disintegration and inflammation. MEASUREMENTS AND MAIN RESULTS: Interestingly, deficiency of both IL-1ß and IL-18 additively prevented LPS-induced mortality. The detrimental role of IL-1ß and IL-18 was confirmed in mice subjected to a lethal dose of TNF, or to a lethal CLP procedure. Although their upstream activator, CASP1, and its amplifier, CASP11, are considered potential therapeutic targets because of their crucial involvement in endotoxin-induced toxicity, CASP11- or CASP1/11-deficient mice were not, or hardly, protected against a lethal TNF or CLP challenge. In line with our results obtained in genetically deficient mice, only the combined neutralization of IL-1 and IL-18, using the IL-1 receptor antagonist anakinra and anti-IL-18 antibodies, conferred complete protection against endotoxin-induced lethality. CONCLUSIONS: Our data point toward the therapeutic potential of neutralizing IL-1 and IL-18 simultaneously in sepsis, rather than inhibiting the upstream inflammatory caspases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Autoanticuerpos/uso terapéutico , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-18/deficiencia , Interleucina-1beta/deficiencia , Choque Séptico/prevención & control , Animales , Biomarcadores/sangre , Caspasa 1/sangre , Caspasa 1/deficiencia , Caspasa 7/sangre , Caspasa 7/deficiencia , Caspasas/sangre , Caspasas/deficiencia , Caspasas Iniciadoras , Ciego/cirugía , Quimioterapia Combinada , Interleucina-18/antagonistas & inhibidores , Interleucina-18/sangre , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/sangre , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Choque Séptico/sangre , Choque Séptico/etiología , Factor de Necrosis Tumoral alfa
3.
J Virol ; 87(6): 3314-23, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23302879

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in infants worldwide. Despite decades of research, there is still no registered vaccine available for this major pathogen. We investigated the protective efficacy of a recombinant influenza virus, PR8/NA-F(85-93), that carries the RSV CD8(+) T cell epitope F(85-93) in its neuraminidase stalk. F(85-93)-specific cytotoxic T lymphocytes (CTLs) were induced in mice after a single intranasal immunization with PR8/NA-F(85-93) virus, and these CTLs provided a significant reduction in the lung viral load upon a subsequent challenge with RSV. To avoid influenza-induced morbidity, we treated mice with matrix protein 2 (M2e)-specific monoclonal antibodies before PR8/NA-F(85-93) virus infection. Treatment with anti-M2e antibodies reduced the infiltration of immune cells in the lungs upon PR8/NA-F(85-93) infection, whereas the formation of inducible bronchus-associated lymphoid tissue was not affected. Moreover, this treatment prevented body weight loss yet still permitted the induction of RSV F-specific T cell responses and significantly reduced RSV replication upon challenge. These results demonstrate that it is possible to take advantage of the infection-permissive protection of M2e-specific antibodies against influenza A virus to induce heterologous CD8(+) T cell-mediated immunity by an influenza A virus vector expressing the RSV F(85-93) epitope.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Virus de la Influenza A/genética , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/inmunología , Replicación Viral , Animales , Peso Corporal , Epítopos de Linfocito T/genética , Femenino , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Recombinación Genética , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Proteínas Virales de Fusión/genética , Proteínas Virales/genética
4.
PLoS Pathog ; 8(3): e1002570, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22396652

RESUMEN

The innate immune response provides the first line of defense against viruses and other pathogens by responding to specific microbial molecules. Influenza A virus (IAV) produces double-stranded RNA as an intermediate during the replication life cycle, which activates the intracellular pathogen recognition receptor RIG-I and induces the production of proinflammatory cytokines and antiviral interferon. Understanding the mechanisms that regulate innate immune responses to IAV and other viruses is of key importance to develop novel therapeutic strategies. Here we used myeloid cell specific A20 knockout mice to examine the role of the ubiquitin-editing protein A20 in the response of myeloid cells to IAV infection. A20 deficient macrophages were hyperresponsive to double stranded RNA and IAV infection, as illustrated by enhanced NF-κB and IRF3 activation, concomitant with increased production of proinflammatory cytokines, chemokines and type I interferon. In vivo this was associated with an increased number of alveolar macrophages and neutrophils in the lungs of IAV infected mice. Surprisingly, myeloid cell specific A20 knockout mice are protected against lethal IAV infection. These results challenge the general belief that an excessive host proinflammatory response is associated with IAV-induced lethality, and suggest that under certain conditions inhibition of A20 might be of interest in the management of IAV infections.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Virus de la Influenza A/patogenicidad , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Macrófagos Alveolares/enzimología , Neutrófilos/enzimología , Ubiquitina-Proteína Ligasas/deficiencia , Animales , Cisteína Endopeptidasas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica , Virus de la Influenza A/fisiología , Factor 3 Regulador del Interferón/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/enzimología , Pulmón/inmunología , Pulmón/virología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/virología , Ratones , Ratones Noqueados , FN-kappa B/biosíntesis , Neutrófilos/inmunología , Neutrófilos/virología , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Mensajero/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
5.
Mol Ther ; 21(1): 251-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23011030

RESUMEN

The use of DNA and viral vector-based vaccines for the induction of cellular immune responses is increasingly gaining interest. However, concerns have been raised regarding the safety of these immunization strategies. Due to the lack of their genome integration, mRNA-based vaccines have emerged as a promising alternative. In this study, we evaluated the potency of antigen-encoding mRNA complexed with the cationic lipid 1,2-dioleoyl-3trimethylammonium-propane/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOTAP/DOPE ) as a novel vaccination approach. We demonstrate that subcutaneous immunization of mice with mRNA encoding the HIV-1 antigen Gag complexed with DOTAP/DOPE elicits antigen-specific, functional T cell responses resulting in specific killing of Gag peptide-pulsed cells and the induction of humoral responses. In addition, we show that DOTAP/DOPE complexed antigen-encoding mRNA displays immune-activating properties characterized by secretion of type I interferon (IFN) and the recruitment of proinflammatory monocytes to the draining lymph nodes. Finally, we demonstrate that type I IFN inhibit the expression of DOTAP/DOPE complexed antigen-encoding mRNA and the subsequent induction of antigen-specific immune responses. These results are of high relevance as they will stimulate the design and development of improved mRNA-based vaccination approaches.


Asunto(s)
Antígenos/inmunología , Inmunidad Celular/efectos de los fármacos , Interferón Tipo I/farmacología , ARN Mensajero/inmunología , Animales , Ratones , ARN Mensajero/administración & dosificación
6.
Am J Pathol ; 181(1): 174-84, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22613023

RESUMEN

Resident alveolar macrophages (rAMs) residing in the bronchoalveolar lumen of the airways play an important role in limiting excessive inflammatory responses in the respiratory tract. High phagocytic activity along with hyporesponsiveness to inflammatory insults and lack of autonomous IFN-ß production are crucial assets in this regulatory function. Using a mouse model of asthma, we analyzed the fate of rAMs both during and after allergic bronchial inflammation. Although nearly indistinguishable phenotypically from naïve rAMs, postinflammation rAMs exhibited a strongly reduced basal phagocytic capacity, accompanied by a markedly increased inflammatory reactivity to Toll-like receptors TLR-3 (poly I:C), TLR-4 [lipopolysaccharide (LPS)], and TLR-7 (imiquimod). Importantly, after inflammation, rAMs exhibited a switch from an IFN-ß-defective to an IFN-ß-competent phenotype, thus indicating the occurrence of a new, inflammatory-released rAM population in the postallergic lung. Analysis of rAM turnover revealed a rapid disappearance of naïve rAMs after the onset of inflammation. This inflammation-induced rAM turnover is critical for the development of the hyperinflammatory rAM phenotype observed after clearance of bronchial inflammation. These data document a novel mechanism of innate imprinting in which noninfectious bronchial inflammation causes alveolar macrophages to acquire a highly modified innate reactivity. The resulting increase in secretion of inflammatory mediators on TLR stimulation implies a role for this phenomenon of innate imprinting in the increased sensitivity of postallergic lungs to inflammatory insults.


Asunto(s)
Asma/inmunología , Bronquitis/inmunología , Macrófagos Alveolares/inmunología , Animales , Asma/fisiopatología , Hiperreactividad Bronquial/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/biosíntesis , Progresión de la Enfermedad , Femenino , Mediadores de Inflamación/inmunología , Interferón beta/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Fagocitosis/inmunología , Fenotipo , Receptores Toll-Like/inmunología
7.
Blood ; 117(21): 5620-30, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21355089

RESUMEN

Zeb2 (Sip1/Zfhx1b) is a member of the zinc-finger E-box-binding (ZEB) family of transcriptional repressors previously demonstrated to regulate epithelial-to-mesenchymal transition (EMT) processes during embryogenesis and tumor progression. We found high Zeb2 mRNA expression levels in HSCs and hematopoietic progenitor cells (HPCs), and examined Zeb2 function in hematopoiesis through a conditional deletion approach using the Tie2-Cre and Vav-iCre recombination mouse lines. Detailed cellular analysis demonstrated that Zeb2 is dispensable for hematopoietic cluster and HSC formation in the aorta-gonadomesonephros region of the embryo, but is essential for normal HSC/HPC differentiation. In addition, Zeb2-deficient HSCs/HPCs fail to properly colonize the fetal liver and/or bone marrow and show enhanced adhesive properties associated with increased ß1 integrin and Cxcr4 expression. Moreover, deletion of Zeb2 resulted in embryonic (Tie2-Cre) and perinatal (Vav-icre) lethality due to severe cephalic hemorrhaging and decreased levels of angiopoietin-1 and, subsequently, improper pericyte coverage of the cephalic vasculature. These results reveal essential roles for Zeb2 in embryonic hematopoiesis and are suggestive of a role for Zeb2 in hematopoietic-related pathologies in the adult.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Transición Epitelial-Mesenquimal , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/fisiología , Proteínas Represoras/fisiología , Animales , Cadherinas/metabolismo , Movimiento Celular , Femenino , Citometría de Flujo , Genes Letales , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Dedos de Zinc
8.
J Immunol ; 184(1): 203-11, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949090

RESUMEN

Because of their large surface area, the lungs appear an attractive route for noninvasive vaccine delivery, harboring the potential to induce local mucosal immune responses in addition to systemic immunity. To evoke adaptive immunity, Ags require the addition of adjuvants that not only enhance the strength of the immune response but also determine the type of response elicited. In this study, we evaluate the adjuvant characteristics of polyelectrolyte microcapsules (PEMs) consisting of the biopolymers dextran-sulfate and poly-L-arginine. PEMs form an entirely new class of microcapsules that are generated by the sequential adsorption of oppositely charged polymers (polyelectrolytes) onto a sacrificial colloidal template, which is subsequently dissolved leaving a hollow microcapsule surrounded by a thin shell. Following intratracheal instillation, PEMs were not only efficiently taken up by APCs but also enhanced their activation status. Pulmonary adaptive immune responses were characterized by the induction of a strongly Th17-polarized response. When compared with a mixture of soluble Ag with empty microcapsules, Ag encapsulation significantly enhanced the strength of this local mucosal response. Given their unique property to selectively generate Th17-polarized immune responses, PEMs may become of significant interest in the development of effective vaccines against fungal and bacterial species.


Asunto(s)
Implantes Absorbibles , Adyuvantes Inmunológicos/administración & dosificación , Cápsulas/administración & dosificación , Interleucina-17/inmunología , Pulmón/inmunología , Vacunación/métodos , Adyuvantes Inmunológicos/síntesis química , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos/administración & dosificación , Antígenos/inmunología , Lavado Broncoalveolar , Cápsulas/síntesis química , Sulfato de Dextran/inmunología , Electrólitos/inmunología , Femenino , Citometría de Flujo , Inmunidad Mucosa/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Péptidos/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Infect Dis ; 204(11): 1692-701, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21998474

RESUMEN

Despite the medical importance of respiratory syncytial virus (RSV) infections, there is no vaccine or therapeutic agent available. Prophylactic administration of palivizumab, a humanized monoclonal RSV fusion (F) protein-specific antibody, can protect high-risk children. Previously, we have demonstrated that RSV can be neutralized by picomolar concentrations of a camelid immunoglobulin single-variable domain that binds the RSV protein F (F-VHHb nanobodies). Here, we investigated the mechanism by which these nanobodies neutralize RSV and tested their antiviral activity in vivo. We demonstrate that bivalent RSV F-specific nanobodies neutralize RSV infection by inhibiting fusion without affecting viral attachment. The ability of RSV F-specific nanobodies to protect against RSV infection was investigated in vivo. Intranasal administration of bivalent RSV F-specific nanobodies protected BALB/c mice from RSV infection, and associated pulmonary inflammation. Moreover, therapeutic treatment with these nanobodies after RSV infection could reduce viral replication and reduced pulmonary inflammation. Thus, nanobodies are promising therapeutic molecules for treatment of RSV.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Antivirales/inmunología , Antivirales/farmacología , Femenino , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Factores de Tiempo , Proteínas Virales de Fusión/inmunología , Carga Viral/efectos de los fármacos
10.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L679-90, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21335522

RESUMEN

Contrary to the T-helper (Th)-2 bias and eosinophil-dominated bronchial inflammation encountered in most asthmatic subjects, other patients may exhibit neutrophil-predominant asthma subphenotypes, along with Th-1 and Th-17 cells. However, the etiology of many neutrophil-dominated asthma subphenotypes remains ill-understood, in part due to a lack of appropriate experimental models. To better understand the distinct immune-pathological features of eosinophilic vs. neutrophilic asthma types, we developed an ovalbumin (OVA)-based mouse model of neutrophil-dominated allergic pulmonary inflammation. Consequently, we probed for particular inflammatory signatures and checkpoints underlying the immune pathology in this new model, as well as in a conventional, eosinophil-dominated asthma model. Briefly, mice were OVA sensitized using either aluminum hydroxide (alum) or complete Freund's adjuvants, followed by OVA aerosol challenge. T-cell, granulocyte, and inflammatory mediator profiles were determined, along with alveolar macrophage genomewide transcriptome profiling. In contrast to the Th-2-dominated phenotype provoked by alum, OVA/ complete Freund's adjuvants adjuvant-based sensitization, followed by allergen challenge, elicited a pulmonary inflammation that was poorly controlled by dexamethasone, and in which Th-1 and Th-17 cells additionally participated. Analysis of the overall pulmonary and alveolar macrophage inflammatory mediator profiles revealed remarkable similarities between both models. Nevertheless, we observed pronounced differences in the IL-12/IFN-γ axis and its control by IL-18 and IL-18 binding protein, but also in macrophage arachidonic acid metabolism and expression of T-cell instructive ligands. These differential signatures, superimposed onto a generic inflammatory signature, denote distinctive inflammatory checkpoints potentially involved in orchestrating neutrophil-dominated asthma.


Asunto(s)
Eosinófilos/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Animales , Asma/inmunología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund/inmunología , Perfilación de la Expresión Génica , Mediadores de Inflamación/análisis , Interleucina-12/inmunología , Interleucina-18/inmunología , Pulmón/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología
11.
Blood ; 114(21): 4664-74, 2009 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19726720

RESUMEN

Alternatively activated macrophages (AAMs), triggered by interleukin-4 (IL-4) and IL-13, play a modulating role during Th2 cytokine-driven pathologies, but their molecular armament remains poorly characterized. Here, we established E-cadherin (Cdh1) as a selective marker for IL-4/IL-13-exposed mouse and human macrophages, which is STAT6-dependently induced during polarized Th2 responses associated with Taenia crassiceps helminth infections or allergic airway inflammation. The IL-4-dependent, arginase-1/ornithine decarboxylase-mediated production of polyamines is important for maximal Cdh1 induction, unveiling a novel mechanism for IL-4-dependent gene transcription. At the macrophage surface, E-cadherin forms a functional complex with the catenins that accumulates at sites of cell contact. Macrophage-specific deletion of the Cdh1 gene illustrates the implication of E-cadherin in IL-4-driven macrophage fusion and heterotypic interactions with CD103(+) and KLRG1(+) T cells. This study identifies the E-cadherin/catenin complex as a discriminative, partly polyamine-regulated feature of IL-4/IL-13-exposed alternatively activated macrophages that contributes to homotypic and heterotypic cellular interactions.


Asunto(s)
Cadherinas/inmunología , Interleucina-4/inmunología , Macrófagos/inmunología , Poliaminas/inmunología , Transducción de Señal/inmunología , alfa Catenina/inmunología , Animales , Asma/inmunología , Western Blotting , Cadherinas/metabolismo , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Hipersensibilidad/inmunología , Inmunoprecipitación , Interleucina-13/inmunología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Microscopía Fluorescente , Teniasis/inmunología , alfa Catenina/metabolismo
12.
Infect Immun ; 78(9): 3848-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20643856

RESUMEN

Infection with Salmonella enterica serovar Typhimurium is a complex disease in which the host-bacterium interactions are strongly influenced by genetic factors of the host. We demonstrate that SPRET/Ei, an inbred mouse strain derived from Mus spretus, is resistant to S. Typhimurium infections. The kinetics of bacterial proliferation, as well as histological examinations of tissue sections, suggest that SPRET/Ei mice can control bacterial multiplication and spreading despite significant attenuation of the cytokine response. The resistance of SPRET/Ei mice to S. Typhimurium infection is associated with increased leukocyte counts in the circulation and enhanced neutrophil influx into the peritoneum during the course of infection. A critical role of neutrophils was confirmed by neutrophil depletion: neutropenic SPRET/Ei mice were sensitive to infection with S. Typhimurium and showed much higher bacterial loads. To identify genes that modulate the natural resistance of SPRET/Ei mice to S. Typhimurium infection, we performed a genome-wide study using an interspecific backcross between C3H/HeN and SPRET/Ei mice. The results of this analysis demonstrate that at least two loci, located on chromosomes 6 and 11, affect survival following lethal infection with S. Typhimurium. These two loci contain several interesting candidate genes which may have important implications for the search for genetic factors controlling Salmonella infections in humans and for our understanding of complex host-pathogen interactions in general.


Asunto(s)
Neutrófilos/fisiología , Salmonelosis Animal/genética , Salmonelosis Animal/inmunología , Salmonella typhimurium , Animales , Caspasa 2/fisiología , Movimiento Celular , Citocinas/biosíntesis , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Sistema Mononuclear Fagocítico/microbiología , Sitios de Carácter Cuantitativo
13.
Am J Pathol ; 174(1): 3-13, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19074616

RESUMEN

Asthma is a type-I allergic airway disease characterized by Th(2) cells and IgE. Episodes of bronchial inflammation, eosinophilic in nature and promoting bronchoconstriction, may become chronic and lead to persistent respiratory symptoms and irreversible structural airway changes. Representative mostly of mild to moderate asthma, this clinical definition fails to account for the atypical and often more severe phenotype found in a considerable proportion of asthmatics who have increased neutrophil cell counts in the airways as a distinguishing trait. Neutrophilic inflammation is a hallmark of another type of allergic airway pathology, hypersensitivity pneumonitis. Considered as an immune counterpart of asthma, hypersensitivity pneumonitis is a prototypical type-III allergic inflammatory reaction involving the alveoli and lung interstitium, steered by Th(1) cells and IgG and, in its chronic form, accompanied by fibrosis. Although pathologically very different and commonly approached as separate disorders, as discussed in this review, clinical studies as well as data from animal models reveal undeniable parallels between both airway diseases. Danger signaling elicited by the allergenic agent or by accompanying microbial patterns emerges as critical in enabling immune sensitization and in determining the type of sensitization and ensuing allergic disease. On this basis, we propose that asthma allergens cause severe noneosinophilic asthma because of sensitization in the presence of hypersensitivity pneumonitis-promoting danger signaling.


Asunto(s)
Alveolitis Alérgica Extrínseca/inmunología , Asma/inmunología , Animales , Eosinófilos/inmunología , Humanos , Inflamación/inmunología
15.
J Leukoc Biol ; 77(3): 321-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15591125

RESUMEN

Molecular markers, especially surface markers associated with type II, cytokine-dependent, alternatively activated macrophages (aaMF), remain scarce. Besides the earlier documented markers, macrophage mannose receptor and arginase 1, we demonstrated recently that murine aaMF are characterized by increased expression of found in inflammatory zone 1 (FIZZ1) and the secretory lectin Ym. We now document that expression of the two members of the mouse macrophage galactose-type C-type lectin gene family (mMGL1 and mMGL2) is induced in diverse populations of aaMF, including peritoneal macrophages elicited during infection with the protozoan Trypanosoma brucei brucei or the Helminth Taenia crassiceps and alveolar macrophages elicited in a mouse model of allergic asthma. In addition, we demonstrate that in vitro, interleukin-4 (IL-4) and IL-13 up-regulate mMGL1 and mMGL2 expression and that in vivo, induction of mMGL1 and mMGL2 is dependent on IL-4 receptor signaling. Moreover, we show that expression of MGL on human monocytes is also up-regulated by IL-4. Hence, macrophage galactose-type C-type lectins represent novel surface markers for murine and human aaMF.


Asunto(s)
Asma/inmunología , Hiperreactividad Bronquial/inmunología , Lectinas Tipo C/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Enfermedades Parasitarias/inmunología , Animales , Asialoglicoproteínas , Asma/patología , Biomarcadores/metabolismo , Hiperreactividad Bronquial/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/inmunología , Inflamación/patología , Lectinas Tipo C/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Parasitarias/parasitología , ARN Mensajero/genética , Taenia/inmunología , Taenia/parasitología , Trypanosoma brucei brucei/inmunología
16.
Nat Commun ; 6: 5794, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25565005

RESUMEN

Early T-cell precursor leukaemia (ETP-ALL) is a high-risk subtype of human leukaemia that is poorly understood at the molecular level. Here we report translocations targeting the zinc finger E-box-binding transcription factor ZEB2 as a recurrent genetic lesion in immature/ETP-ALL. Using a conditional gain-of-function mouse model, we demonstrate that sustained Zeb2 expression initiates T-cell leukaemia. Moreover, Zeb2-driven mouse leukaemia exhibit some features of the human immature/ETP-ALL gene expression signature, as well as an enhanced leukaemia-initiation potential and activated Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signalling through transcriptional activation of IL7R. This study reveals ZEB2 as an oncogene in the biology of immature/ETP-ALL and paves the way towards pre-clinical studies of novel compounds for the treatment of this aggressive subtype of human T-ALL using our Zeb2-driven mouse model.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Leucemia de Células T/fisiopatología , Proteínas Represoras/genética , Transducción de Señal/fisiología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Técnicas Histológicas , Proteínas de Homeodominio/inmunología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Quinasas Janus/metabolismo , Estimación de Kaplan-Meier , Cariotipificación , Luciferasas , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-7/metabolismo , Proteínas Represoras/inmunología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
17.
PLoS One ; 7(7): e41547, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844497

RESUMEN

The type III secretion system (T3SS) is a complex nanomachine of many pathogenic gram-negative bacteria. It forms a proteinaceous channel that is inserted into the host eukaryotic cell membrane for injection of bacterial proteins that manipulate host cell signaling. However, few studies have focused on the effector-independent functions of the T3SS. Using a murine model of acute lung infection with Pseudomonas aeruginosa, an important human opportunistic pathogen, we compared the pathogenicity of mutant bacteria that lack all of the known effector toxins ( ΔSTY), with mutant bacteria that also lack the major translocator protein PopB (ΔSTY/ΔPopB) and so cannot form a functional T3SS channel in the host cell membrane. Mortality was higher among mice challenged with ΔSTY compared to mice challenged with ΔSTY/ΔPopB mutant bacteria. In addition, mice infected with ΔSTY showed decreased bacterial clearance from the lungs compared to those infected with ΔSTY/ΔPopB. Infection was in both cases associated with substantial killing of lung infiltrating macrophages. However, macrophages from ΔSTY-infected mice died by pro-inflammatory necrosis characterized by membrane permeabilization and caspase-1 mediated IL-1ß production, whereas macrophages from ΔSTY/ΔPopB infected mice died by apoptosis, which is characterized by annexin V positive staining of the cell membrane and caspase-3 activation. This was confirmed in macrophages infected in vitro. These results demonstrate a T3SS effector toxin independent role for the T3SS, in particular the T3SS translocator protein PopB, in the pathogenicity of P. aeruginosa during acute lung infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Enfermedades Pulmonares/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Enfermedad Aguda , Animales , Antígenos Bacterianos/metabolismo , Muerte Celular , Línea Celular , Exotoxinas/metabolismo , Femenino , Interleucina-1beta/metabolismo , Pulmón/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Mutación , Porosidad , Transporte de Proteínas , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/genética
18.
ACS Nano ; 6(3): 2136-49, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22303914

RESUMEN

Recombinant antigens hold high potential to develop vaccines against lethal intracellular pathogens and cancer. However, they are poorly immunogenic and fail to induce potent cellular immunity. In this paper, we demonstrate that polymeric multilayer capsules (PMLC) strongly increase antigen delivery toward professional antigen-presenting cells in vivo, including dendritic cells (DCs), macrophages, and B cells, thereby enforcing antigen presentation and stimulating T cell proliferation. A thorough analysis of the T cell response demonstrated their capacity to induce IFN-γ secreting CD4 and CD8 T cells, in addition to follicular T-helper cells, a recently identified CD4 T cell subset supporting antibody responses. On the B cell level, PMLC-mediated antigen delivery promoted the formation of germinal centers, resulting in increased numbers of antibody-secreting plasma cells and elevated antibody titers. The functional relevance of the induced immune responses was validated in murine models of influenza and melanoma. On a mechanistic level, we have demonstrated the capacity of PMLC to activate the NALP3 inflammasome and trigger the release of the potent pro-inflammatory cytokine IL-1ß. Finally, using DC-depleted mice, we have identified DCs as the key mediators of the immunogenic properties of PMLC.


Asunto(s)
Portadores de Fármacos/química , Gripe Humana/inmunología , Gripe Humana/prevención & control , Melanoma Experimental/inmunología , Melanoma Experimental/prevención & control , Polímeros/química , Vacunación/métodos , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Materiales Biocompatibles/química , Cápsulas , Sulfato de Dextran/química , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/inmunología , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inflamasomas/metabolismo , Ratones , Péptidos/química
19.
ACS Nano ; 5(9): 6886-93, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21866940

RESUMEN

Here we present a single-step all-aqueous approach to encapsulate biomolecules such as enzymes and proteins into stable microreactors. Key in this method is the use of spray-drying of the biomolecules of interest in combination with oppositely charged polyelectrolytes and mannitol as the sacrificial template. Remarkably, upon spray-drying in the presence of polyelectrolyte, mannitol crystallization is suppressed and the obtained amorphous mannitol offers enhanced preservation of the biomolecules' activity. Moreover, the use of mannitol allows the formation of nanopores within the microparticles upon rehydration of the microparticles in aqueous medium and subsequent dissolution of the mannitol. The oppositely charged polyelectrolytes provide a polymeric framework which stabilizes the microparticles upon rehydration. The versatility of this approach is demonstrated using horseradish peroxidase as the model enzyme and ovalbumin as the model antigen.


Asunto(s)
Reactores Biológicos , Cristalización , Electrólitos/química , Manitol/química
20.
EMBO Mol Med ; 3(4): 222-34, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21328541

RESUMEN

Mycobacterium bovis bacille Calmette-Guerin (BCG) provides only limited protection against pulmonary tuberculosis. We tested the hypothesis that BCG might have retained immunomodulatory properties from its pathogenic parent that limit its protective immunogenicity. Mutation of the molecules involved in immunomodulation might then improve its vaccine potential. We studied the vaccine potential of BCG mutants deficient in the secreted acid phosphatase, SapM, or in the capping of the immunomodulatory ManLAM cell wall component with α-1,2-oligomannoside. Both systemic and intratracheal challenge of mice with Mycobacterium tuberculosis following vaccination showed that the SapM mutant, compared to the parental BCG vaccine, provided better protection: it led to longer-term survival. Persistence of the SapM-mutated BCG in vivo resembled that of the parental BCG indicating that this mutation will likely not compromise the safety of the BCG vaccine. The SapM mutant BCG vaccine was more effective than the parental vaccine in inducing recruitment and activation of CD11c(+) MHC-II(int) CD40(int) dendritic cells (DCs) to the draining lymph nodes. Thus, SapM acts by inhibiting recruitment of DCs and their activation at the site of vaccination.


Asunto(s)
Fosfatasa Ácida/genética , Vacuna BCG/inmunología , Proteínas Bacterianas/genética , Mycobacterium bovis/enzimología , Mycobacterium tuberculosis/inmunología , Eliminación de Secuencia , Tuberculosis Pulmonar/prevención & control , Fosfatasa Ácida/inmunología , Animales , Vacuna BCG/administración & dosificación , Vacuna BCG/genética , Proteínas Bacterianas/inmunología , Citocinas/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium bovis/genética , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/fisiología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA