Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 15(9): e1008036, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31525249

RESUMEN

Cytomegalovirus (CMV) is a ubiquitous ß-herpesvirus that establishes life-long latent infection in a high percentage of the population worldwide. CMV induces the strongest and most durable CD8+ T cell response known in human clinical medicine. Due to its unique properties, the virus represents a promising candidate vaccine vector for the induction of persistent cellular immunity. To take advantage of this, we constructed a recombinant murine CMV (MCMV) expressing an MHC-I restricted epitope from influenza A virus (IAV) H1N1 within the immediate early 2 (ie2) gene. Only mice that were immunized intranasally (i.n.) were capable of controlling IAV infection, despite the greater potency of the intraperitoneally (i.p.) vaccination in inducing a systemic IAV-specific CD8+ T cell response. The protective capacity of the i.n. immunization was associated with its ability to induce IAV-specific tissue-resident memory CD8+ T (CD8TRM) cells in the lungs. Our data demonstrate that the protective effect exerted by the i.n. immunization was critically mediated by antigen-specific CD8+ T cells. CD8TRM cells promoted the induction of IFNγ and chemokines that facilitate the recruitment of antigen-specific CD8+ T cells to the lungs. Overall, our results showed that locally applied MCMV vectors could induce mucosal immunity at sites of entry, providing superior immune protection against respiratory infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa , Vacunas contra la Influenza/inmunología , Muromegalovirus/inmunología , Administración Intranasal , Secuencia de Aminoácidos , Animales , Línea Celular , Quimiocinas/biosíntesis , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Productos del Gen env/administración & dosificación , Productos del Gen env/genética , Productos del Gen env/inmunología , Vectores Genéticos , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Humana/inmunología , Gripe Humana/prevención & control , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/genética , Células 3T3 NIH , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
2.
Int J Mol Sci ; 19(9)2018 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-30200513

RESUMEN

The respiratory tract is constantly exposed to the environment and displays a favorable niche for colonizing microorganisms. However, the effects of respiratory bacterial carriage on the immune system and its implications for secondary responses remain largely unclear. We have employed respiratory carriage with Bordetella bronchiseptica as the underlying model to comprehensively address effects on subsequent immune responses. Carriage was associated with the stimulation of Bordetella-specific CD4⁺, CD8⁺, and CD4⁺CD25⁺Foxp3⁺ T cell responses, and broad transcriptional activation was observed in CD4⁺CD25⁺ T cells. Importantly, transfer of leukocytes from carriers to acutely B. bronchiseptica infected mice, resulted in a significantly increased bacterial burden in the recipient's upper respiratory tract. In contrast, we found that respiratory B. bronchiseptica carriage resulted in a significant benefit for the host in systemic infection with Listeria monocytogenes. Adaptive responses to vaccination and influenza A virus infection, were unaffected by B. bronchiseptica carriage. These data showed that there were significant immune modulatory processes triggered by B. bronchiseptica carriage, that differentially affect subsequent immune responses. Therefore, our results demonstrated the complexity of immune regulation induced by respiratory bacterial carriage, which can be beneficial or detrimental to the host, depending on the pathogen and the considered compartment.


Asunto(s)
Bordetella bronchiseptica/inmunología , Coinfección/inmunología , Infecciones del Sistema Respiratorio/inmunología , Linfocitos T Reguladores/microbiología , Vacunación , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Infecciones por Bordetella/sangre , Infecciones por Bordetella/inmunología , Infecciones por Bordetella/microbiología , Infecciones por Bordetella/prevención & control , Bordetella bronchiseptica/genética , Antígenos CD5/análisis , Portador Sano/inmunología , Portador Sano/microbiología , Coinfección/sangre , Coinfección/microbiología , Coinfección/prevención & control , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones del Sistema Respiratorio/sangre , Infecciones del Sistema Respiratorio/prevención & control , Linfocitos T Reguladores/inmunología
3.
J Nat Prod ; 80(5): 1531-1540, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28398049

RESUMEN

Six novel bioactive bicyclic polyketides (1-6) were isolated from cultures of an endophytic fungus of the medicinal plant Globularia alypum collected in Batna, Algeria. The producer organism was identified as Preussia similis using morphological and molecular phylogenetic methods. The structures of metabolites 1-6, for which the trivial names preussilides A-F are proposed, were elucidated using a combination of spectral methods, including extensive 2D NMR spectroscopy, high-resolution mass spectrometry, and CD spectroscopy. Preussilides were tested for antimicrobial and antiproliferative effects, and, in particular, compounds 1 and 3 showed selective activities against eukaryotes. Subsequent studies on the influence of 1 and 3 on the morphology of human osteosarcoma cells (U2OS) suggest that these two polyketides might target an enzyme involved in coordination of the cell division cycle. Hence, they might, for instance, affect timing or spindle assembly mechanisms, leading to defects in chromosome segregation and/or spindle geometry.


Asunto(s)
Antiinfecciosos/aislamiento & purificación , Antiinfecciosos/farmacología , Penicillium/química , Policétidos/aislamiento & purificación , Policétidos/farmacología , Antiinfecciosos/química , Humanos , Espectroscopía de Resonancia Magnética , Estructura Molecular , Filogenia , Policétidos/química
4.
Nature ; 479(7374): 547-51, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22080947

RESUMEN

Upon the aberrant activation of oncogenes, normal cells can enter the cellular senescence program, a state of stable cell-cycle arrest, which represents an important barrier against tumour development in vivo. Senescent cells communicate with their environment by secreting various cytokines and growth factors, and it was reported that this 'secretory phenotype' can have pro- as well as anti-tumorigenic effects. Here we show that oncogene-induced senescence occurs in otherwise normal murine hepatocytes in vivo. Pre-malignant senescent hepatocytes secrete chemo- and cytokines and are subject to immune-mediated clearance (designated as 'senescence surveillance'), which depends on an intact CD4(+) T-cell-mediated adaptive immune response. Impaired immune surveillance of pre-malignant senescent hepatocytes results in the development of murine hepatocellular carcinomas (HCCs), thus showing that senescence surveillance is important for tumour suppression in vivo. In accordance with these observations, ras-specific Th1 lymphocytes could be detected in mice, in which oncogene-induced senescence had been triggered by hepatic expression of Nras(G12V). We also found that CD4(+) T cells require monocytes/macrophages to execute the clearance of senescent hepatocytes. Our study indicates that senescence surveillance represents an important extrinsic component of the senescence anti-tumour barrier, and illustrates how the cellular senescence program is involved in tumour immune surveillance by mounting specific immune responses against antigens expressed in pre-malignant senescent cells.


Asunto(s)
Senescencia Celular/inmunología , Hepatocitos/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/patología , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/prevención & control , Senescencia Celular/genética , Progresión de la Enfermedad , Genes ras/genética , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/citología , Hígado/inmunología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/prevención & control , Ratones , Ratones SCID , Fagocitosis , Lesiones Precancerosas/genética , Lesiones Precancerosas/prevención & control
5.
Lung ; 195(1): 59-68, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27864635

RESUMEN

BACKGROUND: COPD represents a multifactorial lung disorder with high morbidity and mortality. Despite intensive research concerning the underlying disease mechanisms, the involvement of the CD200/CD200R axis in supporting or preventing the onset of COPD has not yet been addressed. Since the CD200/CD200R axis is crucially implicated in the maintenance of pulmonary immune homeostasis, we hypothesized that it might be involved in controlling the onset of COPD. METHODS: To address this, we analyzed the serum samples from COPD patients and normal controls for soluble (s) CD200 and correlated the data to COPD-relevant clinical parameters. In addition, basic studies were conducted in CD200-deficient and wild-type mice in which COPD-like inflammation was induced with elastase/LPS followed by lung and serum component analysis. RESULTS: We observed a positive correlation between serum sCD200 and IL-6 levels as well as a trend toward a negative correlation of sCD200 with vitamin D3 in COPD patients. Further investigations in mice revealed that despite elevated serum concentration of MMP-9 in CD200KO mice, the early onset of COPD-like lung inflammation was similar in CD200-deficient and wild-type animals in terms of immune cell infiltration, emphysematous changes, and mucus overproduction. CONCLUSIONS: While our murine studies suggest that the co-inhibitory molecule CD200 does not appear to play a prominent role in the early onset of COPD-like features, correlation of sCD200 serum levels with COPD-related parameters in humans with established disease revealed that the CD200/CD200R axis may be mechanistically linked to the disease course in COPD patients.


Asunto(s)
Antígenos CD/sangre , Antígenos CD/genética , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/patología , Anciano , Animales , Antígenos de Superficie/metabolismo , Estudios de Casos y Controles , Colecalciferol/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-6/sangre , Lipopolisacáridos , Linfocitos/patología , Macrófagos Alveolares/patología , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neutrófilos/patología , Receptores de Orexina , Elastasa Pancreática , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Receptores de Superficie Celular/metabolismo
6.
Int J Cancer ; 138(8): 1982-93, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26619320

RESUMEN

The importance of tumor associated neutrophils (TANs) in cancer development is in the meantime well established. Numerous of clinical data document the adverse prognostic effects of neutrophil infiltration in solid tumors. However, certain tumor therapies need functional neutrophils to be effective, suggesting altered neutrophil polarization associated with different outcomes for cancer patients. Therefore, modulation of neutrophilic phenotypes represents a potent therapeutic option, but factors mediating neutrophil polarization are still poorly defined. In this manuscript we provide evidence that type I IFNs alter neutrophilic phenotype into anti-tumor, both in mice and human. In the absence of IFN-ß, pro-tumor properties, such as reduced tumor cytotoxicity with low neutrophil extracellular traps (NETs) expression, low ICAM1 and TNF-α expression, dominated neutrophil phenotypes in primary lesion and premetastatic lung. Interestingly, such neutrophils have significantly prolonged life-span. Notably, interferon therapy in mice altered TAN polarization towards anti-tumor N1. Similar changes in neutrophil activation could be observed in melanoma patients undergoing type I IFN therapy. Altogether, these data highlight the therapeutic potential of interferons, suggesting optimization of its clinical use as potent anti-tumor agent.


Asunto(s)
Interferón Tipo I/inmunología , Melanoma/inmunología , Neoplasias Experimentales/inmunología , Neutrófilos/inmunología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Separación Celular , Femenino , Citometría de Flujo , Humanos , Interferón Tipo I/uso terapéutico , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias Experimentales/patología , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Fenotipo
7.
Eur J Immunol ; 44(12): 3632-45, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25159127

RESUMEN

Under physiological conditions, studies on the biology of naturally induced Foxp3(+) Treg cells of intra- and extrathymic origin have been hampered by the lack of unambiguous markers to discriminate the mature progeny of such developmental Treg-cell sublineages. Here, we report on experiments in double-transgenic mice, in which red fluorescent protein (RFP) is expressed in all Foxp3(+) Treg cells, whereas Foxp3-dependent GFP expression is exclusively confined to intrathymically induced Foxp3(+) Treg cells. This novel molecular genetic tool enabled us to faithfully track and characterize naturally induced Treg cells of intrathymic (RFP(+) GFP(+) ) and extrathymic (RFP(+) GFP(-) ) origin in otherwise unmanipulated mice. These experiments directly demonstrate that extrathymically induced Treg cells substantially contribute to the overall pool of mature Foxp3(+) Treg cells residing in peripheral lymphoid tissues of steady-state mice. Furthermore, we provide evidence that intra- and extrathymically induced Foxp3(+) Treg cells represent distinct phenotypic and functional sublineages.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Proteínas Fluorescentes Verdes/inmunología , Proteínas Luminiscentes/inmunología , Linfocitos T Reguladores , Timo , Animales , Factores de Transcripción Forkhead/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Timo/citología , Timo/inmunología , Proteína Fluorescente Roja
8.
Eur J Immunol ; 43(6): 1499-510, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23505065

RESUMEN

Dysregulation of apoptosis caused by an imbalance of pro- and anti-apoptotic protein expression can lead to cancer, neurodegenerative, and autoimmune diseases. Cellular-FLIP (c-FLIP) proteins inhibit apoptosis directly at the death-inducing signaling complex of death receptors, such as CD95, and have been linked to apoptosis regulation during immune responses. While the isoforms c-FLIPL and c-FLIPS are well characterized, the function of c-FLIPR remains poorly understood. Here, we demonstrate the induction of endogenous murine c-FLIPR in activated lymphocytes for the first time. To analyze c-FLIPR function in vivo, we generated transgenic mice expressing murine c-FLIPR specifically in hematopoietic cells. As expected, lymphocytes from c-FLIPR transgenic mice were protected against CD95-induced apoptosis in vitro. In the steady state, transgenic mice had normal cell numbers and unaltered frequencies of B cells and T-cell subsets in lymphoid organs. However, when challenged with Listeria monocytogenes, c-FLIPR transgenic mice showed less liver necrosis and better bacterial clearance compared with infected wild-type mice. We conclude that c-FLIPR expression in hematopoietic cells supports an efficient immune response against bacterial infections.


Asunto(s)
Linfocitos B/inmunología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Hígado/inmunología , Isoformas de Proteínas/metabolismo , Linfocitos T/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/inmunología , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Hígado/microbiología , Hígado/patología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Necrosis/genética , Necrosis/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptor fas/metabolismo
9.
J Immunol ; 186(11): 6106-18, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21518973

RESUMEN

Every person harbors a population of potentially self-reactive lymphocytes controlled by tightly balanced tolerance mechanisms. Failures in this balance evoke immune activation and autoimmunity. In this study, we investigated the contribution of self-reactive CD8(+) T lymphocytes to chronic pulmonary inflammation and a possible role for naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (nTregs) in counterbalancing this process. Using a transgenic murine model for autoimmune-mediated lung disease, we demonstrated that despite pulmonary inflammation, lung-specific CD8(+) T cells can reside quiescently in close proximity to self-antigen. Whereas self-reactive CD8(+) T cells in the inflamed lung and lung-draining lymph nodes downregulated the expression of effector molecules, those located in the spleen appeared to be partly Ag-experienced and displayed a memory-like phenotype. Because ex vivo-reisolated self-reactive CD8(+) T cells were very well capable of responding to the Ag in vitro, we investigated a possible contribution of nTregs to the immune control over autoaggressive CD8(+) T cells in the lung. Notably, CD8(+) T cell tolerance established in the lung depends only partially on the function of nTregs, because self-reactive CD8(+) T cells underwent only biased activation and did not acquire effector function after nTreg depletion. However, although transient ablation of nTregs did not expand the population of self-reactive CD8(+) T cells or exacerbate the disease, it provoked rapid accumulation of activated CD103(+)CD62L(lo) Tregs in bronchial lymph nodes, a finding suggesting an adaptive phenotypic switch in the nTreg population that acts in concert with other yet-undefined mechanisms to prevent the detrimental activation of self-reactive CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Neumonía/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Tolerancia Inmunológica/inmunología , Memoria Inmunológica/inmunología , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/inmunología , Cadenas alfa de Integrinas/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Selectina L/genética , Selectina L/inmunología , Selectina L/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Neumonía/genética , Neumonía/metabolismo , Pruebas de Función Respiratoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/metabolismo
10.
Am J Respir Cell Mol Biol ; 47(6): 869-78, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22984087

RESUMEN

Although the contribution of CD8(+) T cells to the pathogenesis of noncommunicable lung diseases has become increasingly appreciated, our knowledge about the mechanisms controlling self-reactive CD8(+) T cells in the respiratory tract remains largely elusive. The outcome of the encounter between pulmonary self-antigen and naive CD8(+) T cells, in the presence or absence of inflammation, was traced after adoptive transfer of fluorescence-labeled CD8(+) T cells specific for the neo-self-antigen influenza A hemagglutinin into transgenic mice expressing hemagglutinin specifically in alveolar type II epithelial cells in order: to study the outcome of alveolar antigen encounter in the steady state and under inflammatory conditions; to define the phenotype and fate of CD8(+) T cells primed in the respiratory tract; and, finally, to correlate these findings with the onset of autoimmunity in the lung. We found that CD8(+) T cells remain ignorant in the steady state, whereas transient proliferation of self-reactive CD8(+) T cells is induced by forced maturation or licensing of dendritic cells, increases in the antigenic threshold, and targeted release of alveolar self-antigen by epithelial injury. However, these cells fail to acquire effector functions, lack the expression of the high-affinity IL-2 receptor CD25, and do not precipitate autoimmunity in the lung. We conclude that inadvertent activation of CD8(+) T cells in the lung is prevented in the absence of "danger signals," whereas tissue damage after infection or noninfectious inflammation creates an environment that allows the priming of previously ignorant T cells. Failure in effector cell differentiation after abortive priming, however, precludes the establishment of self-perpetuating autoimmunity in the lung.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Alveolos Pulmonares/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/fisiología , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular , Proliferación Celular , Citotoxicidad Inmunológica , Mediadores de Inflamación/metabolismo , Interleucina-2/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Transducción de Señal
11.
Am J Respir Crit Care Med ; 179(5): 344-55, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19096007

RESUMEN

RATIONALE: Although the contribution of alveolar type II epithelial cells (AECIIs) in respiratory immunity has become increasingly appreciated, their precise function in the induction and regulation of T-cell reactivity to self-antigen remains poorly understood. OBJECTIVES: To investigate the role of AECII in the initiation of T-cell reactivity to alveolar self-antigen, and to clarify their function in the peripheral induction of Foxp3(+) regulatory CD4(+) T cells. METHODS: To dissect the complex cellular and molecular functions of AECIIs in lung inflammation and immune regulation, we use a transgenic mouse model for CD4(+) T-cell-mediated pulmonary inflammation. MEASUREMENTS AND MAIN RESULTS: Here we report that AECIIs present endogenously expressed antigen on major histocompatibility complex class II molecules to CD4(+) T cells. Epithelial antigen display was sufficient to induce primary T-cell activation and pulmonary inflammation. Upon inflammation, AECIIs induce the differentiation of Foxp3(+) regulatory T cells by a mechanism involving antiproliferative soluble factors, including transforming growth factor (TGF)-beta. Whereas, in acute inflammation, TGF-beta appears to be the dominant factor to induce regulatory T cells, other AECII-derived factors can substitute for and/or synergize with TGF-beta in chronic pulmonary inflammations. CONCLUSIONS: AECIIs are capable of priming naive CD4(+) T cells, demonstrating an active participation of these cells in respiratory immunity. Moreover, AECIIs display as yet unrecognized functions in balancing inflammatory and regulatory T-cell responses in the lung by connecting innate and adaptive immune mechanisms to establish peripheral T-cell tolerance to respiratory self-antigen.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Neumonía/inmunología , Alveolos Pulmonares/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoantígenos/inmunología , Factores de Transcripción Forkhead/biosíntesis , Hemaglutininas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neumonía/patología , Alveolos Pulmonares/citología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/inmunología
12.
PLoS One ; 14(10): e0223060, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31577832

RESUMEN

In this study, we analyzed PRRS virus (PRRSv) specific lymphocyte function in piglets vaccinated with Ingelvac PRRSFLEX EU® at two and three weeks of age in the presence of homologous maternal immunity. Complete analysis of maternal immunity to PRRSv was evaluated postpartum, as well as passive transfer of antibodies and T cells to the piglet through colostrum intake and before and after challenge with a heterologous PRRSv at ten weeks of age. Maternal-derived antibodies were detected in piglets but declined quickly after weaning. However, vaccinated animals restored PRRSv-specific antibody levels by anamnestic response to vaccination. Cell analysis in colostrum and milk revealed presence of PRRSv-specific immune cells at suckling with higher concentrations found in colostrum than in milk. In addition, colostrum and milk contained PRRSv-specific IgA and IgG that may contribute to protection of newborn piglets. Despite the presence of PRRSv-specific Peripheral Blood Mononuclear cells (PBMCs) in colostrum and milk, no PRRSv-specific cells could be detected from blood of the piglets at one or two weeks of life. Nevertheless, cellular immunity was detectable in pre-challenged piglets up to 7 weeks after vaccination while the non-vaccinated control group showed no interferon (IFN) γ response to PRRSv stimulation. After challenge, all piglets developed a PRRSv-specific IFNγ-response, which was more robust at significantly higher levels in vaccinated animals compared to the primary response to PRRSv in non-vaccinated animals. Cytokine analysis in the lung lumen showed a reduction of pro-inflammatory responses to PRRSv challenge in vaccinated animals, especially reduced interferon (IFN) α levels. In conclusion, vaccination of maternally positive piglets at 2 and 3 weeks of age with Ingelvac PRRSFLEX EU induced a humoral and cellular immune response to PRRSv and provided protection against virulent, heterologous PRRSv challenge.


Asunto(s)
Inmunidad Materno-Adquirida , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vacunación , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Líquido del Lavado Bronquioalveolar/virología , Calostro/citología , Citocinas/metabolismo , Femenino , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Pulmón/patología , Leche/citología , Especificidad de la Especie , Porcinos , Viremia/inmunología , Viremia/virología
13.
PLoS One ; 13(12): e0207358, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30540779

RESUMEN

The adhesion molecule and co-receptor of receptor tyrosine kinases, CD44, is expressed in all cells of the immune system, but also in numerous non-immune cells. CD44 plays roles in the cellular response to different pathogens. The molecular actions of CD44 during these processes are by and large still unknown. The CD44 molecule undergoes a sequential proteolytic cleavage which leads to the release of a soluble intracellular domain (CD44-ICD). Previous reports had shown that the CD44-ICD is taken up into the nucleus where it enhances transcription of specific target genes. By RNA profiling we identified a CD44-dependent transcriptional increase of interferon-responsive genes, among them IFI16. IFI16 is important in the innate immune response. It senses and binds pathogenic DNA and, together with cGAS, activates the cGAS-cGAMP-STING pathway and induces the expression of genes relevant for the response, e.g. IFN-ß. Our results show that the enhancement of IFI16 expression depended on CD44 cleavage. A CD44-negative tumor cell line, embryonic fibroblasts and bone marrow-derived macrophages from cd44-/- mice were reduced in their response to IFN-γ, to viral DNA fragments and to Listeria monocytogenes infection. We could rescue the deficiency of CD44 negative RPM-MC cells and cd44-/- MEFs by expressing only the soluble CD44-ICD in the absence of any other CD44 domain. Expression of the CD44-ICD carrying a mutation that prevented the uptake into the nucleus, could not rescue the absence of CD44. This molecular aspect of regulation by CD44 may explain part of the immune phenotypes of mice with cd44 gene disruption.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Receptores de Hialuranos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Células Cultivadas , Diaminas/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Receptores de Hialuranos/genética , Inmunidad Innata/efectos de los fármacos , Interferón beta/genética , Interferón beta/metabolismo , Interferón gamma/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Mutagénesis , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética , Tiazoles/farmacología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
14.
Respir Res ; 8: 47, 2007 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-17610738

RESUMEN

BACKGROUND: Although the contribution of alveolar type II epithelial cell (AEC II) activities in various aspects of respiratory immune regulation has become increasingly appreciated, our understanding of the contribution of AEC II transcriptosome in immunopathologic lung injury remains poorly understood. We have previously established a mouse model for chronic T cell-mediated pulmonary inflammation in which influenza hemagglutinin (HA) is expressed as a transgene in AEC II, in mice expressing a transgenic T cell receptor specific for a class II-restricted epitope of HA. Pulmonary inflammation in these mice occurs as a result of CD4+ T cell recognition of alveolar antigen. This model was utilized to assess the profile of inflammatory mediators expressed by alveolar epithelial target cells triggered by antigen-specific recognition in CD4+ T cell-mediated lung inflammation. METHODS: We established a method that allows the flow cytometric negative selection and isolation of primary AEC II of high viability and purity. Genome wide transcriptional profiling was performed on mRNA isolated from AEC II isolated from healthy mice and from mice with acute and chronic CD4+ T cell-mediated pulmonary inflammation. RESULTS: T cell-mediated inflammation was associated with expression of a broad array of cytokine and chemokine genes by AEC II cell, indicating a potential contribution of epithelial-derived chemoattractants to the inflammatory cell parenchymal infiltration. Morphologically, there was an increase in the size of activated epithelial cells, and on the molecular level, comparative transcriptome analyses of AEC II from inflamed versus normal lungs provide a detailed characterization of the specific inflammatory genes expressed in AEC II induced in the context of CD4+ T cell-mediated pneumonitis. CONCLUSION: An important contribution of AEC II gene expression to the orchestration and regulation of interstitial pneumonitis is suggested by the panoply of inflammatory genes expressed by this cell population, and this may provide insight into the molecular pathogenesis of pulmonary inflammatory states. CD4+ T cell recognition of antigen presented by AEC II cells appears to be a potent trigger for activation of the alveolar cell inflammatory transcriptosome.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunofenotipificación , Pulmón/patología , Alveolos Pulmonares/citología , Alveolos Pulmonares/inmunología , Mucosa Respiratoria/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Alveolos Pulmonares/metabolismo , Mucosa Respiratoria/metabolismo
15.
J Leukoc Biol ; 101(2): 407-419, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27605210

RESUMEN

The adhesion and degranulation promoting adaptor protein (ADAP) is a multifunctional scaffold involved in many different signaling pathways that are important for the function of T cells, including the inside-out and outside-in signaling of integrins, the activation of NF-κB, and the subsequent production of proinflammatory cytokines (e.g., IFN-γ and IL-2). Strikingly, despite its well-established role in T cells, previous studies did not distinguish between CD4+ and CD8+ T cells, and thus, it is unknown whether ADAP fulfills equally important functions in both T cell subsets. We show here that despite comparable ADAP expression levels in CD4+ and CD8+ T cells, their function is differentially dependent on ADAP. Whereas in vitro TCR-stimulation experiments revealed that activation, proliferation, and adhesion are severely compromised in CD4+ T cells lacking ADAP, their CD8+ counterparts are hardly affected by ADAP deficiency. Accordingly, antigen-specific in vivo stimulation of adoptively transferred CD8+ T cells during Listeria monocytogenes (Lm) and influenza A virus (IAV) infection revealed only moderate effects of ADAP deficiency in terms of CD8+ T cell activation, proliferation, and differentiation, which, however, did not impair pathogen-specific immunity. Thus, we show for the first time that ADAP fulfills different functions in CD4+ and CD8+ T cells, with CD8+ T cells being less dependent on ADAP. Our data identify ADAP as a potential molecular target for T cell subset-specific therapeutic interventions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Inmunidad , Listeria monocytogenes/inmunología , Activación de Linfocitos/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Epítopos/inmunología , Interferón gamma/biosíntesis , Subgrupos Linfocitarios/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Quimiocina/metabolismo
16.
Sci Rep ; 7: 43985, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28266658

RESUMEN

Hepatotropic viruses such as hepatitis C virus cause life-threatening chronic liver infections in millions of people worldwide. Targeted in vivo antigen-delivery to cross-presenting dendritic cells (DCs) has proven to be extraordinarily efficient in stimulating antigen-specific T cell responses. To determine whether this approach would as well be suitable to induce local antiviral effector T cells in the liver we compared different vaccine formulations based on either the targeting of DEC-205 or TLR2/6 on cross-presenting DCs or formulations not involving in vivo DC targeting. As read-outs we used in vivo hepatotropic adenovirus challenge, histology and automated multidimensional fluorescence microscopy (MELC). We show that targeted in vivo antigen delivery to cross-presenting DCs is highly effective in inducing antiviral CTLs capable of eliminating virus-infected hepatocytes, while control vaccine formulation not involving DC targeting failed to induce immunity against hepatotropic virus. Moreover, we observed distinct patterns of CD8+ T cell interaction with virus-infected and apoptotic hepatocytes in the two DC-targeting groups suggesting that the different vaccine formulations may stimulate distinct types of effector functions. Our findings represent an important step toward the future development of vaccines against hepatotropic viruses and the treatment of patients with hepatic virus infection after liver transplantation to avoid reinfection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunización/métodos , Hígado/inmunología , Ovalbúmina/inmunología , Animales , Antígenos CD/metabolismo , Reactividad Cruzada , Femenino , Lectinas Tipo C/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Toll-Like/metabolismo
17.
Curr Med Chem ; 23(15): 1549-70, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27048335

RESUMEN

The isoprenoid stress-associated phytohormone abscisic acid (ABA) has recently been recognized to possess multifaceted biological functions in mammals and to exert potent curative effects in a number of clinically relevant human diseases. Studies with human specimens have unequivocally shown that ABA retains its stress-related functional attributes, previously identified in plants, which contribute to enhanced inflammatory defense mechanisms in mammals. Besides, studies performed in animal models revealed prominent anti-inflammatory properties of ABA as indicated by a marked reduction of immune cell infiltrates at the sites of inflammation. Thus, ABA treatment ultimately leads to the profound improvement of both non-communicable and communicable diseases which are associated with an overall alleviated course of inflammation. In addition to its action on the mammalian immune system, ABA was also shown to exert diverse physiological functions on non-immune components. One of the most remarkable features of ABA is to stimulate and expand mesenchymal stem cells, which may open a new avenue for its potential use in the field of regenerative medicine. Furthermore, ABA has been reported to play an important role in the maintenance of glycemic control. In this review, we summarize current understanding of the significance of ABA in the mammalian system, its prophylactic and therapeutic effects in various disease settings and the future directions for the development of ABA as novel drug candidate for the improved treatment of inflammatory and infectious human diseases.


Asunto(s)
Ácido Abscísico/farmacología , Antiinflamatorios no Esteroideos/farmacología , Enfermedades Transmisibles/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Ácido Abscísico/química , Animales , Antiinflamatorios no Esteroideos/química , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos
18.
mBio ; 7(3)2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27143386

RESUMEN

UNLABELLED: Influenza A virus (IAV) periodically causes substantial morbidity and mortality in the human population. In the lower lung, the primary targets for IAV replication are type II alveolar epithelial cells (AECII), which are increasingly recognized for their immunological potential. So far, little is known about their reaction to IAV and their contribution to respiratory antiviral immunity in vivo Therefore, we characterized the AECII response during early IAV infection by analyzing transcriptional regulation in cells sorted from the lungs of infected mice. We detected rapid and extensive regulation of gene expression in AECII following in vivo IAV infection. The comparison to transcriptional regulation in lung tissue revealed a strong contribution of AECII to the respiratory response. IAV infection triggered the expression of a plethora of antiviral factors and immune mediators in AECII with a high prevalence for interferon-stimulated genes. Functional pathway analyses revealed high activity in pathogen recognition, immune cell recruitment, and antigen presentation. Ultimately, our analyses of transcriptional regulation in AECII and lung tissue as well as interferon I/III levels and cell recruitment indicated AECII to integrate signals provided by direct pathogen recognition and surrounding cells. Ex vivo analysis of AECII proved a powerful tool to increase our understanding of their role in respiratory immune responses, and our results clearly show that AECII need to be considered a part of the surveillance and effector system of the lower respiratory tract. IMPORTANCE: In order to confront the health hazard posed by IAV, we need to complete our understanding of its pathogenesis. AECII are primary targets for IAV replication in the lung, and while we are beginning to understand their importance for respiratory immunity, the in vivo AECII response during IAV infection has not been analyzed. In contrast to studies addressing the response of AECII infected with IAV ex vivo, we have performed detailed gene transcriptional profiling of AECII isolated from the lungs of infected mice. Thereby, we have identified an exceptionally rapid and versatile response to IAV infection that is shaped by pathogen-derived as well as microenvironment-derived signals and aims at the induction of antiviral measures and the recruitment and activation of immune cells. In conclusion, our study presents AECII as active players in antiviral defense in vivo that need to be considered part of the sentinel and effector immune system of the lung.


Asunto(s)
Células Epiteliales Alveolares/inmunología , Virus de la Influenza A/inmunología , Pulmón/inmunología , Pulmón/patología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Interferones/análisis , Leucocitos/inmunología , Ratones
19.
Int J Genomics ; 2015: 638032, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25811019

RESUMEN

Asthma is the consequence of allergic inflammation in the lung compartments and lung-draining lymph nodes. Dendritic cells initiate and promote T cell response and drive it to immunity or allergy. However, their modes of action during asthma are poorly understood. In this study, an allergic inflammation with ovalbumin was induced in 38 mice versus 42 control animals. After ovalbumin aerosol challenge, conventional dendritic cells (CD11c/MHCII/CD8) were isolated from the lungs and the draining lymph nodes by means of magnetic cell sorting followed by fluorescence-activated cell sorting. A comparative transcriptional analysis was performed using gene arrays. In general, many transcripts are up- and downregulated in the CD8(-) dendritic cells of the allergic inflamed lung tissue, whereas few genes are regulated in CD8(+) dendritic cells. The dendritic cells of the lymph nodes also showed minor transcriptional changes. The data support the relevance of the CD8(-) conventional dendritic cells but do not exclude distinct functions of the small population of CD8(+) dendritic cells, such as cross presentation of external antigen. So far, this is the first approach performing gene arrays in dendritic cells obtained from lung tissue and lung-draining lymph nodes of asthmatic-like mice.

20.
PLoS One ; 9(7): e100970, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25029240

RESUMEN

Inducible Co-stimulator (ICOS) plays a critical role in mediating T cell differentiation and function and is considered a key player in balancing T effector and T regulatory (Treg) cell responses. Here we show that activation of the ICOS signalling pathway during acute influenza A virus (IAV) infection by application of an agonistic ICOS antibody reduced the frequency of CD8+ T cells in the respiratory tract of IAV infected animals and delayed pathogen elimination. In line with this, immune-mediated influenza pneumonia was significantly ameliorated in mice that received ICOS agonist as indicated by significantly reduced alveolar infiltrations and bronchointerstitial pneumonia, while at the same time virus-related pathology remained unaffected. Importantly, ICOS agonist treatment resulted in expansion of CD4+Foxp3+ Tregs in IAV infected mice, which was associated with elevated levels of the immunosuppressive cytokine IL-10 in the alveolar space. Together, our findings suggest a prominent role of ICOS signaling during acute IAV infection by increasing the Treg/CD8+ T cell ratio with beneficial outcome on immune-mediated pneumonia and underline the suitability of ICOS as potential therapeutic target for immune intervention in those infectious conditions characterized by strong immunopathology rather than virus-mediated cytopathic effects.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Terapia Molecular Dirigida/métodos , Infecciones por Orthomyxoviridae/complicaciones , Neumonía/tratamiento farmacológico , Neumonía/inmunología , Animales , Femenino , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/agonistas , Interleucina-10/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Ratones , Neumonía/complicaciones , Neumonía/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA