Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nat Immunol ; 18(5): 509-518, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28319098

RESUMEN

The retention of intracellular Toll-like receptors (TLRs) in the endoplasmic reticulum prevents their activation under basal conditions. TLR9 is activated by sensing ligands in specific endosomal-lysosomal compartments. Here we identified IRAP+ endosomes as major cellular compartments for the early steps of TLR9 activation in dendritic cells (DCs). Both TLR9 and its ligand, the dinucleotide CpG, were present as cargo in IRAP+ endosomes. In the absence of the aminopeptidase IRAP, the trafficking of CpG and TLR9 to lysosomes and signaling via TLR9 were enhanced in DCs and in mice following bacterial infection. IRAP stabilized CpG-containing endosomes by interacting with the actin-nucleation factor FHOD4, which slowed the trafficking of TLR9 toward lysosomes. Thus, endosomal retention of TLR9 via the interaction of IRAP with the actin cytoskeleton is a mechanism that prevents hyper-activation of TLR9 in DCs.


Asunto(s)
Cistinil Aminopeptidasa/metabolismo , Citoesqueleto/metabolismo , Células Dendríticas/fisiología , Endosomas/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Células Cultivadas , Islas de CpG/genética , Cistinil Aminopeptidasa/genética , Células Dendríticas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Transducción de Señal
2.
Immunity ; 46(2): 301-314, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228284

RESUMEN

Respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infections in infants and is characterized by pulmonary infiltration of B cells in fatal cases. We analyzed the B cell compartment in human newborns and identified a population of neonatal regulatory B lymphocytes (nBreg cells) that produced interleukin 10 (IL-10) in response to RSV infection. The polyreactive B cell receptor of nBreg cells interacted with RSV protein F and induced upregulation of chemokine receptor CX3CR1. CX3CR1 interacted with RSV glycoprotein G, leading to nBreg cell infection and IL-10 production that dampened T helper 1 (Th1) cytokine production. In the respiratory tract of neonates with severe RSV-induced acute bronchiolitis, RSV-infected nBreg cell frequencies correlated with increased viral load and decreased blood memory Th1 cell frequencies. Thus, the frequency of nBreg cells is predictive of the severity of acute bronchiolitis disease and nBreg cell activity may constitute an early-life host response that favors microbial pathogenesis.


Asunto(s)
Linfocitos B Reguladores/inmunología , Bronquiolitis Viral/inmunología , Receptores de Quimiocina/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Linfocitos B Reguladores/virología , Bronquiolitis Viral/patología , Linfocitos T CD4-Positivos/inmunología , Receptor 1 de Quimiocinas CX3C , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Perfilación de la Expresión Génica , Humanos , Recién Nacido , Activación de Linfocitos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios , Transcriptoma
3.
Vet Res ; 54(1): 36, 2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069656

RESUMEN

Bovine respiratory syncytial virus (BRSV) is a pathogenic pneumovirus and a major cause of acute respiratory infections in calves. Although different vaccines are available against BRSV, their efficiency remains limited, and no efficient and large-scale treatment exists. Here, we developed a new reverse genetics system for BRSV expressing the red fluorescent protein mCherry, based on a field strain isolated from a sick calf in Sweden. Although this recombinant fluorescent virus replicated slightly less efficiently compared to the wild type virus, both viruses were shown to be sensitive to the natural steroidal alkaloid cyclopamine, which was previously shown to inhibit human RSV replication. Our data thus point to the potential of this recombinant fluorescent BRSV as a powerful tool in preclinical drug discovery to enable high throughput compound screening.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Bovino , Virus Sincitial Respiratorio Humano , Animales , Bovinos , Humanos , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/veterinaria , Antivirales/farmacología , Antivirales/uso terapéutico , Virus Sincitial Respiratorio Humano/metabolismo , Anticuerpos Antivirales
4.
J Virol ; 95(22): e0091221, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34431698

RESUMEN

Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de Neoplasias/inmunología , Proteínas de la Nucleocápside/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Línea Celular , Cricetinae , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Replicación Viral
5.
FASEB J ; 35(4): e21348, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33715218

RESUMEN

The gut microbiota contributes to shaping efficient and safe immune defenses in the gut. However, little is known about the role of the gut and/or lung microbiota in the education of pulmonary innate immune responses. Here, we tested whether the endogenous microbiota in general can modulate the reactivity of pulmonary tissue to pathogen stimuli by comparing the response of specific-pathogen-free (SPF) and germ-free (GF) mice. Thus, we observed earlier and greater inflammation in the pulmonary compartment of GF mice than that of SPF mice after intranasal instillation to lipopolysaccharide (LPS), a component of Gram-negative bacteria. Toll-like receptor 4 (TLR4) was more abundantly expressed in the lungs of GF mice than those of SPF mice at steady state, which could predispose the innate immunity of GF mice to strongly react to the environmental stimuli. Lung explants were stimulated with different TLR agonists or infected with the human airways pathogen, respiratory syncytial virus (RSV), resulting in greater inflammation under almost all conditions for the GF explants. Finally, alveolar macrophages (AM) from GF mice presented a higher innate immune response upon RSV infection than those of SPF mice. Overall, these data suggest that the presence of microbiota in SPF mice induced a process of innate immune tolerance in the lungs by a mechanism which remains to be elucidated. Our study represents a step forward to establishing the link between the microbiota and the immune reactivity of the lungs.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Vida Libre de Gérmenes , Lipopolisacáridos/toxicidad , Pulmón/inmunología , Pulmón/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/metabolismo , Enfermedades Pulmonares/inducido químicamente , Masculino , Ratones , Organismos Libres de Patógenos Específicos , Técnicas de Cultivo de Tejidos , Receptor Toll-Like 4/genética
6.
PLoS Biol ; 17(2): e3000164, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30789898

RESUMEN

Throughout the last several decades, vaccination has been key to prevent and eradicate infectious diseases. However, many pathogens (e.g., respiratory syncytial virus [RSV], influenza, dengue, and others) have resisted vaccine development efforts, largely because of the failure to induce potent antibody responses targeting conserved epitopes. Deep profiling of human B cells often reveals potent neutralizing antibodies that emerge from natural infection, but these specificities are generally subdominant (i.e., are present in low titers). A major challenge for next-generation vaccines is to overcome established immunodominance hierarchies and focus antibody responses on crucial neutralization epitopes. Here, we show that a computationally designed epitope-focused immunogen presenting a single RSV neutralization epitope elicits superior epitope-specific responses compared to the viral fusion protein. In addition, the epitope-focused immunogen efficiently boosts antibodies targeting the palivizumab epitope, resulting in enhanced neutralization. Overall, we show that epitope-focused immunogens can boost subdominant neutralizing antibody responses in vivo and reshape established antibody hierarchies.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Epítopos/química , Receptores de Antígenos de Linfocitos B/inmunología , Proteínas Recombinantes de Fusión/química , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/química , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Clonación Molecular , Diseño Asistido por Computadora , Epítopos/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inmunización/métodos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Palivizumab/química , Palivizumab/inmunología , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/biosíntesis , Vacunas contra Virus Sincitial Respiratorio/genética , Homología Estructural de Proteína , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
7.
J Neurochem ; 155(2): 137-153, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31811775

RESUMEN

The olfactory mucosa, where the first step of odor detection occurs, is a privileged pathway for environmental toxicants and pathogens toward the central nervous system. Indeed, some pathogens can infect olfactory sensory neurons including their axons projecting to the olfactory bulb allowing them to bypass the blood-brain barrier and reach the central nervous system (CNS) through the so-called olfactory pathway. The respiratory syncytial virus (RSV) is a major respiratory tract pathogen but there is growing evidence that RSV may lead to CNS impairments. However, the mechanisms involved in RSV entering into the CNS have been poorly described. In this study, we wanted to explore the capacity of RSV to reach the CNS via the olfactory pathway and to better characterize RSV cellular tropism in the nasal cavity. We first explored the distribution of RSV infectious sites in the nasal cavity by in vivo bioluminescence imaging and a tissue clearing protocol combined with deep-tissue imaging and 3D image analyses. This whole tissue characterization was confirmed with immunohistochemistry and molecular biology approaches. Together, our results provide a novel 3D atlas of mouse nasal cavity anatomy and show that RSV can infect olfactory sensory neurons giving access to the central nervous system by entering the olfactory bulb. Cover Image for this issue: doi: 10.1111/jnc.14765.


Asunto(s)
Mucosa Olfatoria/inervación , Mucosa Olfatoria/virología , Neuronas Receptoras Olfatorias/virología , Virus Sincitiales Respiratorios , Animales , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/virología , Enfermedades del Sistema Nervioso Central/diagnóstico por imagen , Enfermedades del Sistema Nervioso Central/virología , Femenino , Cabeza/anatomía & histología , Imagenología Tridimensional , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/virología , Bulbo Olfatorio/virología , Mucosa Olfatoria/diagnóstico por imagen , ARN Viral/aislamiento & purificación , Tropismo , Replicación Viral
8.
Eur J Immunol ; 46(4): 874-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26681580

RESUMEN

Respiratory syncytial virus (RSV) causes severe bronchiolitis in infants worldwide. The immunological factors responsible for RSV susceptibility in infants are poorly understood. Here, we used the BALB/c mouse model of neonatal RSV infection to study the mechanisms leading to severe disease upon reexposure to the virus when adults. Two major deficiencies in neonatal lung innate responses were found: a poor DCs mobilization, and a weak engagement of the IFNI pathway. The administration of Flt3 ligand (Flt3-L), a growth factor that stimulates the proliferation of hematopoietic cells, to neonates before RSV-infection, resulted in increased lung DC number, and reconditioned the IFNI pathway upon RSV neonatal infection. Besides, neonates treated with Flt3-L were protected against exacerbated airway disease upon adult reexposure to RSV. This was associated with a reorientation of RSV-specific responses toward Th1-mediated immunity. Thus, the poor lung DCs and IFNI responses to RSV in neonates may be partly responsible for the deleterious long-term consequences revealed upon adult reexposure to RSV, which could be prevented by Flt3-L treatment. These results open new perspectives for developing neonatal immuno-modulating strategies to reduce the burden of bronchiolitis.


Asunto(s)
Bronquiolitis Viral/inmunología , Inmunidad Innata/inmunología , Proteínas de la Membrana/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Animales Recién Nacidos , Bronquiolitis Viral/prevención & control , Bronquiolitis Viral/virología , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Transducción de Señal/inmunología , Células TH1/inmunología
9.
Nanomedicine ; 13(2): 411-420, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27553073

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of acute respiratory infections in children, yet no vaccine is available. The sole licensed preventive treatment against RSV is composed of a monoclonal neutralizing antibody (palivizumab), which targets a conformational epitope located on the fusion protein (F). Palivizumab reduces the burden of bronchiolitis but does not prevent infection. Thus, the development of RSV vaccines remains a priority. We previously evaluated nanorings formed by RSV nucleoprotein (N) as an RSV vaccine, as well as an immunostimulatory carrier for heterologous antigens. Here, we linked the palivizumab-targeted epitope (called FsII) to N, to generate N-FsII-nanorings. Intranasal N-FsII immunization elicited anti-F antibodies in mice that were non-neutralizing in vitro. Nevertheless, RSV-challenged animals were better protected against virus replication than mice immunized with N-nanorings, especially in the upper airways. In conclusion, an N-FsII-focused vaccine is an attractive candidate combining N-specific cellular immunity and F-specific antibodies for protection.


Asunto(s)
Epítopos , Nanopartículas , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitiales Respiratorios , Proteínas Virales de Fusión , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Ratones , Palivizumab , Infecciones por Virus Sincitial Respiratorio/prevención & control , Sigmodontinae
10.
Proc Natl Acad Sci U S A ; 109(5): 1619-24, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307620

RESUMEN

A deficit in early clearance of Pseudomonas aeruginosa (P. aeruginosa) is crucial in nosocomial pneumonia and in chronic lung infections. Few studies have addressed the role of Toll-like receptors (TLRs), which are early pathogen associated molecular pattern receptors, in pathogen uptake and clearance by alveolar macrophages (AMs). Here, we report that TLR5 engagement is crucial for bacterial clearance by AMs in vitro and in vivo because unflagellated P. aeruginosa or different mutants defective in TLR5 activation were resistant to AM phagocytosis and killing. In addition, the clearance of PAK (a wild-type P. aeruginosa strain) by primary AMs was causally associated with increased IL-1ß release, which was dramatically reduced with PAK mutants or in WT PAK-infected primary TLR5(-/-) AMs, demonstrating the dependence of IL-1ß production on TLR5. We showed that this IL-1ß production was important in endosomal pH acidification and in inducing the killing of bacteria by AMs through asparagine endopeptidase (AEP), a key endosomal cysteine protease. In agreement, AMs from IL-1R1(-/-) and AEP(-/-) mice were unable to kill P. aeruginosa. Altogether, these findings demonstrate that TLR5 engagement plays a major role in P. aeruginosa internalization and in triggering IL-1ß formation.


Asunto(s)
Endopeptidasas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Alveolares/inmunología , Fagocitosis , Pseudomonas aeruginosa/inmunología , Receptor Toll-Like 5/fisiología , Animales , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL
11.
PLoS Pathog ; 8(8): e1002841, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916010

RESUMEN

Intracellular Toll-like receptors (TLRs) expressed by dendritic cells recognize nucleic acids derived from pathogens and play an important role in the immune responses against the influenza virus (IAV), a single-stranded RNA sensed by different receptors including TLR7. However, the importance of TLR7 processing in the development of anti-viral immune responses is not known. Here we report that asparagine endopeptidase (AEP) deficient mice are unable to generate a strong anti-IAV response, as demonstrated by reduced inflammation, cross presentation of cell-associated antigens and priming of CD8(+) T cells following TLR7-dependent pulmonary infection induced by IAV. Moreover, AEP deficient lung epithelial- or myeloid-cells exhibit impaired TLR7 signaling due to defective processing of this receptor. Indeed, TLR7 requires a proteolytic cleavage by AEP to generate a C-terminal fragment competent for signaling. Thus, AEP activity is critical for TLR7 processing, opening new possibilities for the treatment of influenza and TLR7-dependent inflammatory diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Endopeptidasas/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
12.
Am J Respir Crit Care Med ; 187(2): 170-9, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23220915

RESUMEN

RATIONALE: Cystic fibrosis transmembrane conductance regulator (CFTR) protein is a chloride channel regulating fluid homeostasis at epithelial surfaces. Its loss of function induces hypohydration, mucus accumulation, and bacterial infections in CF and potentially other lung chronic diseases. OBJECTIVES: To test whether neutrophil elastase (NE) and neutrophil-mediated inflammation negatively impact CFTR structure and function, in vitro and in vivo. METHODS: Using an adenovirus-CFTR overexpression approach, we showed that NE degrades wild-type (WT)- and ΔF508-CFTR in vitro and WT-CFTR in mice through a new pathway involving the activation of intracellular calpains. MEASUREMENTS AND MAIN RESULTS: CFTR degradation triggered a loss of function, as measured in vitro by channel patch-clamp and in vivo by nasal potential recording in mice. Importantly, this mechanism was also shown to be operative in a Pseudomonas aeruginosa lung infection murine model, and was NE-dependent, because CFTR integrity was significantly protected in NE(-/-) mice compared with WT mice. CONCLUSIONS: These data provide a new mechanism and show for the first time a link between NE-calpains activation and CFTR loss of function in bacterial lung infections relevant to CF and to other chronic inflammatory lung conditions.


Asunto(s)
Calpaína/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Elastasa de Leucocito/fisiología , Animales , Calpaína/metabolismo , Canales de Cloruro/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Epitelio/fisiología , Humanos , Elastasa de Leucocito/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Neumonía Bacteriana/etiología , Neumonía Bacteriana/fisiopatología , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/fisiopatología
13.
Int Immunopharmacol ; 128: 111557, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38266451

RESUMEN

BACKGROUND AND PURPOSE: Lung macrophages (LMs) are critically involved in respiratory diseases. The primary objective of the present study was to determine whether or not an adenosine analog (NECA) and prostaglandin E2 (PGE2) affected the interleukin (IL)-4- and IL-13-induced release of M2a chemokines (CCL13, CCL17, CCL18, and CCL22) by human LMs. EXPERIMENTAL APPROACH: Primary macrophages isolated from resected human lungs were incubated with NECA, PGE2, roflumilast, or vehicle and stimulated with IL-4 or IL-13 for 24 h. The levels of chemokines and PGE2 in the culture supernatants were measured using ELISAs and enzyme immunoassays. KEY RESULTS: Exposure to IL-4 (10 ng/mL) and IL-13 (50 ng/mL) was associated with greater M2a chemokine production but not PGE2 production. PGE2 (10 ng/mL) and NECA (10-6 M) induced the production of M2a chemokines to a lesser extent but significantly enhanced the IL-4/IL-13-induced production of these chemokines. At either a clinically relevant concentration (10-9 M) or at a concentration (10-7 M) that fully inhibited phosphodiesterase 4 (PDE4) activity, roflumilast did not increase the production of M2a chemokines and did not modulate their IL-13-induced production, regardless of the presence or absence of PGE2. CONCLUSIONS: NECA and PGE2 enhanced the IL-4/IL-13-induced production of M2a chemokines. The inhibition of PDE4 by roflumilast did not alter the production of these chemokines. These results contrast totally with the previously reported inhibitory effects of NECA, PGE2, and PDE4 inhibitors on the lipopolysaccharide-induced release of tumor necrosis factor alpha and M1 chemokines in human LMs.


Asunto(s)
Adenosina , Aminopiridinas , Benzamidas , Dinoprostona , Humanos , Dinoprostona/farmacología , Adenosina/farmacología , Interleucina-4/farmacología , Interleucina-13/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Quimiocinas , Macrófagos , Factor de Necrosis Tumoral alfa/farmacología , Quimiocina CCL17 , Pulmón , Células Cultivadas , Ciclopropanos
14.
PLoS Pathog ; 7(8): e1002202, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21901097

RESUMEN

Airway inflammation plays a major role in the pathogenesis of influenza viruses and can lead to a fatal outcome. One of the challenging objectives in the field of influenza research is the identification of the molecular bases associated to the immunopathological disorders developed during infection. While its precise function in the virus cycle is still unclear, the viral protein PB1-F2 is proposed to exert a deleterious activity within the infected host. Using an engineered recombinant virus unable to express PB1-F2 and its wild-type homolog, we analyzed and compared the pathogenicity and host response developed by the two viruses in a mouse model. We confirmed that the deletion of PB1-F2 renders the virus less virulent. The global transcriptomic analyses of the infected lungs revealed a potent impact of PB1-F2 on the response developed by the host. Thus, after two days post-infection, PB1-F2 invalidation severely decreased the number of genes activated by the host. PB1-F2 expression induced an increase in the number and level of expression of activated genes linked to cell death, inflammatory response and neutrophil chemotaxis. When generating interactive gene networks specific to PB1-F2, we identified IFN-γ as a central regulator of PB1-F2-regulated genes. The enhanced cell death of airway-recruited leukocytes was evidenced using an apoptosis assay, confirming the pro-apoptotic properties of PB1-F2. Using a NF-kB luciferase adenoviral vector, we were able to quantify in vivo the implication of NF-kB in the inflammation mediated by the influenza virus infection; we found that PB1-F2 expression intensifies the NF-kB activity. Finally, we quantified the neutrophil recruitment within the airways, and showed that this type of leukocyte is more abundant during the infection of the wild-type virus. Collectively, these data demonstrate that PB1-F2 strongly influences the early host response during IAV infection and provides new insights into the mechanisms by which PB1-F2 mediates virulence.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/inmunología , Transcriptoma , Proteínas Virales/metabolismo , Animales , Apoptosis , Muerte Celular , Quimiotaxis , Femenino , Eliminación de Gen , Regulación Viral de la Expresión Génica , Ingeniería Genética , Interferón beta/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Neutrófilos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Viral/genética , Transcripción Genética , Proteínas Virales/genética , Virulencia
15.
Virologie (Montrouge) ; 17(5): 369-378, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31910592

RESUMEN

Influenza is a very common and contagious respiratory disease where viral virulence can lead to the hospitalization of the individual. The emergence of new viruses and pandemics risks increase the need to improve our knowledge on the interaction of the host with the influenza virus (IAV). In particular, it is necessary to understand how the immune system is activated when it encounters the pathogen and how it controls influenza infection. Innate immune cells play a critical role in controlling the infection by influenza virus. The objective of this review is to provide an overview of the cellular partners of the immune system that are involved in the recognition of IAV, and particularly on the role of Toll-like receptors (TLR) in the initiation of anti-influenza immune responses. We will also discuss the role of different lysosomal proteases in the activation and maturation of TLRs during influenza infection.

16.
Nutrients ; 15(2)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36678133

RESUMEN

Microbiota studies have dramatically increased over these last two decades, and the repertoire of microorganisms with potential health benefits has been considerably enlarged. The development of next generation probiotics from new bacterial candidates is a long-term strategy that may be more efficient and rapid with discriminative in vitro tests. Streptococcus strains have received attention regarding their antimicrobial potential against pathogens of the upper and, more recently, the lower respiratory tracts. Pathogenic bacterial strains, such as non-typable Haemophilus influenzae (NTHi), Pseudomonas aeruginosa (P. aeruginosa) and Staphylococcus aureus (S. aureus), are commonly associated with acute and chronic respiratory diseases, and it could be interesting to fight against pathogens with probiotics. In this study, we show that a Streptococcus mitis (S. mitis) EM-371 strain, isolated from the buccal cavity of a human newborn and previously selected for promising anti-inflammatory effects, displayed in vitro antimicrobial activity against NTHi, P. aeruginosa or S. aureus. However, the anti-pathogenic in vitro activity was not sufficient to predict an efficient protective effect in a preclinical model. Two weeks of treatment with S. mitis EM-371 did not protect against, and even exacerbated, NTHi lung infection.


Asunto(s)
Neumonía , Infecciones del Sistema Respiratorio , Infecciones Estafilocócicas , Recién Nacido , Humanos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología , Staphylococcus aureus , Streptococcus mitis , Bacterias , Haemophilus influenzae , Antibacterianos/farmacología , Pulmón
17.
Front Immunol ; 14: 1142228, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37465668

RESUMEN

In response to the increasing demand for lung transplantation, ex vivo lung perfusion (EVLP) has extended the number of suitable donor lungs by rehabilitating marginal organs. However despite an expanding use in clinical practice, the responses of the different lung cell types to EVLP are not known. In order to advance our mechanistic understanding and establish a refine tool for improvement of EVLP, we conducted a pioneer study involving single cell RNA-seq on human lungs declined for transplantation. Functional enrichment analyses were performed upon integration of data sets generated at 4 h (clinical duration) and 10 h (prolonged duration) from two human lungs processed to EVLP. Pathways related to inflammation were predicted activated in epithelial and blood endothelial cells, in monocyte-derived macrophages and temporally at 4 h in alveolar macrophages. Pathways related to cytoskeleton signaling/organization were predicted reduced in most cell types mainly at 10 h. We identified a division of labor between cell types for the selected expression of cytokine and chemokine genes that varied according to time. Immune cells including CD4+ and CD8+ T cells, NK cells, mast cells and conventional dendritic cells displayed gene expression patterns indicating blunted activation, already at 4 h in several instances and further more at 10 h. Therefore despite inducing inflammatory responses, EVLP appears to dampen the activation of major lung immune cell types, what may be beneficial to the outcome of transplantation. Our results also support that therapeutics approaches aiming at reducing inflammation upon EVLP should target both the alveolar and vascular compartments.


Asunto(s)
Linfocitos T CD8-positivos , Trasplante de Pulmón , Humanos , Perfusión/métodos , Células Endoteliales , Trasplante de Pulmón/métodos , Pulmón/fisiología , Inflamación
18.
Am J Respir Cell Mol Biol ; 47(2): 149-57, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22383584

RESUMEN

Mucins, the main glycoproteins present within mucus, modulate the rheologic properties of airways and participate in lung defense. They are thought to be able to trap and eliminate microorganisms from the lung. Among the mucins secreted in the lung, MUC5AC is the most prominent factor secreted by surface epithelial cells. Although much is known about the signaling pathways involved in the regulation of MUC5AC by host factors such as cytokines or proteases, less is known about the pathways triggered by microorganisms and, specifically, by influenza A virus (IAV). We therefore set up experiments to dissect the molecular mechanisms responsible for the potential modulation of MUC5AC by IAV. Using epithelial cells, C57/Bl6 mice, and IAV strains, we measured MUC5AC expression at the RNA and protein levels, specificity protein 1 (Sp1) activation, and protease activity. Intermediate molecular partners were confirmed using pharmacological inhibitors, blocking antibodies, and small interfering (si)RNAs. We showed in vitro and in vivo that IAV up-regulates epithelial cell-derived MUC5AC and Muc5ac expression in mice, both at transcriptional (through the induction of Sp1) and translational levels. In addition, we determined that this induction was dependent on a protease-epithelial growth factor receptor-extracellular regulated kinase-Sp1 signaling cascade, involving in particular the human airway trypsin. Our data point to MUC5AC as a potential modulatory mechanism by which the lung epithelia respond to IAV infection, and we dissect, for the first time to the best of our knowledge, the molecular partners involved. Future experiments using MUC5AC-targeted strategies should help further unravel the pathophysiological consequences of IAV-induced MUC5AC expression for lung homeostasis.


Asunto(s)
Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Virus de la Influenza A/metabolismo , Pulmón/metabolismo , Mucina 5AC/biosíntesis , Péptido Hidrolasas/genética , Factor de Transcripción Sp1/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/virología , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Gripe Humana/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mucina 5AC/genética , Mucina 5AC/metabolismo , Péptido Hidrolasas/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/genética , Tripsina/genética , Tripsina/metabolismo , Regulación hacia Arriba , Replicación Viral/genética
19.
Gastroenterology ; 140(4): 1272-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21199654

RESUMEN

BACKGROUND & AIMS: Colonic tissues of patients with inflammatory bowel disease have been reported to have increased proteolytic activity, but no studies have clearly addressed the role of the balance between proteases and antiproteases in the pathogenesis of colitis. We investigated the role of Elafin, a serine protease inhibitor expressed by skin and mucosal surfaces in human inflammatory conditions, and the proteases neutrophil elastase (NE) and proteinase-3 (PR-3) in mice with colitis. METHODS: We studied mice with heterozygous disruptions in NE and PR-3, mice that express human elafin (an inhibitor of NE and PR-3), and naïve mice that received intracolonic adenoviral vectors that express elafin. Trinitrobenzene sulfonic acid (TNBS) or dextran sodium sulphate (DSS) was used to induce colitis. Protease, cytokine levels, and NF-κB activity were measured in colons of mice. Caco-2 and HT29 cells were studied in assays for cytokine expression, permeability, and NF-κB activity. RESULTS: Elafin expression or delivery re-equilibrated the proteolytic balance in inflamed colons of mice. In mice given TNBS or DSS, transgenic expression of elafin or disruption of NE and PR-3 protected against the development of colitis. Similarly, adenoviral delivery of Elafin significantly inhibited inflammatory parameters. Elafin modulated a variety of inflammatory mediators in vitro and in vivo and strengthened intestinal epithelial barrier functions. CONCLUSIONS: The protease inhibitor Elafin prevents intestinal inflammation in mouse models of colitis and might be developed as a therapeutic agent for inflammatory bowel disease.


Asunto(s)
Colitis , Elafina/genética , Terapia Genética/métodos , Elastasa de Leucocito/metabolismo , Inhibidores de Proteasas/metabolismo , Adenoviridae/genética , Animales , Células CACO-2 , Quimiocinas/metabolismo , Colitis/genética , Colitis/metabolismo , Colitis/terapia , Citocinas/metabolismo , Elafina/metabolismo , Expresión Génica/fisiología , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Mieloblastina/metabolismo , FN-kappa B/metabolismo , Neutrófilos/enzimología , Neutrófilos/inmunología , Inhibidores de Serina Proteinasa/metabolismo
20.
Viruses ; 13(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525646

RESUMEN

BACKGROUND: Respiratory Syncytial Virus (RSV) is the major cause of severe acute respiratory tract illness in young children worldwide and a main pathogen for the elderly and immune-compromised people. In the absence of vaccines or effective treatments, a better characterization of the pathogenesis of RSV infection is required. To date, the pathophysiology of the disease and its diagnosis has mostly relied on chest X-ray and genome detection in nasopharyngeal swabs. The development of new imaging approaches is instrumental to further the description of RSV spread, virus-host interactions and related acute respiratory disease, at the level of the entire lung. METHODS: By combining tissue clearing, 3D microscopy and image processing, we developed a novel visualization tool of RSV infection in undissected mouse lungs. RESULTS: Whole tissue analysis allowed the identification of infected cell subtypes, based on both morphological traits and position within the cellular network. Furthermore, 3D imaging was also valuable to detect the cytoplasmic viral factories, also called inclusion bodies, a hallmark of RSV infection. CONCLUSIONS: Whole lung clearing and 3D deep imaging represents an unprecedented visualization method of infected lungs to allow insight into RSV pathophysiology and improve the 2D histology analyses.


Asunto(s)
Imagenología Tridimensional , Pulmón/patología , Pulmón/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/virología , Cuerpos de Inclusión Viral/patología , Ratones , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA