Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur Respir J ; 59(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34289975

RESUMEN

BACKGROUND: Benralizumab is a humanised, anti-interleukin-5 receptor α monoclonal antibody with anti-eosinophilic activity. Lack of fucose (afucosylation) increases its affinity to CD16a and significantly enhances antibody-dependent cell-mediated cytotoxicity by natural killer (NK) cells. Although benralizumab proved clinically efficacious in clinical trials for patients with severe asthma and hypereosinophilic syndrome, in-depth characterisation of its anti-eosinophilic mechanisms of action remains elusive. METHODS: Here, we further investigated the mechanisms involved in benralizumab's anti-eosinophilic activities by employing relevant primary human autologous cell co-cultures and real-time-lapse imaging combined with flow cytometry. RESULTS: In the presence of NK cells, benralizumab induced potent eosinophil apoptosis as demonstrated by the upstream induction of Caspase-3/7 and upregulation of cytochrome c. In addition, we uncovered a previously unrecognised mechanism whereby benralizumab can induce eosinophil phagocytosis/efferocytosis by macrophages, a process called antibody-dependent cellular phagocytosis. Using live cell imaging, we unravelled the stepwise processes leading to eosinophil apoptosis and uptake by activated macrophages. Through careful observations of cellular co-culture assays, we identified a novel role for macrophage-derived tumour necrosis factor (TNF) to further enhance benralizumab-mediated eosinophil apoptosis through activation of TNF receptor 1 on eosinophils. TNF-induced eosinophil apoptosis was associated with cytochrome c upregulation, mitochondrial membrane depolarisation and increased Caspase-3/7 activity. Moreover, activated NK cells were found to amplify this axis through the secretion of interferon-γ, subsequently driving TNF expression by macrophages. CONCLUSIONS: Our data provide deeper insights into the timely appearance of events leading to benralizumab-induced eosinophil apoptosis and suggest that additional mechanisms may contribute to the potent anti-eosinophilic activity of benralizumab in vivo. Importantly, afucosylation of benralizumab strongly enhanced its potency for all mechanisms investigated.


Asunto(s)
Antiasmáticos , Asma , Antiasmáticos/farmacología , Antiasmáticos/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Eosinófilos , Humanos
2.
Nat Commun ; 11(1): 4786, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32963227

RESUMEN

Evidence points to an indispensable function of macrophages in tissue regeneration, yet the underlying molecular mechanisms remain elusive. Here we demonstrate a protective function for the IL-33-ST2 axis in bronchial epithelial repair, and implicate ST2 in myeloid cell differentiation. ST2 deficiency in mice leads to reduced lung myeloid cell infiltration, abnormal alternatively activated macrophage (AAM) function, and impaired epithelial repair post naphthalene-induced injury. Reconstitution of wild type (WT) AAMs to ST2-deficient mice completely restores bronchial re-epithelialization. Central to this mechanism is the direct effect of IL-33-ST2 signaling on monocyte/macrophage differentiation, self-renewal and repairing ability, as evidenced by the downregulation of key pathways regulating myeloid cell cycle, maturation and regenerative function of the epithelial niche in ST2-/- mice. Thus, the IL-33-ST2 axis controls epithelial niche regeneration by activating a large multi-cellular circuit, including monocyte differentiation into competent repairing AAMs, as well as group-2 innate lymphoid cell (ILC2)-mediated AAM activation.


Asunto(s)
Bronquiolos/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Interleucina-33/farmacología , Animales , Bronquiolos/lesiones , Bronquiolos/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Proteína 1 Similar al Receptor de Interleucina-1/genética , Pulmón/patología , Activación de Linfocitos , Linfocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
3.
PLoS Pathog ; 13(4): e1006309, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28384349

RESUMEN

Early responses mounted by both tissue-resident and recruited innate immune cells are essential for host defense against bacterial pathogens. In particular, both neutrophils and Ly6Chi monocytes are rapidly recruited to sites of infection. While neutrophils and monocytes produce bactericidal molecules, such as reactive nitrogen and oxygen species, both cell types are also capable of synthesizing overlapping sets of cytokines important for host defense. Whether neutrophils and monocytes perform redundant or non-redundant functions in the generation of anti-microbial cytokine responses remains elusive. Here, we sought to define the contributions of neutrophils and Ly6Chi monocytes to cytokine production and host defense during pulmonary infection with Legionella pneumophila, responsible for the severe pneumonia Legionnaires' disease. We found that both neutrophils and monocytes are critical for host defense against L. pneumophila. Both monocytes and neutrophils contribute to maximal IL-12 and IFNγ responses, and monocytes are also required for TNF production. Moreover, natural killer (NK) cells, NKT cells, and γδ T cells are sources of IFNγ, and monocytes direct IFNγ production by these cell types. Thus, neutrophils and monocytes cooperate in eliciting an optimal cytokine response that promotes effective control of bacterial infection.


Asunto(s)
Antígenos Ly/inmunología , Citocinas/inmunología , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/inmunología , Pulmón/microbiología , Monocitos/inmunología , Neutrófilos/inmunología , Animales , Antígenos Ly/genética , Citocinas/genética , Humanos , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/prevención & control , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL
6.
Nat Immunol ; 17(6): 646-55, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111142

RESUMEN

Group 2 innate lymphoid cells (ILC2 cells) are important for type 2 immune responses and are activated by the epithelial cytokines interleukin 33 (IL-33), IL-25 and thymic stromal lymphopoietin (TSLP). Here we demonstrated that IL-1ß was a critical activator of ILC2 cells, inducing proliferation and cytokine production and regulating the expression of epithelial cytokine receptors. IL-1ß also governed ILC2 plasticity by inducing low expression of the transcription factor T-bet and the cytokine receptor chain IL-12Rß2, which enabled the conversion of these cells into an ILC1 phenotype in response to IL-12. This transition was marked by an atypical chromatin landscape characterized by the simultaneous transcriptional accessibility of the locus encoding interferon-γ (IFN-γ) and the loci encoding IL-5 and IL-13. Finally, IL-1ß potentiated ILC2 activation and plasticity in vivo, and IL-12 acted as the switch that determined an ILC2-versus-ILC1 response. Thus, we have identified a previously unknown role for IL-1ß in facilitating ILC2 maturation and plasticity.


Asunto(s)
Plasticidad de la Célula , Inmunidad Innata , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Linfocitos/inmunología , Animales , Diferenciación Celular , Plasticidad de la Célula/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Ratones , Ratones SCID , Receptores de Interleucina-12/genética , Receptores de Interleucina-12/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Balance Th1 - Th2 , Células Th2/inmunología , Linfopoyetina del Estroma Tímico
7.
Nat Immunol ; 17(6): 626-35, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111143

RESUMEN

Innate lymphoid cells (ILCs) are critical mediators of mucosal immunity, and group 1 ILCs (ILC1 cells) and group 3 ILCs (ILC3 cells) have been shown to be functionally plastic. Here we found that group 2 ILCs (ILC2 cells) also exhibited phenotypic plasticity in response to infectious or noxious agents, characterized by substantially lower expression of the transcription factor GATA-3 and a concomitant switch to being ILC1 cells that produced interferon-γ (IFN-γ). Interleukin 12 (IL-12) and IL-18 regulated this conversion, and during viral infection, ILC2 cells clustered within inflamed areas and acquired an ILC1-like phenotype. Mechanistically, these ILC1 cells augmented virus-induced inflammation in a manner dependent on the transcription factor T-bet. Notably, IL-12 converted human ILC2 cells into ILC1 cells, and the frequency of ILC1 cells in patients with chronic obstructive pulmonary disease (COPD) correlated with disease severity and susceptibility to exacerbations. Thus, functional plasticity of ILC2 cells exacerbates anti-viral immunity, which may have adverse consequences in respiratory diseases such as COPD.


Asunto(s)
Infecciones por Haemophilus/inmunología , Haemophilus influenzae/inmunología , Virus de la Influenza A/inmunología , Pulmón/inmunología , Linfocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Células TH1/inmunología , Células Th2/inmunología , Anciano , Animales , Diferenciación Celular , Plasticidad de la Célula/inmunología , Células Cultivadas , Citocinas/metabolismo , Femenino , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fenotipo , Fumar/efectos adversos , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
8.
Proc Natl Acad Sci U S A ; 112(24): 7557-62, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26034289

RESUMEN

The innate immune system is critical for host defense against microbial pathogens, yet many pathogens express virulence factors that impair immune function. Here, we used the bacterial pathogen Legionella pneumophila to understand how the immune system successfully overcomes pathogen subversion mechanisms. L. pneumophila replicates within macrophages by using a type IV secretion system to translocate bacterial effectors into the host cell cytosol. As a consequence of effector delivery, host protein synthesis is blocked at several steps, including translation initiation and elongation. Despite this translation block, infected cells robustly produce proinflammatory cytokines, but the basis for this is poorly understood. By using a reporter system that specifically discriminates between infected and uninfected cells within a population, we demonstrate here that infected macrophages produced IL-1α and IL-1ß, but were poor producers of IL-6, TNF, and IL-12, which are critical mediators of host protection. Uninfected bystander cells robustly produced IL-6, TNF, and IL-12, and this bystander response required IL-1 receptor (IL-1R) signaling during early pulmonary infection. Our data demonstrate functional heterogeneity in production of critical protective cytokines and suggest that collaboration between infected and uninfected cells enables the immune system to bypass pathogen-mediated translation inhibition to generate an effective immune response.


Asunto(s)
Receptores Tipo I de Interleucina-1/metabolismo , Animales , Antígeno B7-2/biosíntesis , Citocinas/biosíntesis , Femenino , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interleucina-1/metabolismo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Virulencia/inmunología
9.
Proc Natl Acad Sci U S A ; 112(21): 6688-93, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25964352

RESUMEN

Inflammasomes are critical for host defense against bacterial pathogens. In murine macrophages infected by gram-negative bacteria, the canonical inflammasome activates caspase-1 to mediate pyroptotic cell death and release of IL-1 family cytokines. Additionally, a noncanonical inflammasome controlled by caspase-11 induces cell death and IL-1 release. However, humans do not encode caspase-11. Instead, humans encode two putative orthologs: caspase-4 and caspase-5. Whether either ortholog functions similar to caspase-11 is poorly defined. Therefore, we sought to define the inflammatory caspases in primary human macrophages that regulate inflammasome responses to gram-negative bacteria. We find that human macrophages activate inflammasomes specifically in response to diverse gram-negative bacterial pathogens that introduce bacterial products into the host cytosol using specialized secretion systems. In primary human macrophages, IL-1ß secretion requires the caspase-1 inflammasome, whereas IL-1α release and cell death are caspase-1-independent. Instead, caspase-4 mediates IL-1α release and cell death. Our findings implicate human caspase-4 as a critical regulator of noncanonical inflammasome activation that initiates defense against bacterial pathogens in primary human macrophages.


Asunto(s)
Caspasas Iniciadoras/inmunología , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/patogenicidad , Inflamasomas/inmunología , Animales , Caspasa 1/inmunología , Muerte Celular , Células Cultivadas , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Lipopolisacáridos/toxicidad , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad
10.
Infect Immun ; 82(10): 4325-36, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092908

RESUMEN

Legionella pneumophila, an intracellular pathogen responsible for the severe pneumonia Legionnaires' disease, uses its dot/icm-encoded type IV secretion system (T4SS) to translocate effector proteins that promote its survival and replication into the host cell cytosol. However, by introducing bacterial products into the host cytosol, L. pneumophila also activates cytosolic immunosurveillance pathways, thereby triggering robust proinflammatory responses that mediate the control of infection. Thus, the pulmonary cell types that L. pneumophila infects not only may act as an intracellular niche that facilitates its pathogenesis but also may contribute to the immune response against L. pneumophila. The identity of these host cells remains poorly understood. Here, we developed a strain of L. pneumophila producing a fusion protein consisting of ß-lactamase fused to the T4SS-translocated effector RalF, which allowed us to track cells injected by the T4SS. Our data reveal that alveolar macrophages and neutrophils both are the primary recipients of T4SS-translocated effectors and harbor viable L. pneumophila during pulmonary infection of mice. Moreover, both alveolar macrophages and neutrophils from infected mice produced tumor necrosis factor and interleukin-1α in response to T4SS-sufficient, but not T4SS-deficient, L. pneumophila. Collectively, our data suggest that alveolar macrophages and neutrophils are both an intracellular reservoir for L. pneumophila and a source of proinflammatory cytokines that contribute to the host immune response against L. pneumophila during pulmonary infection.


Asunto(s)
Sistemas de Secreción Bacterianos , Legionella pneumophila/inmunología , Legionella pneumophila/fisiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Neutrófilos/inmunología , Neutrófilos/microbiología , Animales , Citosol/metabolismo , Citosol/microbiología , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Interleucina-1alfa/metabolismo , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/microbiología , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(20): 7385-90, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24799700

RESUMEN

Toll-like receptor signaling and subsequent activation of NF-κB- and MAPK-dependent genes during infection play an important role in antimicrobial host defense. The YopJ protein of pathogenic Yersinia species inhibits NF-κB and MAPK signaling, resulting in blockade of NF-κB-dependent cytokine production and target cell death. Nevertheless, Yersinia infection induces inflammatory responses in vivo. Moreover, increasing the extent of YopJ-dependent cytotoxicity induced by Yersinia pestis and Yersinia pseudotuberculosis paradoxically leads to decreased virulence in vivo, suggesting that cell death promotes anti-Yersinia host defense. However, the specific pathways responsible for YopJ-induced cell death and how this cell death mediates immune defense against Yersinia remain poorly defined. YopJ activity induces processing of multiple caspases, including caspase-1, independently of inflammasome components or the adaptor protein ASC. Unexpectedly, caspase-1 activation in response to the activity of YopJ required caspase-8, receptor-interacting serine/threonine kinase 1 (RIPK1), and Fas-associated death domain (FADD), but not RIPK3. Furthermore, whereas RIPK3 deficiency did not affect YopJ-induced cell death or caspase-1 activation, deficiency of both RIPK3 and caspase-8 or FADD completely abrogated Yersinia-induced cell death and caspase-1 activation. Mice lacking RIPK3 and caspase-8 in their hematopoietic compartment showed extreme susceptibility to Yersinia and were deficient in monocyte and neutrophil-derived production of proinflammatory cytokines. Our data demonstrate for the first time to our knowledge that RIPK1, FADD, and caspase-8 are required for YopJ-induced cell death and caspase-1 activation and suggest that caspase-8-mediated cell death overrides blockade of immune signaling by YopJ to promote anti-Yersinia immune defense.


Asunto(s)
Caspasa 1/metabolismo , Caspasa 8/metabolismo , Inmunidad Innata , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Animales , Apoptosis , Proteínas Bacterianas/genética , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Regulación Enzimológica de la Expresión Génica , Ratones , Ratones Transgénicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Yersiniosis/microbiología , Yersinia pseudotuberculosis
12.
PLoS Pathog ; 9(6): e1003400, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762026

RESUMEN

Inflammasome activation is important for antimicrobial defense because it induces cell death and regulates the secretion of IL-1 family cytokines, which play a critical role in inflammatory responses. The inflammasome activates caspase-1 to process and secrete IL-1ß. However, the mechanisms governing IL-1α release are less clear. Recently, a non-canonical inflammasome was described that activates caspase-11 and mediates pyroptosis and release of IL-1α and IL-1ß. Caspase-11 activation in response to Gram-negative bacteria requires Toll-like receptor 4 (TLR4) and TIR-domain-containing adaptor-inducing interferon-ß (TRIF)-dependent interferon production. Whether additional bacterial signals trigger caspase-11 activation is unknown. Many bacterial pathogens use specialized secretion systems to translocate effector proteins into the cytosol of host cells. These secretion systems can also deliver flagellin into the cytosol, which triggers caspase-1 activation and pyroptosis. However, even in the absence of flagellin, these secretion systems induce inflammasome activation and the release of IL-1α and IL-1ß, but the inflammasome pathways that mediate this response are unclear. We observe rapid IL-1α and IL-1ß release and cell death in response to the type IV or type III secretion systems of Legionella pneumophila and Yersinia pseudotuberculosis. Unlike IL-1ß, IL-1α secretion does not require caspase-1. Instead, caspase-11 activation is required for both IL-1α secretion and cell death in response to the activity of these secretion systems. Interestingly, whereas caspase-11 promotes IL-1ß release in response to the type IV secretion system through the NLRP3/ASC inflammasome, caspase-11-dependent release of IL-1α is independent of both the NAIP5/NLRC4 and NLRP3/ASC inflammasomes as well as TRIF and type I interferon signaling. Furthermore, we find both overlapping and non-redundant roles for IL-1α and IL-1ß in mediating neutrophil recruitment and bacterial clearance in response to pulmonary infection by L. pneumophila. Our findings demonstrate that virulent, but not avirulent, bacteria trigger a rapid caspase-11-dependent innate immune response important for host defense.


Asunto(s)
Sistemas de Secreción Bacterianos/inmunología , Caspasas/inmunología , Citosol/inmunología , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Macrófagos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas Iniciadoras , Línea Celular , Citosol/microbiología , Activación Enzimática/inmunología , Inmunidad Innata/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...