Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 208(5): 1272-1279, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35110420

RESUMEN

Interstitial macrophages (IMs) are key regulators of allergic inflammation. We previously showed that the absence of semaphorin 3E (Sema3E) exacerbates asthma features in both acute and chronic asthma models. However, it has not been studied whether Sema3E, via its receptor plexinD1, regulates IM function in allergic asthma. Therefore, we investigated the role of plexinD1 deficiency on IMs in allergic asthma. We found that the absence of plexinD1 in IMs increased airway hyperresponsiveness, airway leukocyte numbers, allergen-specific IgE, goblet cell hyperplasia, and Th2/Th17 cytokine response in the house dust mite (HDM)-induced allergic asthma model. Muc5ac, Muc5b, and α-SMA genes were increased in mice with Plxnd1-deficient IMs compared with wild-type mice. Furthermore, plexinD1-deficient bone marrow-derived macrophages displayed reduced IL-10 mRNA expression, at both the baseline and following HDM challenge, compared with their wild-type counterpart mice. Our data suggest that Sema3E/plexinD1 signaling in IMs is a critical pathway that modulates airway inflammation, airway resistance, and tissue remodeling in the HDM murine model of allergic asthma. Reduced IL-10 expression by plexinD1-deficient macrophages may account for these enhanced allergic asthma features.


Asunto(s)
Asma/patología , Dermatophagoides pteronyssinus/inmunología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Semaforinas/genética , Actinas/genética , Actinas/metabolismo , Resistencia de las Vías Respiratorias/inmunología , Animales , Asma/inmunología , Modelos Animales de Enfermedad , Femenino , Células Caliciformes/inmunología , Inmunoglobulina E/inmunología , Interleucina-10/genética , Recuento de Leucocitos , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Noqueados , Mucina 5AC/genética , Mucina 5AC/metabolismo , Mucina 5B/genética , Mucina 5B/metabolismo , ARN Mensajero/genética , Células Th17/inmunología , Células Th2/inmunología
2.
J Immunol ; 206(6): 1251-1265, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33504621

RESUMEN

Recent studies have identified semaphorin 3E (Sema3E) as a novel mediator of immune responses. However, its function in immunity to infection has yet to be investigated. Using a mouse model of chlamydial lung infection, we show that Sema3E plays a significant role in the host immune response to the infection. We found that Sema3E is induced in the lung after chlamydial infection, and Sema3E deficiency has a detrimental impact on disease course, dendritic cell (DC) function, and T cell responses. Specifically, we found that Sema3E knockout (KO) mice exhibited higher bacterial burden, severe body weight loss, and pathological changes after Chlamydia muridarum lung infection compared with wild-type (WT) mice. The severity of disease in Sema3E KO mice was correlated with reduced Th1/Th17 cytokine responses, increased Th2 response, altered Ab response, and a higher number of regulatory CD4 T cells. Moreover, DCs isolated from Sema3E KO mice showed lower surface expression of costimulatory molecules and production of IL-12, but higher expression of PD-L1, PD-L2, and IL-10 production. Functional DC-T cell coculture studies revealed that DCs from infected Sema3E KO mice failed to induce Th1 and Th17 cell responses compared with DCs from infected WT mice. Upon adoptive transfer, mice receiving DCs from Sema3E KO mice, unlike those receiving DCs from WT mice, were not protected against challenge infection. In conclusion, our data evidenced that Sema3E acts as a critical factor for protective immunity against intracellular bacterial infection by modulating DC functions and T cell subsets.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Chlamydia/inmunología , Células Dendríticas/inmunología , Semaforinas/metabolismo , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/metabolismo , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/inmunología , Técnicas de Cocultivo , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Humanos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Noqueados , Semaforinas/genética , Índice de Severidad de la Enfermedad , Subgrupos de Linfocitos T/metabolismo
3.
J Immunol ; 206(3): 588-598, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33443083

RESUMEN

Protective immunity to cutaneous leishmaniasis is mediated by IFN-γ-secreting CD4+ Th1 cells. IFN-γ binds to its receptor on Leishmania-infected macrophages, resulting in their activation, production of NO, and subsequent destruction of parasites. This study investigated the role of Semaphorin 3E (Sema3E) in host immunity to Leishmania major infection in mice. We observed a significant increase in Sema3E expression at the infection site at different timepoints following L. major infection. Sema3E-deficient (Sema3E knockout [KO]) mice were highly resistant to L. major infection, as evidenced by significantly (p < 0.05-0.01) reduced lesion sizes and lower parasite burdens at different times postinfection when compared with their infected wild-type counterpart mice. The enhanced resistance of Sema3E KO mice was associated with significantly (p < 0.05) increased IFN-γ production by CD4+ T cells. CD11c+ cells from Sema3E KO mice displayed increased expression of costimulatory molecules and IL-12p40 production following L. major infection and were more efficient at inducing the differentiation of Leishmania-specific CD4+ T cells to Th1 cells than their wild-type counterpart cells. Furthermore, purified CD4+ T cells from Sema3E KO mice showed increased propensity to differentiate into Th1 cells in vitro, and this was significantly inhibited by the addition of recombinant Sema3E in vitro. These findings collectively show that Sema3E is a negative regulator of protective CD4+ Th1 immunity in mice infected with L. major and suggest that its neutralization may be a potential therapeutic option for treating individuals suffering from cutaneous leishmaniasis.


Asunto(s)
Leishmania major/inmunología , Leishmaniasis Cutánea/metabolismo , Semaforinas/metabolismo , Células TH1/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Tolerancia Inmunológica , Leishmaniasis Cutánea/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Semaforinas/genética
4.
J Immunol ; 204(1): 128-136, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31776203

RESUMEN

Semaphorin 3E (Sema3E) is a secreted protein that was initially discovered as a neuronal guidance cue. Recent evidence showed that Sema3E plays an essential role in regulating the activities of various immune cells. However, the exact role of Sema3E in macrophage function, particularly during inflammation, is not fully understood. We studied the impact of Sema3E gene deletion on macrophage function during the LPS-induced acute inflammatory response. We found that Sema3E-deficient (Sema3e-/- ) mice were better protected from LPS-induced acute inflammation as exemplified by their superior clinical score and effective temperature control compared with their wild-type littermates. This superior control of inflammatory response in Sema3e-/- mice was associated with significantly lower phosphorylation of ERK1/2, AKT, STAT3, and NF-κB, and a concomitant reduction in inducible NO synthase expression and production of TNF and IL-6 compared with their Sema3e+/+ littermates. Sema3e-/- mice also contained significantly higher numbers of activated macrophages compared with their Sema3e+/+ littermates at both baselines and after LPS challenge. In vivo-specific deletion of the Sema3E high-affinity receptor, plexinD1, on macrophages led to the improvement in clinical disease following exposure to a lethal dose of LPS. Collectively, our data show that Sema3E plays an essential role in dampening the early inflammatory response to LPS by regulating macrophage function, suggesting an essential role of this pathway in macrophage inflammatory response.


Asunto(s)
Inflamación/inmunología , Macrófagos/inmunología , Semaforinas/inmunología , Animales , Células Cultivadas , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Noqueados , Ratones Transgénicos , Semaforinas/deficiencia
5.
Am J Pathol ; 189(4): 762-772, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711489

RESUMEN

Increased angiogenesis is a characteristic feature of remodeling in asthmatic airways and stems from the imbalance between pro-angiogenic and anti-angiogenic factors. Surprisingly, the factors regulating this process in allergic asthma are poorly defined. Previously, we showed an important role of semaphorins 3E (Sema3E) in growth factor-induced airway smooth muscle proliferation and migration in vitro, and in down-regulating airway inflammation, T helper 2/T helper 17 cytokine response, mucus cell hyperplasia, and airway hyperresponsiveness in vivo. However, the role of Sema3E in airway angiogenesis is not fully understood. Here, we investigated the role of Sema3E in airway angiogenesis using a house dust mite (HDM) murine model of allergic asthma. Intranasal treatment with recombinant Sema3E significantly reduced the expression of angiogenesis markers within the airways of HDM-challenged mice compared with untreated mice. HDM-induced expression of vascular endothelial growth factor (VEGF) and VEGF receptor 2 protein were diminished substantially on Sema3E treatment. Interestingly, Sema3E-treated mice showed an enhanced expression of the negative regulator of angiogenesis, soluble VEGF receptor 1, compared with the untreated mice. These events were reversed in Sema3E-deficient mice at baseline or on HDM challenge. Taken together, this study provides the first evidence that Sema3E modulates angiogenesis in allergic asthmatic airways via modulating pro- and anti-angiogenic factors.


Asunto(s)
Asma/prevención & control , Proteínas del Citoesqueleto/fisiología , Modelos Animales de Enfermedad , Inflamación/prevención & control , Proteínas de la Membrana/fisiología , Neovascularización Patológica/prevención & control , Pyroglyphidae/patogenicidad , Hipersensibilidad Respiratoria/prevención & control , Remodelación de las Vías Aéreas (Respiratorias) , Alérgenos/inmunología , Inductores de la Angiogénesis/inmunología , Inductores de la Angiogénesis/metabolismo , Animales , Asma/etiología , Asma/patología , Femenino , Inflamación/etiología , Inflamación/patología , Ratones , Ratones Noqueados , Neovascularización Patológica/etiología , Neovascularización Patológica/patología , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/patología , Semaforinas
6.
J Immunol ; 198(5): 1805-1814, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28108561

RESUMEN

Semaphorin 3E (Sema3E) plays a crucial role in axon guidance, vascular patterning, and immune regulation. Nevertheless, the role of Sema3E in asthma is still elusive. In this study, we show that genetic ablation of Sema3E in mice results in increased lung granulocytosis, airway hyperresponsiveness, mucus overproduction, collagen deposition, and Th2/Th17 inflammation. Transfer of Sema3e-/- bone marrow progenitor cells to irradiated wild-type (WT) recipients exacerbates airway hyperresponsiveness and inflammation, whereas transfer of WT bone marrow progenitor cells ameliorates asthma pathology in Sema3e-/- recipients. Sema3e-/- mice display a higher frequency of CD11b+ pulmonary dendritic cells than their WT controls at the baseline and after sensitization with house dust mite. Adoptive transfer of CD11b+ pulmonary dendritic cells from Sema3e-/- mice into WT recipients increases house dust mite-induced Th2/Th17 inflammation in the airway. Together, these findings identify Sema3E as a novel regulatory molecule in allergic asthma that acts upstream of proallergic events and suggest that targeting this molecule could be a novel approach to treat allergic asthma.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Glicoproteínas/deficiencia , Glicoproteínas/fisiología , Inflamación/inmunología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/fisiología , Hipersensibilidad Respiratoria/inmunología , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Proteínas del Citoesqueleto , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glicoproteínas/genética , Pulmón/inmunología , Pulmón/fisiopatología , Proteínas de la Membrana/genética , Ratones , Pyroglyphidae/inmunología , Semaforinas , Células Th17/inmunología , Células Th17/metabolismo
7.
J Immunol ; 198(3): 1023-1033, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27913633

RESUMEN

Neutrophil migration is an essential step in leukocyte trafficking during inflammatory responses. Semaphorins, originally discovered as axon guidance cues in neural development, have been shown to regulate cell migration beyond the nervous system. However, the potential contribution of semaphorins in the regulation of neutrophil migration is not well understood. This study examines the possible role of a secreted chemorepellent, Semaphorin 3E (Sema3E), in neutrophil migration. In this study, we demonstrated that human neutrophils constitutively express Sema3E high-affinity receptor, PlexinD1. Sema3E displayed a potent ability to inhibit CXCL8/IL-8-induced neutrophil migration as determined using a microfluidic device coupled to real-time microscopy and a transwell system in vitro. The antimigratory effect of Sema3E on human neutrophil migration was associated with suppression of CXCL8/IL-8-mediated Ras-related C3 botulinum toxin substrate 1 GTPase activity and actin polymerization. We further addressed the regulatory role of Sema3E in the regulation of neutrophil migration in vivo. Allergen airway exposure induced higher neutrophil recruitment into the lungs of Sema3e-/- mice compared with wild-type controls. Administration of exogenous recombinant Sema3E markedly reduced allergen-induced neutrophil recruitment into the lungs, which was associated with alleviation of allergic airway inflammation and improvement of lung function. Our data suggest that Sema3E could be considered an essential regulatory mediator involved in modulation of neutrophil migration throughout the course of neutrophilic inflammation.


Asunto(s)
Neutrófilos/fisiología , Semaforinas/fisiología , Actinas/metabolismo , Moléculas de Adhesión Celular Neuronal/análisis , Movimiento Celular , Quimiotaxis de Leucocito , Humanos , Interleucina-8/fisiología , Péptidos y Proteínas de Señalización Intracelular , Dispositivos Laboratorio en un Chip , Glicoproteínas de Membrana , Proteína de Unión al GTP rac1/metabolismo
8.
Am J Pathol ; 187(7): 1566-1576, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28634005

RESUMEN

Semaphorins are an essential family of guidance cues ubiquitously expressed in various organs, which play diverse developmental, homeostatic, and pathological roles. Semaphorin 3E (Sema3E), initially identified as a neuronal chemorepellent, is involved in the regulation of cell migration, proliferation, and angiogenesis. However, expression and function of Sema3E in allergic asthma has not been extensively investigated. We determined the expression of Sema3E in the airways and its effect on airway inflammation, hyperresponsiveness, and remodeling as pathological features of allergic asthma provoked by house dust mite in vivo. Our data indicate that exposure to house dust mite markedly reduces Sema3E expression in mouse airways. More important, replenishment of Sema3E by intranasal administration of exogenous Sema3E protects mice from allergic asthma by reducing eosinophilic inflammation, serum IgE level, and T helper cell 2/T helper cell 17 cytokine response. The regulatory effect of Sema3E on cytokine response was sustained on allergen recall response in the lymph nodes and spleen. Furthermore, goblet cell hyperplasia, collagen deposition, and airway hyperresponsiveness were significantly diminished on Sema3E treatment. The inhibitory effect of Sema3E was associated with a reduction of pulmonary CD11b+ conventional dendritic cells and regulation of CD4+ T-cell cytokine response. Collectively, our data represent a novel approach to treating allergic asthma via regulation of immune response to house dust mite.


Asunto(s)
Asma/prevención & control , Regulación de la Expresión Génica , Glicoproteínas/administración & dosificación , Proteínas de la Membrana/administración & dosificación , Pyroglyphidae/inmunología , Hipersensibilidad Respiratoria/prevención & control , Administración Intranasal , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Alérgenos/inmunología , Animales , Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Citocinas/inmunología , Proteínas del Citoesqueleto , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/genética , Glicoproteínas/metabolismo , Inflamación/inmunología , Inflamación/prevención & control , Pulmón/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes , Hipersensibilidad Respiratoria/inmunología , Semaforinas , Células Th17/inmunología , Células Th2/inmunología
9.
J Allergy Clin Immunol ; 139(3): 950-963.e9, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27567326

RESUMEN

BACKGROUND: Pentraxin 3 (PTX3) is a multifunctional molecule that plays a nonredundant role at the crossroads between pathogen clearance, innate immune system, matrix deposition, female fertility, and vascular biology. It is produced at sites of infection and inflammation by both structural and inflammatory cells. However, its role in allergen-induced inflammation remains to be tested. OBJECTIVE: We sought to determine the effect of Ptx3 deletion on ovalbumin (OVA)-induced allergic inflammation in a murine model of asthma. METHODS: Bronchoalveolar lavage fluid was collected from patients with severe asthma and healthy subjects, and the level of PTX3 was determined by using ELISA. Ptx3+/+ and Ptx3-/- mice were sensitized and challenged with OVA and bronchoalveolar lavage fluid, and the lungs were collected for assessing inflammation. Lung tissue inflammation and mucus production were assessed by means of flow cytometry and hematoxylin and eosin and periodic acid-Schiff staining, respectively. flexiVent was used to determine airway resistance to methacholine in these mice. RESULTS: Here we report that mice with severe asthma and OVA-sensitized/challenged mice had increased PTX3 levels in the lungs compared with healthy control mice. Mice lacking PTX3 have exaggerated neutrophilic/eosinophilic lung inflammation, mucus production, and airway hyperresponsiveness in an experimental model of OVA-induced asthma. Furthermore, OVA-exposed lung Ptx3-/- CD4 T cells exhibit an increased production of IL-17A, an effect that is accompanied by an increased signal transducer and activator of transcription 3 phosphorylation, reduced IL-2 production, and enhanced activation and survival. Also, we observed an increase in numbers of IL-6- and IL-23-producing dendritic cells in OVA-exposed Ptx3-/- mice compared with those in wild-type control mice. CONCLUSION: Altogether, PTX3 deficiency results in augmented airway hyperresponsiveness, mucus production, and IL-17A-dominant pulmonary inflammation, suggesting a regulatory role of PTX3 in the development of allergic inflammation.


Asunto(s)
Asma/inmunología , Proteína C-Reactiva/inmunología , Linfocitos T CD4-Positivos/inmunología , Componente Amiloide P Sérico/inmunología , Adulto , Anciano , Alérgenos , Animales , Asma/metabolismo , Asma/fisiopatología , Líquido del Lavado Bronquioalveolar , Proteína C-Reactiva/genética , Citocinas/inmunología , Femenino , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Moco/metabolismo , Ovalbúmina , Componente Amiloide P Sérico/genética
10.
J Allergy Clin Immunol ; 133(2): 560-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23932461

RESUMEN

BACKGROUND: Chronic airway diseases, including asthma, are characterized by increased airway smooth muscle (ASM) mass that is due in part to growth factor-mediated ASM cell proliferation and migration. However, the molecular mechanisms underlying these effects are not completely understood. Semaphorin 3E (Sema3E) has emerged as an essential mediator involved in cell migration, proliferation, and angiogenesis, although its role in ASM cell function is not investigated. OBJECTIVES: We sought to determine the expression of Sema3E receptor, plexinD1, in human ASM cells (HASMCs); effect of Sema3E on basal and platelet-derived growth factor (PDGF)-induced proliferation and migration; and underlying signaling pathways. METHODS: Expression of plexinD1 in HASMCs was studied with RT-PCR, immunostaining, and flow cytometry. The effect of Sema3E on HASMC proliferation and migration was evaluated by 5-ethynyl-2'-deoxyuridine (EdU) incorporation, cell count, and Boyden chamber assay. Sema3E-mediated intracellular signaling was investigated with fluorescent microscopy, flow cytometry, Rac1 activation, and Western blot analysis. RESULTS: HASMCs from healthy persons expressed plexinD1 more than HASMCs from asthmatic patients. Sema3E increased plexinD1 expression in HASMCs from asthmatic patients. Recombinant Sema3E inhibited PDGF-mediated HASMC proliferation and migration, which was associated with F-actin depolymerization, suppression of PDGF-induced Rac1 guanosine triphosphatase activity, and Akt and extracellular signal-regulated kinase 1 and 2 phosphorylation. Bronchial biopsies from patients with mild asthma displayed immunoreactivity of plexinD1, suggesting the potential in vivo role of Sema3E-PlexinD1 axis in HASMC function. CONCLUSION: This study provides the first evidence that Sema3E receptor is expressed and plays functional roles in HASMCs. Our data suggest a regulatory role of Sema3E in PDGF-mediated proliferation and migration, leading to downregulation of ASM remodeling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/patología , Miocitos del Músculo Liso/fisiología , Semaforinas/fisiología , Adulto , Asma/fisiopatología , Becaplermina , Bronquios/citología , Moléculas de Adhesión Celular Neuronal/fisiología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Glicoproteínas de Membrana , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-sis/farmacología , Tráquea/citología , Adulto Joven
13.
Pharmacol Ther ; 242: 108351, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36706796

RESUMEN

Asthma is a heterogenous airway disease characterized by airway inflammation and remodeling. It affects more than 300 million people worldwide and poses a significant burden on society. Semaphorins, discovered initially as neural guidance molecules, are ubiquitously expressed in various organs and regulate multiple signaling pathways. Interestingly, Semaphorin3E is a critical molecule in lung pathophysiology through its role in both lung development and homeostasis. Semaphorin3E binds to plexinD1, mediating regulatory effects on cell migration, proliferation, and angiogenesis. Recent in vitro and in vivo studies have demonstrated that the Semaphorin3E-plexinD1 axis is implicated in asthma, impacting inflammatory and structural cells associated with airway inflammation, tissue remodeling, and airway hyperresponsiveness. This review details the Semaphorin3E-plexinD1 axis in various aspects of asthma and highlights future directions in research including its potential role as a therapeutic target in airway allergic diseases.


Asunto(s)
Asma , Hipersensibilidad , Humanos , Animales , Sistema Respiratorio/metabolismo , Inflamación/metabolismo , Movimiento Celular , Remodelación de las Vías Aéreas (Respiratorias) , Pulmón/metabolismo , Modelos Animales de Enfermedad
14.
J Immunol ; 184(12): 7134-43, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483734

RESUMEN

Thymic stromal lymphopoietin (TSLP) plays a pivotal role in allergic diseases such as asthma, chronic obstructive pulmonary disease, and atopic dermatitis. Enhanced TSLP expression has been detected in asthmatic airways that correlated with both the expression of Th2-attracting chemokines and with disease severity. Although cumulative evidence suggests that human airway smooth muscle (HASM) cells can initiate or perpetuate the airway inflammation by secreting a variety of inflammatory cell products such as cytokines and chemokines, the role of TSLP in this pathway is not known. In the current study, we sought to investigate whether HASM cells express the TSLP receptor (TSLPR) and whether it is functional. We first demonstrated that primary HASM cells express the transcript and protein of both TSLPR subunits (TSLPR and IL-7Ralpha). Functionally, TSLPR-mediated HASM activation induced a significant increase in CXC (IL-8/CXCL8), CC (eotaxin-1/CCL11) chemokines, and proinflammatory cytokine IL-6 expression. Furthermore, using biochemical and genetic approaches, we found that TSLP-induced proinflammatory gene expression in HASM involved the transcriptional mechanisms, MAPKs (ERK1/2, p38, and JNK), and STAT3 activation. Finally, TSLPR immunoreactivity in bronchial sections from mild allergic asthmatics suggested the potential in vivo TSLP targeting of HASM. Altogether, our data suggest that the TSLPR-mediated HASM activation induces proinflammatory cytokine and chemokines release that may facilitate inflammatory immune cells recruitment in airways. In addition, it may be inferred that TSLPR is involved in the pathogenesis of allergic asthma through the activation of HASM cells by TSLP.


Asunto(s)
Quimiocinas CC/biosíntesis , Quimiocinas CXC/biosíntesis , Interleucina-6/biosíntesis , Miocitos del Músculo Liso/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal/inmunología , Separación Celular , Quimiocinas CC/inmunología , Quimiocinas CXC/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Activación Enzimática/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-6/inmunología , Pulmón/inmunología , Pulmón/metabolismo , MAP Quinasa Quinasa 4/inmunología , MAP Quinasa Quinasa 4/metabolismo , Microscopía Confocal , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos del Músculo Liso/inmunología , Receptores de Citocinas/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Linfopoyetina del Estroma Tímico
15.
J Immunol ; 182(6): 3357-65, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265112

RESUMEN

IL-17A has been shown to be expressed at higher levels in respiratory secretions from asthmatics and to correlate with airway hyperresponsiveness. Although these studies raise the possibility that IL-17A may influence allergic disease, the mechanism remains unknown. We previously demonstrated that IL-17A mediates CC chemokine (CCL11) production from human airway smooth muscle (ASM) cells. In this study, we demonstrate that STAT3 activation is critical in IL-17A-mediated CCL11 expression in ASM cells. IL-17A mediated a rapid phosphorylation of STAT3 but not STAT6 or STAT5 in ASM cells. Interestingly, transient transfection with wild-type or mutated CCL11 promoter constructs showed that IL-17A-mediated CCL11 expression relies on the STAT6 binding site. However, STAT3 but not STAT6 in vivo binding to the CCL11 promoter was detected following IL-17A stimulation of ASM cells. Overexpression of DN STAT3 (STAT3beta) abolishes IL-17A-induced CCL11 promoter activity. This effect was not observed with STAT6 DN or the STAT3 mutant at Ser(727). Interestingly, disruption of STAT3 activity with the SH2 domain binding peptide, but not with control peptide, results in a significant reduction of IL-17A-mediated STAT3 phosphorylation and CCL11 promoter activity. IL-17A-mediated CCL11 promoter activity and mRNA were significantly diminished in STAT3- but not STAT6-silenced ASM cells. Finally, IL-17A-induced STAT3 phosphorylation was sensitive to pharmacological inhibitors of JAK2 and ERK1/2. Taken together, our data provide the first evidence of IL-17A-mediated gene expression via STAT3 in ASM cells. Collectively, our results raise the possibility that the IL-17A/STAT3 signaling pathway may play a crucial role in airway inflammatory responses.


Asunto(s)
Bronquios/inmunología , Bronquios/metabolismo , Quimiocina CCL11/biosíntesis , Interleucina-17/fisiología , Factor de Transcripción STAT3/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/inmunología , Bronquios/citología , Células Cultivadas , Quimiocina CCL11/genética , Regulación de la Expresión Génica/inmunología , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/inmunología , Unión Proteica/inmunología , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/inmunología
16.
Front Immunol ; 12: 641311, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305885

RESUMEN

PTX3 is a unique member of the long pentraxins family and plays an indispensable role in regulating the immune system. We previously showed that PTX3 deletion aggravates allergic inflammation via a Th17 -dominant phenotype and enhanced CD4 T cell survival using a murine model of ovalbumin (OVA) induced allergic inflammation. In this study, we identified that upon OVA exposure, increased infiltration of CD11c+CD11b+ dendritic cells (DCs) was observed in the lungs of PTX3-/- mice compared to wild type littermate. Further analysis showed that a short-term OVA exposure led to an increased number of bone marrow common myeloid progenitors (CMP) population concomitantly with increased Ly6Chigh CCR2high monocytes and CD11c+CD11b+ DCs in the lungs. Also, pulmonary CD11c+CD11b+ DCs from OVA-exposed PTX3-/- mice exhibited enhanced expression of maturation markers, chemokines receptors CCR2, and increased OVA uptake and processing compared to wild type controls. Taken together, our data suggest that PTX3 deficiency heightened lung CD11c+CD11b+DC numbers and function, hence exacerbating airway inflammatory response.


Asunto(s)
Proteína C-Reactiva/deficiencia , Proteína C-Reactiva/inmunología , Células Dendríticas/inmunología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/inmunología , Hipersensibilidad Respiratoria/inmunología , Alérgenos/inmunología , Alérgenos/toxicidad , Animales , Antígeno CD11b/inmunología , Antígeno CD11c/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Ovalbúmina/toxicidad
17.
PLoS One ; 16(6): e0252868, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34185781

RESUMEN

Regulation of dendritic cell functions is a complex process in which several mediators play diverse roles as a network in a context-dependent manner. The precise mechanisms underlying dendritic cell functions have remained to be addressed. Semaphorins play crucial roles in regulation of various cell functions. We previously revealed that Semaphorin 3E (Sema3E) contributes to regulation of allergen-induced airway pathology partly mediated by controlling recruitment of conventional dendritic cell subsets in vivo, though the underlying mechanism remained elusive. In this study, we investigate the potential regulatory role of Sema3E in dendritic cells. We demonstrated that bone marrow-derived dendritic cells differentiated from Sema3e-/- progenitors have an enhanced migration capacity both at the baseline and in response to CCL21. The enhanced migration ability of Sema3E dendritic cells was associated with an overexpression of the chemokine receptor (CCR7), elevated Rac1 GTPase activity and F-actin polymerization. Using a mouse model of allergic airway sensitization, we observed that genetic deletion of Sema3E leads to a time dependent upregulation of CCR7 on CD11b+ conventional dendritic cells in the lungs and mediastinal lymph nodes. Furthermore, aeroallergen sensitization of Sema3e-/- mice lead to an enhanced expression of PD-L2 and IRF-4 as well as enhanced allergen uptake in pulmonary CD11b+ DC, compared to wild type littermates. Collectively, these data suggest that Sema3E implicates in regulation of dendritic cell functions which could be considered a basis for novel immunotherapeutic strategies for the diseases associated with defective dendritic cells in the future.


Asunto(s)
Alérgenos/inmunología , Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Neumonía/inmunología , Semaforinas/fisiología , Actinas/metabolismo , Animales , Movimiento Celular , Quimiocina CCL21/metabolismo , Ratones , Ratones Noqueados , Neuropéptidos/metabolismo , Neumonía/metabolismo , Neumonía/patología , Receptores CCR7/metabolismo , Proteína de Unión al GTP rac1/metabolismo
18.
Cell Rep ; 33(11): 108513, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326783

RESUMEN

The long pentraxin 3 (PTX3) plays a critical role in inflammation, tissue repair, and wound healing. Here, we show that PTX3 regulates disease pathogenesis in cutaneous leishmaniasis (CL). PTX3 expression increases in skin lesions in patients and mice during CL, with higher expression correlating with severe disease. PTX3-deficient (PTX3-/-) mice are highly resistant to L. major and L. braziliensis infections. This enhanced resistance is associated with increases in Th17 and IL-17A responses. The neutralization of IL-17A abolishes this enhanced resistance, while rPTX3 treatment results in decrease in Th17 and IL-17A responses and increases susceptibility. PTX3-/- CD4+ T cells display increased differentiation to Th17 and expression of Th17-specific transcription factors. The addition of rPTX3 suppresses the expression of Th17 transcription factors, Th17 differentiation, and IL-17A production by CD4+ T cells from PTX3-/- mice. Collectively, our results show that PTX3 contributes to the pathogenesis of CL by negatively regulating Th17 and IL-17A responses.


Asunto(s)
Proteína C-Reactiva/inmunología , Linfocitos T CD4-Positivos/inmunología , Leishmaniasis Cutánea/inmunología , Proteínas del Tejido Nervioso/inmunología , Componente Amiloide P Sérico/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas de Fase Aguda/inmunología , Animales , Femenino , Humanos , Leishmaniasis Cutánea/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Biochem Biophys Res Commun ; 384(2): 167-72, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19401191

RESUMEN

Interleukin (IL)-9 is associated with key pathological features of asthma such as airway hyperresponsiveness, bronchoconstriction and mucus production. Inflammatory responses mediated by IL-9 rely on the expression of the IL-9R which has been reported on lung epithelial cells, T lymphocytes and recently on airway granulocyte infiltrates. In this study, we assessed the regulatory and constitutive cell surface expression of the IL-9Ralpha in unfractionated and purified human neutrophils from atopic asthmatics, atopic non-asthmatics and healthy normal controls. We demonstrate that T(H)2 cytokines (IL-4 or IL-13) and granulocyte macrophage-colony stimulating factor (GM-CSF) up-regulated mRNA and cell surface expression levels of the IL-9Ralpha in primary human and HL-60 differentiated neutrophils. Pharmacological inhibition of NF-kappaB did not affect T(H)2-mediated IL-9Ralpha expression in human neutrophils although IFN-gamma and IL-10 down-regulated IL-9Ralpha expression when co-incubated with IL-4, IL-13 or GM-CSF. Collectively, our results reveal a regulatory function for IFN-gamma and IL-10 on modulating the inducible IL-9Ralpha expression levels on peripheral blood neutrophils by T(H)2 cytokines.


Asunto(s)
Citocinas/metabolismo , Neutrófilos/inmunología , Receptores de Interleucina-9/biosíntesis , Células Th2/inmunología , Asma/inmunología , Membrana Celular/inmunología , Células HL-60 , Humanos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptores de Interleucina-9/genética , Regulación hacia Arriba
20.
J Allergy Clin Immunol ; 121(2): 492-498.e10, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18036649

RESUMEN

BACKGROUND: Glucocorticoids have been shown to inhibit human neutrophil apoptosis, with implications that this might help accentuate neutrophilic inflammation. OBJECTIVE: The aim of this study was to investigate the molecular mechanisms involved in glucocorticoid-mediated inhibition of primary human neutrophil apoptosis. METHODS: Primary human neutrophils were isolated from peripheral blood of healthy volunteers and cultured in vitro with dexamethasone. RESULTS: Here we confirm that dexamethasone, a classical glucocorticoid, significantly inhibited apoptosis of primary human neutrophils. This inhibition was not dependent on transrepression of proapoptotic molecules but was associated with induction of antiapoptotic Mcl-1. Remarkably, glucocorticoid-mediated enhancement of Mcl-1 and survival were significantly suppressed by pharmacologic inhibition of p38 mitogen-activated protein kinase or phosphatidylinositol 3-kinase. Inhibition of the above kinases also blocked glucocorticoid-induced maintenance of mitochondrial transmembrane potential and suppression of caspases. CONCLUSION: Phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase are protein kinases that regulate the prosurvival effect of glucocorticoids on human neutrophils.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/farmacología , Proteínas de Neoplasias/biosíntesis , Neutrófilos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis/fisiología , Inhibidores de Caspasas , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Dexametasona/antagonistas & inhibidores , Glucocorticoides/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA