Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases 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.
Adv Exp Med Biol ; 1304: 205-213, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019271

RESUMEN

Semaphorin3E belongs to the large family of semaphorin proteins. Semaphorin3E was initially identified as axon guidance cues in the neural system. It is universally expressed beyond the nervous system and contributes to regulating essential cell functions such as cell migration, proliferation, and adhesion. Binding of semaphorin3E to its receptor, plexinD1, triggers diverse signaling pathways involved in the pathogenesis of various diseases from cancer to autoimmune and allergic disorders. Here, we highlight the novel findings on the role of semaphorin3E in airway biology. In particular, we highlight our recent findings on the function and potential mechanisms by which semaphorin3E and its receptor, plexinD1, impact airway inflammation, airway hyperresponsiveness, and remodeling in the context of asthma.


Asunto(s)
Asma , Hipersensibilidad Respiratoria , Semaforinas , Asma/genética , Movimiento Celular , Humanos , Semaforinas/genética , Transducción de Señal
6.
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
7.
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
8.
J Immunol ; 199(3): 1086-1095, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28659355

RESUMEN

Despite decades of clinical and biomedical research, the pathogenesis of sepsis and its spectrum of diseases (severe sepsis and septic shock), which are leading causes of death in intensive care units, are still poorly understood. In this article, we show that signaling via the p110δ isoform of PI3K is critical for survival in experimental sepsis. Mice with an inactive knock-in mutation in the p110δ gene (p110δD910A) succumbed acutely to nonlethal dose LPS challenge. The susceptibility of p110δD910A mice to LPS was associated with increased neutrophil numbers and activities in the tissues, due in part to delayed apoptosis resulting mostly from inherent reduced regulatory T cell (Treg) numbers. Adoptive transfer of wild-type or p110δD910A Tregs abrogated exaggerated neutrophil activity, increased neutrophil apoptosis, and rescued p110δD910A mice from mortality after LPS challenge. We confirmed the clinical relevance of these findings by showing that human Tregs also regulate neutrophil function and survival. Collectively, our results show that PI3K δ is essential for survival during sepsis. In addition, our data highlight the importance of Tregs in regulating the pathogenesis of sepsis and septic shock via their effects on neutrophil survival and function, and provide evidence of regulation of innate immunity by cells of the adaptive immune system.


Asunto(s)
Neutrófilos/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Sepsis/inmunología , Choque Séptico/mortalidad , Linfocitos T Reguladores/inmunología , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular/genética , Proliferación Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase I , Técnicas de Sustitución del Gen , Inmunidad Innata , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones , Neutrófilos/patología , Neutrófilos/fisiología , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética , Isoformas de Proteínas , Sepsis/fisiopatología , Choque Séptico/inmunología , Choque Séptico/fisiopatología , Transducción de Señal
9.
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
11.
Am J Respir Cell Mol Biol ; 58(1): 21-27, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28817310

RESUMEN

Semaphorins are a large family of proteins originally identified as axon guidance cues that play a crucial role in neural development. They are also ubiquitously expressed beyond the nervous system and contribute to regulation of essential cell functions, such as cell migration, proliferation, and adhesion. Binding of semaphorins to their receptors, including plexins and neuropilins, triggers diverse signaling pathways, which are involved in the pathogenesis of various diseases, from cancer to autoimmune and allergic disorders. Despite emerging evidence suggestive of nonredundant roles of semaphorins in cellular and molecular mechanisms of the airway biology, their precise expression and function have not been fully addressed. Here, we first provide an overview about the semaphorin family, their receptors, signaling pathways, and their cellular functions. Then, we highlight the novel findings on the role of semaphorins in airway biology under developmental, homeostatic, and pathological conditions. In particular, we discuss the dual roles of semaphorins in respiratory disorders where they can up- or downregulate processes underlying the pathophysiology of the airway diseases. Next, our recent findings on the expression and function of semaphorin 3E in allergic asthma are further emphasized, and its potential mechanism of action in allergic airway inflammation and remodeling is discussed. Finally, we raise some unanswered questions aiming to develop future research directions.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma/metabolismo , Sistema Respiratorio/metabolismo , Semaforinas/metabolismo , Transducción de Señal , Animales , Asma/patología , Humanos , Sistema Respiratorio/patología
12.
J Neuroinflammation ; 15(1): 53, 2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29467001

RESUMEN

BACKGROUND: Spinal cord injury (SCI) triggers a robust neuroinflammatory response that governs secondary injury mechanisms with both degenerative and pro-regenerative effects. Identifying new immunomodulatory therapies to promote the supportive aspect of immune response is critically needed for the treatment of SCI. We previously demonstrated that SCI results in acute and permanent depletion of the neuronally derived Neuregulin-1 (Nrg-1) in the spinal cord. Increasing the dysregulated level of Nrg-1 through acute intrathecal Nrg-1 treatment enhanced endogenous cell replacement and promoted white matter preservation and functional recovery in rat SCI. Moreover, we identified a neuroprotective role for Nrg-1 in moderating the activity of resident astrocytes and microglia following injury. To date, the impact of Nrg-1 on immune response in SCI has not yet been investigated. In this study, we elucidated the effect of systemic Nrg-1 therapy on the recruitment and function of macrophages, T cells, and B cells, three major leukocyte populations involved in neuroinflammatory processes following SCI. METHODS: We utilized a clinically relevant model of moderately severe compressive SCI in female Sprague-Dawley rats. Nrg-1 (2 µg/day) or saline was delivered subcutaneously through osmotic mini-pumps starting 30 min after SCI. We conducted flow cytometry, quantitative real-time PCR, and immunohistochemistry at acute, subacute, and chronic stages of SCI to investigate the effects of Nrg-1 treatment on systemic and spinal cord immune response as well as cytokine, chemokine, and antibody production. RESULTS: We provide novel evidence that Nrg-1 promotes a pro-regenerative immune response after SCI. Bioavailability of Nrg-1 stimulated a regulatory phenotype in T and B cells and augmented the population of M2 macrophages in the spinal cord and blood during the acute and chronic stages of SCI. Importantly, Nrg-1 fostered a more balanced microenvironment in the injured spinal cord by attenuating antibody deposition and expression of pro-inflammatory cytokines and chemokines while upregulating pro-regenerative mediators. CONCLUSION: We provide the first evidence of a significant regulatory role for Nrg-1 in neuroinflammation after SCI. Importantly, the present study establishes the promise of systemic Nrg-1 treatment as a candidate immunotherapy for traumatic SCI and other CNS neuroinflammatory conditions.


Asunto(s)
Inmunidad Celular/efectos de los fármacos , Neurregulina-1/administración & dosificación , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/inmunología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Femenino , Inmunidad Celular/fisiología , Infusiones Subcutáneas , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/fisiología
13.
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
14.
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
16.
Hepatology ; 63(2): 620-32, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26289140

RESUMEN

UNLABELLED: Visceral leishmaniasis (VL) is associated with severe immune dysfunction and if untreated leads to death. Because the liver is one of the primary target organs in VL, unraveling the mechanisms governing the local hepatic immune response is important for understanding the immunopathogenesis of VL. We previously reported that mice with inactivating knockin mutation in the p110δ gene (p110δ(D910A) ) are resistant to VL, due in part to impaired regulatory T-cell (Treg) expansion. In this study, we investigated the mechanism of this resistance by focusing on hepatic stellate cells (HSCs), which are known to regulate Treg induction and expansion. We show that HSCs are infected with Leishmania donovani in vivo and in vitro and that this infection leads to the production of interleukin-2, interleukin-6, and transforming growth factor-ß, cytokines known to induce Tregs. We further demonstrate that L. donovani infection leads to expansion of HSCs in a p110δ-dependent manner and that this correlated with proliferation of hepatic Tregs in vivo. In vitro studies clearly show that L. donovani-infected HSCs induce CD4(+) T cells to become Tregs and expand Tregs in a p110δ-dependent manner. Targeted depletion of HSCs during infection caused a dramatic reduction in liver Treg numbers and proliferation, which was associated with a decrease in interleukin-10 production by hepatic T cells and a more efficient parasite control. CONCLUSION: These results demonstrate the critical role of HSCs in the pathogenesis of VL and suggest that the enhanced resistance of p110δ(D910A) mice to L. donovani infection is due in part to impaired expansion and inability of their HSCs to induce and expand Tregs in the liver.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/inmunología , Células Estrelladas Hepáticas/inmunología , Inmunidad Celular/inmunología , Leishmaniasis Visceral/inmunología , Hígado/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C
17.
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
18.
Am J Respir Cell Mol Biol ; 50(6): 1053-63, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24393021

RESUMEN

Airway smooth muscle (ASM) cells are thought to contribute to the pathogenesis of allergic asthma by orchestrating and perpetuating airway inflammation and remodeling responses. In this study, we evaluated the IL-17RA signal transduction and gene expression profile in ASM cells from subjects with mild asthma and healthy individuals. Human primary ASM cells were treated with IL-17A and probed by the Affymetrix GeneChip array, and gene targets were validated by real-time quantitative RT-PCR. Genomic analysis underlined the proinflammatory nature of IL-17A, as multiple NF-κB regulatory factors and chemokines were induced in ASM cells. Transcriptional regulators consisting of primary response genes were overrepresented and displayed dynamic expression profiles. IL-17A poorly enhanced IL-1ß or IL-22 gene responses in ASM cells from both subjects with mild asthma and healthy donors. Interestingly, protein modifications to the NF-κB regulatory network were not observed after IL-17A stimulation, although oscillations in IκBε expression were detected. ASM cells from subjects with mild asthma up-regulated more genes with greater overall variability in response to IL-17A than from healthy donors. Finally, in response to IL-17A, ASM cells displayed rapid activation of the extracellular signal-regulated kinase/ribosomal S6 kinase signaling pathway and increased nuclear levels of phosphorylated extracellular signal-regulated kinase. Taken together, our results suggest that IL-17A mediated modest gene expression response, which, in cooperation with the NF-κB signaling network, may regulate the gene expression profile in ASM cells.


Asunto(s)
Asma/genética , Asma/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Miocitos del Músculo Liso/metabolismo , Adulto , Asma/patología , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Miocitos del Músculo Liso/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación , Transducción de Señal , Transcriptoma , Regulación hacia Arriba , Interleucina-22
20.
Pulm Pharmacol Ther ; 26(1): 86-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22580035

RESUMEN

The airway smooth muscle (ASM) is no longer considered as merely a contractile apparatus and passive recipient of growth factors, neurotransmitters and inflammatory mediators signal but a critical player in the perpetuation and modulation of airway inflammation and remodeling. In recent years, a molecular link between ASM and IgE has been established through Fc epsilon receptors (FcεRs) in modulating the phenotype and function of these cells. Particularly, the expression of high affinity IgE receptor (FcεRI) has been noted in primary human ASM cells in vitro and in vivo within bronchial biopsies of allergic asthmatic subjects. The activation of FcεRI on ASM cells suggests a critical yet almost completely ignored network which may modulate ASM cell function in allergic asthma. This review is intended to provide a historical perspective of IgE effects on ASM and highlights the recent updates in the expression and function of FcεRI, and to present future perspectives of activation of this pathway in ASM cells.


Asunto(s)
Inflamación/inmunología , Miocitos del Músculo Liso/metabolismo , Receptores de IgE/genética , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Animales , Asma/inmunología , Asma/fisiopatología , Expresión Génica , Humanos , Inmunoglobulina E/inmunología , Inflamación/patología , Músculo Liso/citología , Músculo Liso/inmunología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/inmunología , Receptores de IgE/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA