Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cancer Sci ; 113(10): 3405-3416, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35879596

RESUMEN

Most breast cancer-related deaths are caused by metastasis in vital organs including the lungs. Development of supportive metastatic microenvironments, referred to as premetastatic niches (PMNs), in certain distant organs before arrival of metastatic cells, is critical in metastasis. However, the mechanisms of PMN formation are not fully clear. Here, we demonstrated that chemoattractant C-C motif chemokine ligand 2 (CCL2) could be stimulated by heat shock protein 60 (HSP60) on the surface of murine 4 T1 breast cancer cell-released LC3+ extracellular vesicles (LC3+ EVs) via the TLR2-MyD88-NF-κB signal cascade in lung fibroblasts, which subsequently promoted lung PMN formation through recruiting monocytes and suppressing T cell function. Consistently, reduction of LC3+ EV release or HSP60 level or neutralization of CCL2 markedly attenuated PMN formation and lung metastasis. Furthermore, the number of circulating LC3+ EVs and HSP60 level on LC3+ EVs in the plasma of breast cancer patients were positively correlated with disease progression and lung metastasis, which might have potential value as biomarkers of lung metastasis in breast cancer patients (AUC = 0.898, 0.694, respectively). These findings illuminate a novel mechanism of PMN formation and might provide therapeutic targets for anti-metastasis therapy for patients with breast cancer.


Asunto(s)
Neoplasias de la Mama , Vesículas Extracelulares , Neoplasias Pulmonares , Animales , Neoplasias de la Mama/patología , Chaperonina 60/metabolismo , Factores Quimiotácticos/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Ligandos , Neoplasias Pulmonares/patología , Ratones , Proteínas Asociadas a Microtúbulos , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Metástasis de la Neoplasia/patología , Receptor Toll-Like 2 , Microambiente Tumoral
2.
FASEB J ; 34(10): 13696-13710, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32786052

RESUMEN

Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome are life-threatening diseases. Despite recent advances in intensive care medicine, the mortality is still as high as 50%, which stems from our insufficient understanding of the underlying mechanisms of the diseases. The roles of C/EBPß and C/EBPδ have been extensively investigated in LPS- and IgG immune complexes-stimulated acute lung injury. However, the effect of C/EBPγ, belonging to the same family as C/EBPß and C/EBPδ, on ALI has not been elucidated. Our previous data have shown that during LPS-/IgG immune complexes-induced ALI, the DNA binding activities of C/EBPγ are obviously reduced. In the present study, we determine whether ALI induced by LPS and IgG immune complexes is affected by C/EBPγ. We find that adenovirus-mediated C/EBPγ expression in the lung tissue alleviates LPS-/IgG immune complexes-stimulated acute pulmonary damage through reducing vascular permeability changes and recruitment of neutrophils into alveolar spaces, which might be linked to a decrease in the production of pro-inflammatory mediators, such as TNF-α and IL-6. Moreover, our data obtained from macrophages in vitro are consistent with the in vivo results. In terms of mechanisms, C/EBPγ might inhibit LPS-/IgG immune complexes-mediated inflammation via alleviating C/EBPß and C/EBPδ transcription activities as reflected by luciferase assays. However, the NF-κB-dependent production of pro-inflammatory mediators is not affected by C/EBPγ. Taken together, C/EBPγ suppresses LPS- and IgG immune complexes-induced pro-inflammatory mediators' production through the downregulation of C/EBP but not NF-κB activation, leading to the subsequent attenuation of ALI. Collectively, our data provide an insight into the critical role of C/EBPγ in acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación hacia Abajo , Activación Transcripcional , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Animales , Complejo Antígeno-Anticuerpo , Permeabilidad Capilar , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Immunol ; 193(7): 3769-78, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172497

RESUMEN

Increasing evidence suggests that the novel anti-inflammatory and proresolving mediators such as the resolvins play an important role during inflammation. However, the functions of these lipid mediators in immune complex-induced lung injury remain unknown. In this study, we determined the role of aspirin-triggered resolvin D1 (AT-RvD1) and its metabolically stable analog, 17R-hydroxy-19-para-fluorophenoxy-resolvin D1 methyl ester (p-RvD1), in IgG immune complex-induced inflammatory responses in myeloid cells and injury in the lung. We show that lung vascular permeability in the AT-RvD1- or p-RvD1-treated mice was significantly reduced when compared with values in mice receiving control vesicle during the injury. Furthermore, i.v. administration of either AT-RvD1 or p-RvD1 caused significant decreases in the bronchoalveolar lavage fluid contents of neutrophils, inflammatory cytokines, and chemokines. Of interest, AT-RvD1 or p-RvD1 significantly reduced bronchoalveolar lavage fluid complement C5a level. By EMSA, we demonstrate that IgG immune complex-induced activation of NF-κB and C/EBPß transcription factors in the lung was significantly inhibited by AT-RvD1 and p-RvD1. Moreover, AT-RvD1 dramatically mitigates IgG immune complex-induced NF-κB and C/EBP activity in alveolar macrophages. Also, secretion of TNF-α, IL-6, keratinocyte cell-derived chemokine, and MIP-1α from IgG immune complex-stimulated alveolar macrophages or neutrophils was significantly decreased by AT-RvD1. These results suggest a new approach to the blocking of immune complex-induced inflammation.


Asunto(s)
Lesión Pulmonar Aguda , Antiinflamatorios no Esteroideos/farmacología , Complejo Antígeno-Anticuerpo/inmunología , Aspirina/farmacología , Complemento C5a/inmunología , Inmunoglobulina G/inmunología , Neumonía , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/prevención & control , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Proteínas Potenciadoras de Unión a CCAAT/inmunología , Línea Celular , Quimiocina CCL3/inmunología , Citocinas/inmunología , Ácidos Docosahexaenoicos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Ratones , FN-kappa B/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Neumonía/prevención & control
4.
Exp Cell Res ; 337(1): 120-7, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26209606

RESUMEN

Compelling evidence indicates that suppressor of cytokine signaling 3 (SOCS3) plays a pivotal regulatory role in inflammation. However, the function of SOCS3 in inflammatory responses mediated by Fcγ receptor (FcγR) remains largely unknown. In the current study, we found that SOCS3 expression was greatly enhanced in peritoneal macrophages treated with IgG immune complex (IgG IC). By over-expressing SOCS3 in macrophages, we observed that SOCS3 promoted IgG immune complex-induced production of inflammatory mediators, including IL-6, TNF-α, MIP-2, and MIP-1α. In contrast, SOCS3-defective peritoneal macrophages generated less inflammatory cytokines and chemokines when compared with their wild type counterparts during IgG IC-induced inflammatory responses. We further demonstrated that CCAAT/enhancer-binding protein (C/EBP) δ transcription factor was the major downstream target of SOCS3 in macrophages. These data suggested that SOCS3 was an inflammatory enhancer in IgG IC-treated macrophages by increasing C/EBPδ activity. To elucidate the role for myeloid-derived SOCS3 in IgG IC-induced inflammation in vivo, LysM-cre SOCS3(fl/fl) mice lacking SOCS3 in macrophages and neutrophils were generated. We found that SOCS3 deficiency greatly alleviated IgG IC-induced generation of pro-inflammatory mediators in lungs, consistent with the in vitro data. Our current findings may provide a new theoretical basis for designing drugs for treatment of IgG IC-associated diseases.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/fisiología , Macrófagos/metabolismo , Receptores de IgG/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Transcripción Genética , Animales , Línea Celular , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/inmunología , Ratones Transgénicos , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas
5.
Mediators Inflamm ; 2016: 9201604, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27194827

RESUMEN

Optimal methods are applied to acute lung injury (ALI) and the acute respiratory distress syndrome (ARDS), but the mortality rate is still high. Accordingly, further studies dedicated to identify novel therapeutic approaches to ALI are urgently needed. Bigelovii A is a new natural product and may exhibit anti-inflammatory activity. Therefore, we sought to investigate its effect on lipopolysaccharide- (LPS-) induced ALI and the underlying mechanisms. We found that LPS-induced ALI was significantly alleviated by Bigelovii A treatment, characterized by reduction of proinflammatory mediator production, neutrophil infiltration, and lung permeability. Furthermore, Bigelovii A also downregulated LPS-stimulated inflammatory mediator expressions in vitro. Moreover, both NF-κB and CCAAT/enhancer-binding protein δ (C/EBPδ) activation were obviously attenuated by Bigelovii A treatment. Additionally, phosphorylation of both p38 MAPK and ERK1/2 (upstream signals of C/EBPδ activation) in response to LPS challenge was also inhibited by Bigelovii A. Therefore, Bigelovii A could attenuate LPS-induced inflammation by suppression of NF-κB, inflammatory mediators, and p38 MAPK/ERK1/2-C/EBPδ, inflammatory mediators signaling pathways, which provide a novel theoretical basis for the possible application of Bigelovii A in clinic.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Proteína delta de Unión al Potenciador CCAAT/metabolismo , FN-kappa B/metabolismo , Saponinas/farmacología , Triterpenos/farmacología , Lesión Pulmonar Aguda/inducido químicamente , Animales , Western Blotting , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos
6.
Am J Pathol ; 182(2): 420-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23177475

RESUMEN

Although inflammation plays a central role in the pathogenesis of acute lung injury, the molecular mechanisms underlying inflammatory responses in acute lung injury are poorly understood, and therapeutic options remain limited. CCAAT/enhancer-binding proteins, C/EBPß and C/EBPδ, are expressed in the lung and have been implicated in the regulation of inflammatory mediators. However, their functions in lung pathobiological characteristics are not well characterized. Herein, we show that C/EBPß and C/EBPδ are activated in mouse lung after intrapulmonary deposition of lipopolysaccharide (LPS). Mice carrying a targeted deletion of the C/EBPδ gene displayed significant attenuation of the lung permeability index (lung vascular leak of albumin), lung neutrophil accumulation (myeloperoxidase activity), and neutrophils in bronchial alveolar lavage fluids compared with wild-type mice. These phenotypes were consistent with morphological evaluation of lung, which showed reduced inflammatory cell influx and minimal intra-alveolar hemorrhage. Moreover, mutant mice expressed considerably less tumor necrosis factor-α, IL-6, and macrophage inflammatory protein-2 in bronchial alveolar lavage fluids in LPS-injured lung compared with wild-type mice. In contrast, C/EBPß deficiency had no effect on LPS-induced lung injury. By using small-interfering RNA-mediated knockdown for C/EBPδ, we demonstrate, for the first time to our knowledge, that C/EBPδ plays a critical role for the tumor necrosis factor-α, IL-6, and macrophage inflammatory protein-2 production in LPS-stimulated alveolar macrophages. These findings demonstrate that C/EBPδ, but not C/EBPß, plays an important role in LPS-induced lung inflammatory responses and injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Animales , Líquido del Lavado Bronquioalveolar , Proteína delta de Unión al Potenciador CCAAT/deficiencia , Línea Celular , Quimiocinas/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Interleucina-6/metabolismo , Lipopolisacáridos , Luciferasas/metabolismo , Pulmón/enzimología , Pulmón/patología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/patología , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
FASEB J ; 27(8): 2967-76, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23585399

RESUMEN

Although uncontrolled inflammatory response plays a central role in the pathogenesis of acute lung injury (ALI), the precise molecular mechanisms underlying the development of this disorder remain poorly understood. SOCS3 is an important negative regulator of IL-6-type cytokine signaling. SOCS3 is induced in lung during LPS-induced lung injury, suggesting that generation of SOCS3 may represent a regulatory product during ALI. In the current study, we created mice lacking SOCS3 expression in macrophages and neutrophils (LysM-cre SOCS3(fl/fl)). We evaluated the lung inflammatory response to LPS in both LysM-cre SOCS3(fl/fl) mice and the wild-type (WT) mice (SOCS3(fl/fl)). LysM-cre SOCS3(fl/fl) mice displayed significant increase of the lung permeability index (lung vascular leak of albumin), neutrophils, lung neutrophil accumulation (myeloperoxidase activity), and proinflammatory cytokines/chemokines in bronchial alveolar lavage fluids compared to WT mice. These phenotypes were consistent with morphological evaluation of lung, which showed enhanced inflammatory cell influx and intra-alveolar hemorrhage. We further identify the transcription factor, CCAAT/enhancer-binding protein (C/EBP) δ as a critical downstream target of SOCS3 in LPS-induced ALI. These results indicate that SOCS3 has a protective role in LPS-induced ALI by suppressing C/EBPδ activity in the lung. Elucidating the function of SOCS3 would represent prospective targets for a new generation of drugs needed to treat ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Proteína delta de Unión al Potenciador CCAAT/inmunología , Células Mieloides/inmunología , Transducción de Señal/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Células Cultivadas , Quimiocinas/inmunología , Quimiocinas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/inmunología , Lipopolisacáridos/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Peroxidasa/inmunología , Peroxidasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética
8.
J Immunol ; 189(3): 1480-90, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22732594

RESUMEN

C/EBPs, particularly C/EBPß and C/EBPδ, are known to participate in the regulation of many genes associated with inflammation. However, very little is known regarding the activation and functions of C/EBPß and C/EBPδ in acute lung inflammation and injury. In this study, we show that both C/EBPß and C/EBPδ activation are triggered in lungs and in alveolar macrophages following intrapulmonary deposition of IgG immune complexes. We further show that mice carrying a targeted deletion of the C/EBPß gene displayed significant attenuation of the permeability index (lung vascular leak of albumin), lung neutrophil accumulation (myeloperoxidase activity), total number of WBCs, and neutrophils in bronchoalveolar lavage fluids compared with wild-type mice. Moreover, the mutant mice expressed considerably less TNF-α, IL-6, and CXC/CC chemokine and soluble ICAM-1 proteins in bronchoalveolar lavage fluids, and corresponding mRNAs in the IgG immune complex-injured lung, compared with wild-type mice. These phenotypes were associated with a significant reduction in morphological lung injury. In contrast, C/EBPδ deficiency had no effect on IgG immune complex-induced lung injury. IgG immune complex-stimulated C/EBPß-deficient alveolar macrophages released significantly less TNF-α, IL-6, MIP-2, keratinocyte cell-derived chemokine, and MIP-1α compared with wild-type cells. Similar decreases in IgG immune complex-induced inflammatory mediator production were observed following small interfering RNA ablation of C/EBPß in a murine alveolar macrophage cell line. These findings implicate C/EBPß as a critical regulator of IgG immune complex-induced inflammatory responses and injury in the lung.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Complejo Antígeno-Anticuerpo/administración & dosificación , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Complejo Antígeno-Anticuerpo/efectos adversos , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Proteína delta de Unión al Potenciador CCAAT/deficiencia , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/fisiología , Línea Celular , Modelos Animales de Enfermedad , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/efectos adversos , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/efectos adversos , Mediadores de Inflamación/fisiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Inflammation ; 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38376609

RESUMEN

The role of programmed death ligand 1 (PD-L1) has been extensively investigated in adaptive immune system. However, increasing data show that innate immune responses are also affected by the immune checkpoint molecule. It has been demonstrated that regulation of PD-L1 signaling in macrophages may be a potential therapeutic method for acute respiratory distress syndrome (ARDS). However, the PD-L1 expression pattern in local macrophages and whole lung tissues remains mysterious, hindering optimization of the potential treatment program. Therefore, we aim to determine the PD-L1 expression pattern during ARDS. Our findings show that PD-L1 levels are markedly increased in lipopolysaccharide (LPS)-stimulated lung tissues, which might be attributable to an increase in the gene expression by immune cells, including macrophages and neutrophils. In vitro experiments are performed to explore the mechanism involved in LPS-induced PD-L1 production. We find that PD-L1 generation is controlled by transcription factors early growth response 1 (Egr-1) and CCAAT/enhancer binding protein delta (C/EBPδ). Strikingly, PD-L1 production is enhanced by phosphoinositide-3 kinase (PI3K)-protein kinase B (AKT) signaling pathway via up-regulation of Egr-1 and C/EBPδ expressions. Additionally, we observe that expressions of Egr-1 and C/EBPδ mutually reinforce each other. Moreover, we observe that PD-L1 is protective for ARDS due to its regulatory role in macrophage-associated inflammatory response. In summary, during LPS-induced ARDS, PD-L1 expression, which is beneficial for the disease, is increased via the PI3K-AKT1-Egr-1/C/EBPδ signaling pathway, providing theoretical basis for application of methods controlling PD-L1 signaling in macrophages for ARDS treatment in clinic.

10.
J Biol Chem ; 287(5): 3217-30, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22147692

RESUMEN

CCAAT/enhancer-binding protein ß (C/EBPß) and C/EBPδ are known to participate in the regulation of many genes associated with inflammation. However, little is known about the activation and function of C/EBPß and -δ in inflammatory responses elicited by Fcγ receptor (FcγR) activation. Here we show that C/EBPß and -δ activation are induced in IgG immune complex (IC)-treated macrophages. The increased expression of C/EBPß and -δ occurred at both mRNA and protein levels. Furthermore, induction of C/EBPß and -δ was mediated, to a large extent, by activating FcγRs. Using siRNA-mediated knockdown as well as macrophages deficient for C/EBPß and/or -δ, we demonstrate that C/EBPß and -δ play a critical role in the production of TNF-α, MIP-2, and MIP-1α in IgG IC-stimulated macrophages. Moreover, both ERK1/2 and p38 MAPK are involved in C/EBP induction and TNF-α, MIP-2, and MIP-1α production induced by IgG IC. We provide the evidence that C5a regulates IgG IC-induced inflammatory responses by enhancing ERK1/2 and p38 MAPK activities as well as C/EBPß and -δ activities. Collectively, these data suggest that C/EBPß and -δ are key regulators for FcγR-mediated induction of cytokines and chemokines in macrophages. Furthermore, C/EBPs may play an important regulatory role in IC-associated inflammatory responses.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Quimiocinas/biosíntesis , Complemento C5a/metabolismo , Receptores de IgG/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/genética , Línea Celular , Complemento C5a/genética , Inflamación/genética , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de IgG/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Inflamm (Lond) ; 20(1): 34, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37864223

RESUMEN

BACKGROUND: Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) are life threatening pulmonary diseases, and we are now lack of effective therapeutic methods. Inflammatory responses are essential for initiating ALI/ARDS. Thus, ameliorating inflammatory reaction might be beneficial for treatment of the disease. There are increasing data that phosphodiesterase-4 (PDE4)-selective inhibitors, which may elevate cellular cyclic adenosine 3', 5'-monophosphate (cAMP) level, could suppress inflammation. However, whether they could be used to treat IgG immune complex (IgG-IC)-associated ALI has not been determined. METHODS: ALI is induced by treating mice with airway deposition of IgG immune complexes. Cellular cAMP concentrations are elevated by treating mice or macrophages with Rolipram/Roflumilast. The degree of pulmonary injury is reflected by lung permeability, leukocyte accumulation, histological change and expressions of pro-inflammatory mediators. 6-Bnz-cAMP and H-89 are used to regulate protein kinase A (PKA) activity, and 8-pCPT-2'-O-Me-cAMP is applied to activate exchange proteins directly activated by cAMP (Epac). Gene expressions are analyzed by real-time PCR, ELISA or Western blot. CCAAT/enhancer binding protein (C/EBP) and activation protein 1 (AP-1) transcription activities are estimated by measuring the luciferase productions. RESULTS: IgG-IC-induced ALI is attenuated by the PDE4-selective inhibitor, which is due to reduced expressions of cytokine and chemokines. Interestingly, we find that cAMP downstream effector molecule PKA but not Epac is involved in negative regulation of IgG-IC-mediated pro-inflammatory mediators' productions. Mechanistically, activation of cAMP-PKA signal axis leads to inactivation of MAPK pathway, resulting in a decrease in C/EBP- and AP-1-mediated transcriptions of pro-inflammatory mediators. CONCLUSIONS: Our data demonstrate, for the first time, that cAMP-PKA signal is involved in down-regulation of IgG-IC-associated inflammatory responses via down-regulating MAPK activation, which is critical for transcriptional activities of C/EBP and AP-1. Collectively, our experiments provide theoretical base for the potential application of PDE4-selective inhibitor to clinic for treatment of IgG-IC-related acute lung injury.

12.
Int Immunopharmacol ; 117: 109744, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36812669

RESUMEN

Thymosin α-1 (Tα-1) is an immunomodulating polypeptide of 28 amino acids, which was the first peptide isolated from thymic tissue and has been widely used for the treatment of viral infections, immunodeficiencies, and especially malignancies. Tα-1 stimulates both innate and adaptive immune responses, and its regulation of innate immune cells and adaptive immune cells varies under different disease conditions. Pleiotropic regulation of immune cells by Tα-1 depends on activation of Toll-like receptors and its downstream signaling pathways in various immune microenvironments. For treatment of malignancies, the combination of Tα-1 and chemotherapy has a strong synergistic effect by enhancing the anti-tumor immune response. On the basis of the pleiotropic effect of Tα-1 on immune cells and the promising results of preclinical studies, Tα-1 may be a favorable immunomodulator to enhance the curative effect and decrease immune-related adverse events of immune checkpoint inhibitors to develop novel cancer therapies.


Asunto(s)
Neoplasias , Timosina , Humanos , Timalfasina/uso terapéutico , Timalfasina/farmacología , Timosina/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Inmunidad , Microambiente Tumoral
13.
Adv Sci (Weinh) ; 10(17): e2206814, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37097733

RESUMEN

Stiffness is an important physical property of biomaterials that determines stem cell fate. Guiding stem cell differentiation via stiffness modulation has been considered in tissue engineering. However, the mechanism by which material stiffness regulates stem cell differentiation into the tendon lineage remains controversial. Increasing evidence demonstrates that immune cells interact with implanted biomaterials and regulate stem cell behaviors via paracrine signaling; however, the role of this mechanism in tendon differentiation is not clear. In this study, polydimethylsiloxane (PDMS) substrates with different stiffnesses are developed, and the tenogenic differentiation of mesenchymal stem cells (MSCs) exposed to different stiffnesses and macrophage paracrine signals is investigated. The results reveal that lower stiffnesses facilitates tenogenic differentiation of MSCs, while macrophage paracrine signals at these stiffnesses suppress the differentiation. When exposed to these two stimuli, MSCs still exhibit enhanced tendon differentiation, which is further elucidated by global proteomic analysis. Following subcutaneous implantation in rats for 2 weeks, soft biomaterial induces only low inflammation and promotes tendon-like tissue formation. In conclusion, the study demonstrates that soft, rather than stiff, material has a greater potential to guide tenogenic differentiation of stem cells, which provides comprehensive evidence for optimized bioactive scaffold design in tendon tissue engineering.


Asunto(s)
Células Madre Mesenquimatosas , Comunicación Paracrina , Ratas , Animales , Proteómica , Diferenciación Celular , Materiales Biocompatibles
14.
FASEB J ; 25(12): 4292-300, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21859893

RESUMEN

Growing evidence suggests that transcription factor signal transducer and activator of transcription (Stat) 3 may play an important regulatory role during inflammation. However, the function of Stat3 in acute lung injury (ALI) is largely unknown. In the current study, by using an adenoviral vector expressing a dominant-negative Stat3 isoform (Ad-Stat3-EVA), we determined the role of Stat3 in IgG immune complex (IC)-induced inflammatory responses and injury in the lung from C57BL/6J mice. We show that IgG IC-induced DNA binding activity of Stat3 in the lung was significantly inhibited by Stat3-EVA. We demonstrate that both lung vascular permeability (albumin leak) and lung myeloperoxidase accumulation in the Ad-Stat-EVA treated mice were substantially reduced when compared with values in mice receiving control virus (Ad-GFP) during the injury. Furthermore, intratracheal administration of Ad-Stat3-EVA caused significant decreases in the contents of neutrophils, inflammatory cytokines (TNF-α and IL-6), chemokines [keratinocyte cell-derived chemokine, macrophage inflammatory protein (MIP)-1α, and MIP-1ß], and complement component C5a in bronchoalveolar lavage fluids. Using Stat3-specific small interfering RNA, we show that knocking down Stat3 expression in alveolar macrophages (MH-S cells) significantly reduced the production of proinflammatory mediators on IgG IC stimulation. These data suggest that Stat3 plays an essential role in the pathogenesis of IgG IC-induced ALI by mediating the acute inflammatory responses in the lung and alveolar macrophages.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Inmunoglobulina G/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Secuencia de Bases , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas/metabolismo , Citocinas/metabolismo , ADN/metabolismo , Técnicas de Silenciamiento del Gen , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética
15.
Cancer Res ; 82(10): 1991-2002, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35364609

RESUMEN

The immunologic effects of chemotherapy-induced tumor cell death are not completely understood. Accumulating evidence suggests that phagocytic clearance of apoptotic tumor cells, also known as efferocytosis, is an immunologically silent process, thus maintaining an immunosuppressive tumor microenvironment (TME). Here we report that, in the breast tumor microenvironment, thymosin α-1 (Tα-1) significantly reverses M2 polarization of IL10-producing tumor-associated macrophages (TAM) during efferocytosis induced by apoptotic cells. Mechanistically, Tα-1, which bound to phosphatidylserine on the surface of apoptotic tumor cells and was internalized by macrophages, triggered the activation of SH2-containing inositol 5'-phosphatase 1 (SHIP1) through the lysosomal Toll-like receptor 7 (TLR7)/MyD88 pathway, subsequently resulting in dephosphorylation of efferocytosis-activated TBK1 and reduction of efferocytosis-induced IL10. Tα-1 combined with epirubicin chemotherapy markedly suppressed tumor growth in an in vivo breast cancer model by reducing macrophage-derived IL10 and enhancing the number and function of tumor-infiltrating CD4+ and CD8+ T cells. In conclusion, Tα-1 improved the curative effect of chemotherapy by reversing M2 polarization of efferocytosis-activated macrophages, suggesting that Tα-1 injection immediately after chemotherapy may contribute to highly synergistic antitumor effects in patients with breast cancer. SIGNIFICANCE: Thymosin α-1 improves the curative effect of chemotherapy by reversing efferocytosis-induced M2 polarization of macrophages via activation of a TLR7/SHIP1 axis.


Asunto(s)
Neoplasias de la Mama , Receptor Toll-Like 7 , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Interleucina-10 , Timalfasina , Microambiente Tumoral , Macrófagos Asociados a Tumores
16.
J Biol Chem ; 285(48): 37227-39, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20876575

RESUMEN

Suppressor of cytokine signaling 3 (SOCS3) is an important intracellular protein that inhibits cytokine signaling in numerous cell types and has been implicated in several inflammatory diseases. However, the expression and function of SOCS3 in osteoblasts are not known. In this study, we demonstrated that SOCS3 expression was transiently induced by LPS in osteoblasts, and apparently contributed to the inhibition of IL-6 induction by LPS treatment. We found that tyrosine 204 of the SOCS box, the SH2 domain, and the N-terminal kinase inhibitory region (KIR) of SOCS3 were all involved in its IL-6 inhibition. Furthermore, we demonstrated that CCAAT/enhancer-binding protein (C/EBP) ß was activated by LPS (increased DNA binding activity), and played a key role in LPS-induced IL-6 expression in osteoblasts. We further provided the evidence that SOCS3 functioned as a negative regulator for LPS response in osteoblasts by suppressing C/EBPß DNA binding activity. In addition, tyrosine 204 of the SOCS box, the SH2 domain, and the N-terminal kinase inhibitory region (KIR) of SOCS3 were all required for its C/EBPß inhibition. These findings suggest that SOCS3 by interfering with C/EBPß activation may have an important regulatory role during bone-associated inflammatory responses.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación hacia Abajo , Interleucina-6/metabolismo , Lipopolisacáridos/metabolismo , Osteoblastos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular , Células Cultivadas , ADN/genética , ADN/metabolismo , Interleucina-6/genética , Ratones , Unión Proteica , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Dominios Homologos src
17.
Front Immunol ; 12: 634889, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717177

RESUMEN

Background: The ligand-activated transcription factor peroxisome proliferator-activated receptor (PPAR) γ plays crucial roles in diverse biological processes including cellular metabolism, differentiation, development, and immune response. However, during IgG immune complex (IgG-IC)-induced acute lung inflammation, its expression and function in the pulmonary tissue remains unknown. Objectives: The study is designed to determine the effect of PPARγ on IgG-IC-triggered acute lung inflammation, and the underlying mechanisms, which might provide theoretical basis for therapy of acute lung inflammation. Setting: Department of Pathogenic Biology and Immunology, Medical School of Southeast University. Subjects: Mice with down-regulated/up-regulated PPARγ activity or down-regulation of Early growth response protein 1 (Egr-1) expression, and the corresponding controls. Interventions: Acute lung inflammation is induced in the mice by airway deposition of IgG-IC. Activation of PPARγ is achieved by using its agonist Rosiglitazone or adenoviral vectors that could mediate overexpression of PPARγ. PPARγ activity is suppressed by application of its antagonist GW9662 or shRNA. Egr-1 expression is down-regulated by using the gene specific shRNA. Measures and Main Results: We find that during IgG-IC-induced acute lung inflammation, PPARγ expression at both RNA and protein levels is repressed, which is consistent with the results obtained from macrophages treated with IgG-IC. Furthermore, both in vivo and in vitro data show that PPARγ activation reduces IgG-IC-mediated pro-inflammatory mediators' production, thereby alleviating lung injury. In terms of mechanism, we observe that the generation of Egr-1 elicited by IgG-IC is inhibited by PPARγ. As an important transcription factor, Egr-1 transcription is substantially increased by IgG-IC in both in vivo and in vitro studies, leading to augmented protein expression, thus amplifying IgG-IC-triggered expressions of inflammatory factors via association with their promoters. Conclusion: During IgG-IC-stimulated acute lung inflammation, PPARγ activation can relieve the inflammatory response by suppressing the expression of its downstream target Egr-1 that directly binds to the promoter regions of several inflammation-associated genes. Therefore, regulation of PPARγ-Egr-1-pro-inflammatory mediators axis by PPARγ agonist Rosiglitazone may represent a novel strategy for blockade of acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Citocinas/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Inmunoglobulina G/metabolismo , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , PPAR gamma/metabolismo , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/prevención & control , Animales , Antiinflamatorios/farmacología , Complejo Antígeno-Anticuerpo/inmunología , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/agonistas , PPAR gamma/genética , Células RAW 264.7 , Rosiglitazona/farmacología , Transducción de Señal
19.
Mol Immunol ; 111: 95-105, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31048100

RESUMEN

During acute lung injury, a large number of monocytes are recruited into the pulmonary tissue, which is mainly mediated by local production of monocyte chemotactic protein 1 (MCP-1). As an essential component of the lung tissues, alveolar type II epithelial cells are one of the major sources of MCP-1. Therefore, uncovering the mechanism whereby MCP-1 production is regulated in the alveolar type II cells will provide a pivotal theoretical basis for clinical intervention in acute lung injury. In the current study, we find that there is a κB binding site in the MCP-1 promoter region, and mutation of the site leads to reduced production of MCP-1 in alveolar type II epithelial cells. In contrast, overexpression of NF-κB p65 significantly increases MCP-1 expression. Furthermore, we elucidate that IKKα/ß-NF-κB p65 signaling pathway and phosphorylation of serine 534 in NF-κB p65 are required for the maximal expression of MCP-1. Also, Activator protein 1 (AP-1) site in the promoter region and JNK1/2-c-Jun signaling are required for MCP-1 generation in alveolar type II epithelial cells. Moreover, a CCAAT/enhancer-binding protein (C/EBP) element is identified in the MCP-1 promoter region through the point mutation technique, and further experiments demonstrate that both C/EBPß and C/EBPδ are involved in basic and IL-1ß-mediated MCP-1 expression. Of note, specificity protein 1-Sp1 expression is not changed in alveolar type II epithelial cells incubated with IL-1ß, but it still control MCP-1 production by binding to the consensus sequence in the promoter region. More importantly, we find that the results derived from the cell line-MLE-12 cells and primary cells are consistent. Taken together, our data provide insights into the molecular mechanism how MCP-1 expression in inflammatory alveolar type II epithelial cells is regulated at transcription level.


Asunto(s)
Quimiocina CCL2/genética , Células Epiteliales/metabolismo , Interleucina-1beta/genética , Factores de Transcripción/genética , Animales , Línea Celular , Quinasa I-kappa B/genética , Inflamación/genética , Ratones , FN-kappa B/genética , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética , Factor de Transcripción ReIA/genética , Transcripción Genética/genética
20.
Mol Immunol ; 101: 585-596, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29887504

RESUMEN

Although participation of IL-6 in lung inflammation has been widely elucidated, the transcriptional regulation of its generation in alveolar type II cells stimulated by TNF-α remain unclear. Here, we find that TNF-α significantly induces IL-6 production, and TNF-α induction of IL-6 is mainly regulated at transcriptional level. Upon stimulated by TNF-α, Activator Protein-1 (AP-1)-mediated transcriptional activity is apparently increased in alveolar type II epithelial cells, which might be derived from elevated phosphorylation of JNK and subsequent activation of c-Jun. Either down-regulation of c-Jun or the AP-1 site mutation leads to significant reduction of IL-6 expression. In contrast, ectopic expression of c-Jun notably increases IL-6 generation. So, c-Jun, one of the AP-1 family members, plays a pivotal role in TNF-α-induced IL-6 generation. CCAAT/enhancer binding protein δ (C/EBPδ) expression is significantly amplified by TNF-α, which may contribute to the rise of C/EBP activity in alveolar type II cells. C/EBPδ shRNA treatment results in attenuation of IL-6 expression in the cells, which is consistent with data by introduction of mutations into the C/EBP site in the promoter. However, overexpression of C/EBPδ greatly increases the IL-6 promoter activity. In addition, data regarding another transactivator in the family-C/EBPß show that it does not affect IL-6 production. We also find that the IKK/NF-κB p65 pathway is activated in TNF-α-treated alveolar type II epithelial cells, and plays an essential role in positive regulation of IL-6 expression in TNF-α-treated alveolar type II epithelial cells via knockdown or forced expression of NF-κB p65, or elimination of κB sites in the IL-6 promoter. Notably, IL-6 promoter-driven luciferase production in primary alveolar type II epithelial cells can also be increased by the ectopic expression of c-Jun, C/EBPδ, and NF-κB p65, respectively. Collectively, our data provide insights into molecular mechanism involved in IL-6 expression in alveolar type II epithelial cells on TNF-α treatment, which provides a theoretical basis for specific inhibition of IL-6 production at the transcriptional level.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Quinasa I-kappa B/metabolismo , Interleucina-6/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Sitios de Unión , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA