Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 33(12): 14354-14369, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31661644

RESUMEN

Targeting activated fibroblasts, including myofibroblast differentiation, has emerged as a key therapeutic strategy in patients with idiopathic pulmonary fibrosis (IPF). However, there is no available therapy capable of selectively eradicating myofibroblasts or limiting their genesis. Through an integrative analysis of the regulator genes that are responsible for the activation of IPF fibroblasts, we noticed the phosphatidylinositol 4,5-bisphosphate (PIP2)-binding protein, myristoylated alanine-rich C-kinase substrate (MARCKS), as a potential target molecule for IPF. Herein, we have employed a 25-mer novel peptide, MARCKS phosphorylation site domain sequence (MPS), to determine if MARCKS inhibition reduces pulmonary fibrosis through the inactivation of PI3K/protein kinase B (AKT) signaling in fibroblast cells. We first observed that higher levels of MARCKS phosphorylation and the myofibroblast marker α-smooth muscle actin (α-SMA) were notably overexpressed in all tested IPF lung tissues and fibroblast cells. Treatment with the MPS peptide suppressed levels of MARCKS phosphorylation in primary IPF fibroblasts. A kinetic assay confirmed that this peptide binds to phospholipids, particularly PIP2, with a dissociation constant of 17.64 nM. As expected, a decrease of phosphatidylinositol (3,4,5)-trisphosphate pools and AKT activity occurred in MPS-treated IPF fibroblast cells. MPS peptide was demonstrated to impair cell proliferation, invasion, and migration in multiple IPF fibroblast cells in vitro as well as to reduce pulmonary fibrosis in bleomycin-treated mice in vivo. Surprisingly, we found that MPS peptide decreases α-SMA expression and synergistically interacts with nintedanib treatment in IPF fibroblasts. Our data suggest MARCKS as a druggable target in pulmonary fibrosis and also provide a promising antifibrotic agent that may lead to effective IPF treatments.-Yang, D. C., Li, J.-M., Xu, J., Oldham, J., Phan, S. H., Last, J. A., Wu, R., Chen, C.-H. Tackling MARCKS-PIP3 circuit attenuates fibroblast activation and fibrosis progression.


Asunto(s)
Fibroblastos/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/metabolismo , Fosfatidilinositoles/metabolismo , Fibrosis Pulmonar/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Proliferación Celular , Células Cultivadas/efectos de los fármacos , Células Cultivadas/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/genética , Fosfatidilinositoles/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/inducido químicamente
2.
Immunology ; 154(3): 500-509, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29377102

RESUMEN

Cholera toxin (CT) is a bacterial component that increases intracellular cAMP levels in host cells and suppresses T-cell activation. Recently, CT was reported to induce T helper type 17-skewing dendritic cells and activate interleukin-17A (IL-17A) production in CD4+ T cells through a cAMP-dependent pathway. However, the underlying mechanism by which cAMP regulates IL-17A production in T cells is not completely defined. In this study, we took advantage of a small molecule protein kinase A (PKA) inhibitor (H89) and different cAMP analogues: a PKA-specific activator (N6-benzoyl-adenosine-cAMP), an exchange protein activated by cAMP-specific activator (Rp-8-chlorophenylthio-2'-O-methyl cAMP), and a PKA inhibitor (Rp-8-bromo-cAMP), to elucidate the signalling cascade of cAMP in IL-17A regulation in T cells. We found that CT induced IL-17A production and IL-17A promoter activity in activated CD4+ T cells through a cAMP/PKA pathway. Moreover, this regulation was via cAMP-response element binding protein (CREB) -mediated transcriptional activation by using the transfection of an IL-17A promoter-luciferase reporter construct and CREB small interfering RNA in Jurkat cells. Also, we showed that CREB bound to the CRE motif located at -183 of the IL-17A promoter in vitro. Most interestingly, not only in CD4+ T cells, CT also enhanced cAMP/PKA-dependent IL-17A production and CREB phosphorylation in CD8+ T cells. In conclusion, our data suggest that CT induces an IL-17A-dominated immune microenvironment through the cAMP/PKA/CREB signalling pathway. Our study also highlights the potentials of CT and cAMP in modulating T helper type 17 responses in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Toxina del Cólera/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Interleucina-17/biosíntesis , Interleucina-17/genética , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Interferencia de ARN , Transducción de Señal , Activación Transcripcional
3.
Int J Mol Sci ; 18(11)2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-29125538

RESUMEN

Cigarette smoke has been shown to trigger aberrant signaling pathways and pathophysiological processes; however, the regulatory mechanisms underlying smoke-induced gene expression remain to be established. Herein, we observed that two smoke-responsive genes, HO-1 and CYP1A1, are robustly induced upon smoke by different mechanisms in human bronchial epithelia. CYP1A1 is mediated by aryl hydrocarbon receptor signaling, while induction of HO-1 is regulated by oxidative stress, and suppressed by N-acetylcysteine treatment. In light of a pivotal role of NRF2 and BACH1 in response to oxidative stress and regulation of HO-1, we examined if smoke-induced HO-1 expression is modulated through the NRF2/BACH1 axis. We demonstrated that smoke causes significant nuclear translocation of NRF2, but only a slight decrease in nuclear BACH1. Knockdown of NRF2 attenuated smoke-induced HO-1 expression while down-regulation of BACH1 had stimulatory effects on both basal and smoke-induced HO-1 with trivial influence on NRF2 nuclear translocation. Chromatin immunoprecipitation assays showed that smoke augments promoter-specific DNA binding of NRF2 but suppresses BACH1 binding to the HO-1 promoter ARE sites, two of which at -1.0 kb and -2.6 kb are newly identified. These results suggest that the regulation of NRF2 activator and BACH1 repressor binding to the ARE sites are critical for smoke-mediated HO-1 induction.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Hemo-Oxigenasa 1/genética , Factor 2 Relacionado con NF-E2/genética , Fumar/genética , Bronquios/metabolismo , Bronquios/patología , Línea Celular , Núcleo Celular/genética , Citocromo P-450 CYP1A1/genética , Proteínas de Unión al ADN/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica/genética , Humanos , Estrés Oxidativo/genética , Regiones Promotoras Genéticas , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética , Fumar/patología
4.
Crit Rev Immunol ; 35(2): 135-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26351147

RESUMEN

Numerous studies have shown that TH17 cells and their signature cytokine IL-17A are critical to host defense against various bacterial and fungal infections. The protective responses mediated by TH17 cells and IL-17A include the recruitment of neutrophils, release of antimicrobial peptides and chemokines, and enhanced tight junction of epithelial cells. Due to the importance of TH17 cells in infections, efforts have been made to develop TH17-based vaccines. The goal of vaccination is to establish a protective immunological memory. Most currently approved vaccines are antibody-based and have limited protection against stereotypically different strains. Studies show that T-cell-based vaccines may overcome this limitation and protect hosts against infection of different strains. Two main strategies are used to develop TH17 vaccines: identification of TH17-specific antigens and TH17-skewing adjuvants. Studies have revealed that cholera toxin (CT) induces a potent Th17 response following vaccination. Antigen vaccination along with CT induces a robust TH17 response, which is sometimes accompanied by TH1 responses. Due to the toxicity of CT, it is hard to apply CT in a clinical setting. Thus, understanding how CT modulates TH17 responses may lead to the development of successful TH17-based vaccines.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Toxina del Cólera/inmunología , Células Th17/citología , Células Th17/inmunología , Vacunas/inmunología , Animales , Diferenciación Celular , Toxina del Cólera/administración & dosificación , AMP Cíclico/metabolismo , Células Dendríticas/inmunología , Humanos , Infecciones/inmunología , Interleucina-17/inmunología , Vacunas/administración & dosificación
5.
Am J Respir Cell Mol Biol ; 53(5): 689-702, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25874372

RESUMEN

Statins are widely used to prevent cardiovascular disease. In addition to their inhibitory effects on cholesterol synthesis, statins have beneficial effects in patients with sepsis and pneumonia, although molecular mechanisms have mostly remained unclear. Using human airway epithelial cells as a proper in vitro model, we show that prior exposure to physiological nanomolar serum concentrations of simvastatin (ranging from 10-1,000 nM) confers significant cellular resistance to the cytotoxicity of pneumolysin, a pore-forming toxin and the main virulence factor of Streptococcus pneumoniae. This protection could be demonstrated with a different statin, pravastatin, or on a different toxin, α-hemolysin. Furthermore, through the use of gene silencing, pharmacological inhibitors, immunofluorescence microscopy, and biochemical and metabolic rescue approaches, we demonstrate that the mechanism of protection conferred by simvastatin at physiological nanomolar concentrations could be different from the canonical mevalonate pathways seen in most other mechanistic studies conducted with statins at micromolar levels. All of these data are integrated into a protein synthesis-dependent, calcium-dependent model showing the interconnected pathways used by statins in airway epithelial cells to elicit an increased resistance to pore-forming toxins. This research fills large gaps in our understanding of how statins may confer host cellular protection against bacterial infections in the context of airway epithelial cells without the confounding effect from the presence of immune cells. In addition, our discovery could be potentially developed into a host-centric strategy for the adjuvant treatment of pore-forming toxin associated bacterial infections.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Células Epiteliales/efectos de los fármacos , Proteínas Hemolisinas/antagonistas & inhibidores , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inmunidad Innata/efectos de los fármacos , Simvastatina/farmacología , Estreptolisinas/antagonistas & inhibidores , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Línea Celular Transformada , Células Epiteliales/inmunología , Células Epiteliales/patología , Proteínas Hemolisinas/toxicidad , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/inmunología , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Pravastatina/inmunología , Pravastatina/farmacología , Cultivo Primario de Células , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Simvastatina/inmunología , Staphylococcus aureus/química , Streptococcus pneumoniae/química , Estreptolisinas/toxicidad
6.
J Immunol ; 191(8): 4095-102, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043897

RESUMEN

The significance of Th17 cells and IL-17A signaling in host defense and disease development has been demonstrated in various infection and autoimmune models. Additionally, the generation of Th17 cells is highly influenced by microbes. However, the specific bacterial components capable of shaping Th17 responses have not been well defined. The goals of this study were to understand how a bacterial toxin, cholera toxin (CT), modulates Th17-dominated response in isolated human CD4(+) T cells, and what are the mechanisms associated with this modulation. CD4(+) cells isolated from human peripheral blood were treated with CT. The levels of cytokine production and specific Th cell responses were determined by ELISA, Luminex assay, and flow cytometry. Along with the decreased production of other proinflammatory cytokines (IFN-γ, TNF-α, and IL-2), we found that CT could directly enhance the IL-17A production through a cAMP-dependent pathway. This enhancement is specific for IL-17A but not for IL-17F, IL-22, and CCL20. Interestingly, CT could increase IL-17A production only from Th17-committed cells, such as CCR6(+)CD4(+) T cells and in vitro-differentiated Th17 cells. Furthermore, we also demonstrated that this direct effect occurs at a transcriptional level because CT stimulates the reporter activity in Jurkat and primary CD4(+) T cells transfected with the IL-17A promoter-reporter construct. This study shows that CT has the capacity to directly shape Th17 responses in the absence of APCs. Our findings highlight the potentials of bacterial toxins in the regulation of human Th17 responses.


Asunto(s)
Toxina del Cólera/metabolismo , Interleucina-17/biosíntesis , Células Th17/metabolismo , Células Cultivadas , Quimiocina CCL20/metabolismo , AMP Cíclico/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucinas/metabolismo , Receptores CCR6/metabolismo , Transducción de Señal , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-22
7.
Am J Respir Crit Care Med ; 190(10): 1127-38, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25318062

RESUMEN

RATIONALE: Phosphorylation of myristoylated alanine-rich C kinase substrate (phospho-MARCKS) at the phosphorylation site domain (PSD) is crucial for mucus granule secretion and cell motility, but little is known concerning its function in lung cancer. OBJECTIVES: We aimed to determine if MARCKS PSD activity can serve as a therapeutic target and to elucidate the molecular basis of this potential. METHODS: The clinical relevance of phospho-MARCKS was first confirmed. Next, we used genetic approaches to verify the functionality and molecular mechanism of phospho-MARCKS. Finally, cancer cells were pharmacologically inhibited for MARCKS activity and subjected to functional bioassays. MEASUREMENTS AND MAIN RESULTS: We demonstrated that higher phospho-MARCKS levels were correlated with shorter overall survival of lung cancer patients. Using shRNA silencing and ectopic expression of wild-type and PSD-mutated (S159/163A) MARCKS, we showed that elevated phospho-MARCKS promoted cancer growth and erlotinib resistance. Further studies demonstrated an interaction of phosphoinositide 3-kinase with MARCKS, but not with phospho-MARCKS. Interestingly, phospho-MARCKS acted in parallel with increased phosphatidylinositol (3,4,5)-triphosphate pools and AKT activation in cells. Through treatment with a 25-mer peptide targeting the MARCKS PSD motif (MPS peptide), we were able to suppress tumor growth and metastasis in vivo, and reduced levels of phospho-MARCKS, phosphatidylinositol (3,4,5)-triphosphate, and AKT activity. This peptide also enhanced the sensitivity of lung cancer cells to erlotinib treatment, especially those with sustained activation of phosphoinositide 3-kinase/AKT signaling. CONCLUSIONS: These results suggest a key role for MARCKS PSD in cancer disease and provide a unique strategy for inhibiting the activity of MARCKS PSD as a treatment for lung cancer.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas de la Membrana/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Modelos Animales de Enfermedad , Clorhidrato de Erlotinib , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Transducción de Señal/fisiología
8.
Am J Respir Cell Mol Biol ; 50(1): 30-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23944933

RESUMEN

IL-17A, IL-17F, and IL-25 belong to the IL-17 family of cytokines, and are well known to play important roles in the host defense against infection and inflammatory diseases. IL-17C, also a member of the IL-17 family, is highly expressed in the epithelium; however, the function and regulatory mechanism of IL-17C in airway epithelium remain poorly understood. In this study, we demonstrate that polyinosinic-polycytidylic acid (polyI:C), the ligand to Toll-like receptor 3, is a potent inducer of IL-17C mRNA and protein expression in primary normal human bronchial epithelial (NHBE) cells. IL-17C induction by polyI:C was both time dependent and dose dependent, and was attenuated by inhibitors of the Toll-IL-1 receptor domain-containing adaptor-inducing INF-ß (TRIF)-NF-κB pathway, Pepinh-TRIF, BAY11, NF-κB inhibitor III, and NF-κB p65 small interfering RNA, suggesting that IL-17C expression is induced by polyI:C via the Toll-like receptor 3-TRIF-NF-κB pathway. Both IL-17C and polyI:C increased the expression of antimicrobial peptides and proinflammatory cytokines, such as human ß-defensin (hBD) 2, colony-stimulating factor 3 (CSF3), and S100A12 in NHBE cells. Knockdown of IL-17 receptor (IL-17R) E, the specific receptor for IL-17C, using IL-17RE small interfering RNA, attenuated polyI:C-induced hBD2, CSF3, and S100A12 expression, without any reduction of polyI:C-induced IL-17C expression, which suggest that IL-17C enhances hBD2, CSF, and S100A12 expression in an autocrine/paracrine manner in NHBE cells. Knockdown of IL-17C also decreased polyI:C-induced hBD2, CSF3, and S100A12 expression. Thus, our data demonstrate that IL-17C is an essential epithelial cell-derived cytokine that enhances mucosal host defense responses in a unique autocrine/paracrine manner in the airway epithelium.


Asunto(s)
Comunicación Autocrina/fisiología , Bronquios/metabolismo , Interleucina-17/metabolismo , Comunicación Paracrina/fisiología , Mucosa Respiratoria/metabolismo , Receptor Toll-Like 3/metabolismo , Comunicación Autocrina/inmunología , Bronquios/inmunología , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Interleucina-17/inmunología , Comunicación Paracrina/inmunología , Poli I-C/inmunología , Poli I-C/metabolismo , Receptores del Factor Estimulante de Colonias/inmunología , Receptores del Factor Estimulante de Colonias/metabolismo , Mucosa Respiratoria/inmunología , Proteínas S100/inmunología , Proteínas S100/metabolismo , Proteína S100A12 , Receptor Toll-Like 3/inmunología , beta-Defensinas/inmunología , beta-Defensinas/metabolismo
9.
Eur J Pharmacol ; 980: 176867, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39111683

RESUMEN

BACKGROUND: MARCKS (myristoylated alanine-rich C kinase substrates) serves as a substrate for protein kinase C, residing in the plasma membrane while acts as an actin filament crosslinking protein. This investigation aims to elucidate phosphorylated MARCKS (p-MARCKS) levels and activity in allergic asthma patients and explore the therapeutic potential of peptide inhibitors targeting p-MARCKS in an acute mouse model of allergic asthma. METHODS: Immunohistochemistry and histology staining were employed on lung tissue slides to evaluate p-MARCKS expression and allergic asthma symptoms. Airway resistance was measured using invasive whole-body plethysmography. Flow cytometry detected lung dendritic cell migration, and migration/maturation assays were conducted on isolated murine bone marrow-derived dendritic cells (BM-DCs). RESULTS: Elevated p-MARCKS expression was observed in both human asthmatic tissues and animal models immunized with ovalbumin or Alternaria alternata. Remarkably, asthmatic individuals showed elevated high p-MARCKS expression in lung tissues. Intraperitoneal injection of the peptide MPS, targeting the MARCKS phosphorylation site domain, before allergen challenged, effectively suppressed MARCKS phosphorylation in murine lung tissues. MPS inhibited both in vivo and in vitro migration and maturation of dendritic cells (BM-DCs) and reduced Th2-related lymphocyte activation in bronchoalveolar lavage fluid (BALF). MPS pretreatment additionally suppressed all symptoms associated with allergic airway asthma, including a reduction in inflammatory cell influx, airway mucous cell metaplasia, and airway hyperreactivity. CONCLUSION: These findings suggest that phosphorylated MARCKS occurs in asthmatic lung tissue, and the inhibition of MARCKS phosphorylation by the MPS peptide reduces dendritic cell migration and Th2-related lymphocytes in the lungs in a murine model of acute asthma.


Asunto(s)
Asma , Movimiento Celular , Células Dendríticas , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Animales , Femenino , Humanos , Masculino , Ratones , Enfermedad Aguda , Asma/inmunología , Asma/tratamiento farmacológico , Asma/patología , Asma/metabolismo , Movimiento Celular/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/inmunología , Pulmón/efectos de los fármacos , Ratones Endogámicos BALB C , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/metabolismo , Péptidos/farmacología , Fosforilación
10.
Am J Respir Cell Mol Biol ; 49(2): 204-11, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23672216

RESUMEN

The incidence of lung diseases and cancer caused by cigarette smoke is increasing. The molecular mechanisms of gene regulation induced by cigarette smoke that ultimately lead to cancer remain unclear. This report describes a novel long noncoding RNA (lncRNA) that is induced by cigarette smoke extract (CSE) both in vitro and in vivo and is elevated in numerous lung cancer cell lines. We have termed this lncRNA the smoke and cancer-associated lncRNA-1 (SCAL1). This lncRNA is located in chromosome 5, and initial sequencing analysis reveals a transcript with four exons and three introns. The expression of SCAL1 is regulated transcriptionally by nuclear factor erythroid 2-related factor (NRF2), as determined by the small, interfering RNA (siRNA) knockdown of NRF2 and kelch-like ECH-associated protein 1 (KEAP1). A nuclear factor erythroid-derived 2 (NF-E2) motif was identified in the promoter region that shows binding to NRF2 after its activation. Functionally, the siRNA knockdown of SCAL1 in human bronchial epithelial cells shows a significant potentiation of cytotoxicity induced by CSE in vitro. Altogether, these results identify a novel and intriguing new noncoding RNA that may act downstream of NRF2 to regulate gene expression and mediate oxidative stress protection in airway epithelial cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Fumar/metabolismo , Línea Celular Tumoral , Cromosomas Humanos Par 5/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteína 1 Asociada A ECH Tipo Kelch , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Masculino , Subunidad p45 del Factor de Transcripción NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Proteínas de Neoplasias/biosíntesis , Fumar/efectos adversos , Fumar/patología
11.
Clin Dev Immunol ; 2013: 267971, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23956759

RESUMEN

The significance of Th17 cells and interleukin- (IL-)17A signaling in host defense and disease development has been demonstrated in various infection and autoimmune models. Numerous studies have indicated that Th17 cells and its signature cytokine IL-17A are critical to the airway's immune response against various bacteria and fungal infection. Cytokines such as IL-23, which are involved in Th17 differentiation, play a critical role in controlling Klebsiella pneumonia (K. pneumonia) infection. IL-17A acts on nonimmune cells in infected tissues to strengthen innate immunity by inducing the expression of antimicrobial proteins, cytokines, and chemokines. Mice deficient in IL-17 receptor (IL-17R) expression are susceptible to infection by various pathogens. In this review, we summarize the recent advances in unraveling the mechanism behind Th17 cell differentiation, IL-17A/IL-17R signaling, and also the importance of IL-17A in pulmonary infection.


Asunto(s)
Interleucina-17/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Neumonía/genética , Neumonía/microbiología , Receptores de Interleucina-17/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Transducción de Señal , Células Th17/citología
12.
Nat Commun ; 14(1): 8393, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110369

RESUMEN

Ferroptosis is an iron-dependent programmed cell death associated with severe kidney diseases, linked to decreased glutathione peroxidase 4 (GPX4). However, the spatial distribution of renal GPX4-mediated ferroptosis and the molecular events causing GPX4 reduction during ischemia-reperfusion (I/R) remain largely unknown. Using spatial transcriptomics, we identify that GPX4 is situated at the interface of the inner cortex and outer medulla, a hyperactive ferroptosis site post-I/R injury. We further discover OTU deubiquitinase 5 (OTUD5) as a GPX4-binding protein that confers ferroptosis resistance by stabilizing GPX4. During I/R, ferroptosis is induced by mTORC1-mediated autophagy, causing OTUD5 degradation and subsequent GPX4 decay. Functionally, OTUD5 deletion intensifies renal tubular cell ferroptosis and exacerbates acute kidney injury, while AAV-mediated OTUD5 delivery mitigates ferroptosis and promotes renal function recovery from I/R injury. Overall, this study highlights a new autophagy-dependent ferroptosis module: hypoxia/ischemia-induced OTUD5 autophagy triggers GPX4 degradation, offering a potential therapeutic avenue for I/R-related kidney diseases.


Asunto(s)
Lesión Renal Aguda , Ferroptosis , Daño por Reperfusión , Humanos , Riñón , Autofagia , Isquemia
13.
Respir Res ; 13: 38, 2012 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-22583375

RESUMEN

BACKGROUND: Asthma causes significant morbidity worldwide in adults and children alike, and incurs large healthcare costs. The statin drugs, which treat hyperlipidemia and cardiovascular diseases, have pleiotropic effects beyond lowering cholesterol, including immunomodulatory, anti-inflammatory, and anti-fibrotic properties which may benefit lung health. Using an allergic mouse model of asthma, we previously demonstrated a benefit of statins in reducing peribronchiolar eosinophilic inflammation, airway hyperreactivity, goblet cell hyperplasia, and lung IL-4 and IL-13 production. OBJECTIVES: In this study, we evaluated whether simvastatin inhibits IL-13-induced pro-inflammatory gene expression of asthma-related cytokines in well-differentiated primary mouse tracheal epithelial (MTE) cell cultures. We hypothesized that simvastatin reduces the expression of IL-13-inducible genes in MTE cells. METHODS: We harvested tracheal epithelial cells from naïve BALB/c mice, grew them under air-liquid interface (ALI) cell culture conditions, then assessed IL-13-induced gene expression in MTE cells using a quantitative real-time PCR mouse gene array kit. RESULTS: We found that simvastatin had differential effects on IL-13-mediated gene expression (inhibited eotaxin-1; MCP-1,-2,-3; and osteopontin (SPP1), while it induced caspase-1 and CCL20 (MIP-3α)) in MTE cells. For other asthma-relevant genes such as TNF, IL-4, IL-10, CCL12 (MCP-5), CCL5 (RANTES), and CCR3, there were no significant IL-13-inducible or statin effects on gene expression. CONCLUSIONS: Simvastatin modulates the gene expression of selected IL-13-inducible pro-inflammatory cytokines and chemokines in primary mouse tracheal epithelial cells. The airway epithelium may be a viable target tissue for the statin drugs. Further research is needed to assess the mechanisms of how statins modulate epithelial gene expression.


Asunto(s)
Citocinas/biosíntesis , Células Epiteliales/metabolismo , Interleucina-13/fisiología , Mucosa Respiratoria/metabolismo , Simvastatina/farmacología , Tráquea/metabolismo , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Tráquea/citología , Tráquea/efectos de los fármacos
14.
Cancers (Basel) ; 14(19)2022 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-36230850

RESUMEN

The myristoylated alanine-rich C-kinase substrate (MARCKS) is a membrane-associated protein kinase C (PKC) substrate ubiquitously expressed in eukaryotic cells. MARCKS plays important roles in multiple cellular processes, including cell adhesion and motility, mucin secretion, exocytosis, and inflammatory response. Aberrant MARCKS signaling has been observed in the development and progression of multiple cancer types. In addition, MARCKS facilitates cancer metastasis through modulating cancer cell migration and invasion. Moreover, MARCKS contributes to treatment resistance, likely by promoting cancer stem cell renewal as well as immunosuppression. In this review, we describe MARCKS protein structure, cellular localization, and biological functions. We then discuss the role of MARCKS in cancer metastasis as well as its mechanisms of action in solid tumors. Finally, we review recent advances in targeting MARCKS as a new therapeutic strategy in cancer management.

15.
Am J Respir Cell Mol Biol ; 45(2): 270-6, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20971882

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent environmental toxicant. Epidemiological studies have associated TCDD exposure with the development of chronic obstructive pulmonary disease, which is manifested by mucous/goblet cell hyperplasia. The purpose of this research was to elucidate the pathway/mechanisms that lead to TCDD-induced gene expression in both primary normal human bronchial epithelial cells and an immortalized cell line, HBE1, under air-liquid interface conditions. TCDD exposure induced a time-dependent elevation of MUC5AC mRNA and protein synthesis, and cytochrome p450 1A1 (CYP1A1) expression in these cells. Treatment with an aryl hydrocarbon receptor antagonist had no effect on TCDD-induced MUC5AC expression, but significantly suppressed CYP1A1 induction. However, treatments with inhibitors of signaling pathways and the expression of dominant negative mutants of epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK) and p38, but not the inhibition of c-Jun N-terminal kinase pathway, abrogated MUC5AC induction, but not that of CYP1A1. These effects also occurred at the MUC5AC promoter-reporter level using the chimeric construct for a transient transfection study. Western blot analysis confirmed the phosphorylation of activated EGFR, ERK, and p38 signaling molecules, but not the c-Jun N-terminal kinase, in cells after TCDD exposure. Specificity protein 1 (Sp1) phosphorylation also occurred in cells after TCDD exposure. Both MUC5AC expression and the promoter activity were inhibited by mithramycin A, an inhibitor specific to Sp1-based transcription. These results lead to the conclusion that TCDD induced MUC5AC expression through a noncanonical aryl hydrocarbon receptor-independent, EGFR/ERK/p38-mediated signaling pathway-mediated/Sp1-based transcriptional mechanism.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Citocromo P-450 CYP1A1/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mucina 5AC/genética , Dibenzodioxinas Policloradas/farmacología , Receptores Inmunológicos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Biomarcadores de Tumor/genética , Western Blotting , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Células Cultivadas , Citocromo P-450 CYP1A1/metabolismo , Contaminantes Ambientales/farmacología , Inhibidores Enzimáticos/farmacología , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Humanos , Mucina 5AC/metabolismo , ARN Mensajero/genética , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
16.
Am J Respir Cell Mol Biol ; 45(2): 246-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20935193

RESUMEN

A major pathological feature of chronic airway diseases is the elevated expression of gel-forming mucins. NF-κB activation in airway epithelial cells has been shown to play a proinflammatory role in chronic airway diseases; however, the specific role of NF-κB in mucin gene expression has not been characterized. In this study, we show that the proinflammatory cytokines, IL-1ß and IL-17A, both of which use the NF-κB pathway, are potent inducers of MUC5B mRNA expression in both well differentiated primary normal human bronchial epithelial cells and the human bronchial epithelial cell line, HBE1. MUC5B induction by these cytokines was both time- and dose-dependent, and was attenuated by the small molecule inhibitor, NF-κB inhibitor III, as well as p65 small interfering RNA, suggesting that the regulation of MUC5B expression by these cytokines is via an NF-κB-based transcriptional mechanism. Deletion analysis of the MUC5B promoter demonstrated that IL-1ß- and IL-17A-induced promoter activity resides within the -4.17-kb to -2.56-kb region relative to the transcriptional start site. This region contains three putative κB-binding sites (NF-κB-1, -3,786/-3,774; NF-κB-2, -3,173/-3,161; and NF-κB-3, -2,921/-2,909). Chromatin immunoprecipitation analysis confirmed enhanced binding of the p50 NF-κB subunit to the NF-κB-3 site after cytokine stimulation. We conclude that an NF-κB-based transcriptional mechanism is involved in MUC5B regulation by IL-1ß and IL-17A in airway epithelium. This is the first demonstration of the participation of NF-κB and its specific binding site in cytokine-mediated airway MUC5B expression.


Asunto(s)
Bronquios/metabolismo , Regulación de la Expresión Génica , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Mucina 5B/genética , Mucina 5B/metabolismo , FN-kappa B/metabolismo , Western Blotting , Bronquios/citología , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Humanos , Interleucina-17/genética , Interleucina-1beta/genética , Mucina 5B/antagonistas & inhibidores , FN-kappa B/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Sistema Respiratorio/citología , Sistema Respiratorio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Immunol ; 183(10): 6236-43, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19841186

RESUMEN

Mucin over-production is one of the hallmarks of chronic airway diseases such as chronic obstructive pulmonary disease, asthma, and cystic fibrosis. NF-kappaB activation in airway epithelial cells has been shown to play a positive inflammatory role in chronic airway diseases; however, the role of NF-kappaB in mucin gene expression is unresolved. In this study, we have shown that the proinflammatory cytokines, IL-1beta and IL-17A, both of which utilize the NF-kappaB pathway, are potent inducers of mucin (MUC)5AC mRNA and protein synthesis by both well-differentiated primary normal human bronchial epithelial cells and the human bronchial epithelial cell line, HBE1. MUC5AC induction by these cytokines was both time- and dose-dependent and occurred at the level of promoter activation, as measured by a reporter gene assay. These effects were attenuated by the small molecule inhibitor NF-kappaB inhibitor III, as well as p65 small-interfering RNA, suggesting that the regulation of MUC5AC expression by these cytokines is via an NF-kappaB-based transcriptional mechanism. Further investigation of the promoter region identified a putative NF-kappaB binding site at position-3594/-3582 in the promoter of MUC5AC as critical for the regulation of MUC5AC expression by both IL-1beta and IL-17A. Chromatin immunoprecipitation analysis confirmed enhanced binding of the NF-kappaB subunit p50 to this region following cytokine stimulation. We conclude that an NF-kappaB-based transcriptional mechanism is involved in MUC5AC regulation by IL-1beta and IL-17A in the airway epithelium. This is the first demonstration of the participation of NF-kappaB and its specific binding site in cytokine-mediated airway MUC5AC expression.


Asunto(s)
Bronquios/inmunología , Mucina 5AC/metabolismo , FN-kappa B/metabolismo , Mucosa Respiratoria/inmunología , Factor de Transcripción ReIA/metabolismo , Bronquios/efectos de los fármacos , Línea Celular , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Humanos , Interleucina-17/farmacología , Interleucina-1beta/farmacología , Mucina 5AC/agonistas , Mucina 5AC/genética , Mucina 5AC/inmunología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/inmunología , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/inmunología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/farmacología , Mucosa Respiratoria/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Transfección
19.
Am J Vet Res ; 71(6): 690-6, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20513186

RESUMEN

OBJECTIVE: To isolate and culture primary equine airway epithelial cells in vitro and elucidate the major cytokines involved in expression of the gel-forming mucin gene MUC5AC in horses. SAMPLE POPULATION: 12 tracheas obtained within 5 hours after euthanasia from horses free from respiratory tract disease. PROCEDURES: Tracheal rings were digested overnight in 0.2% protease, and dissociated airway epithelial cells were grown in a serum-free defined medium at an air-liquid interface until confluence was achieved. Differentiated airway epithelial cells were treated with a panel of recombinant equine cytokines followed by quantitative reverse transcriptase PCR assay for mRNA of equine MUC5AC and the control gene glyceraldehyde 3-phosphate dehydrogenase. Cultures were incubated in the presence of isohelenin, a nuclear factor kappaB-DNA-binding inhibitor, to investigate transcriptional regulation of MUC5AC. RESULTS: Light and electron microscopy revealed a differentiated epithelium with ciliated cells, nonciliated mucous cells, and basal-like cells. Recombinant equine tumor necrosis factor-alpha was the major mediator in the cytokine panel that significantly increased MUC5AC mRNA by a factor of 5 in a dose- and time-dependent manner. This enhancement was attenuated by isohelenin. CONCLUSIONS AND CLINICAL RELEVANCE: Data suggested that a nuclear factor KB-based transcriptional mechanism is involved in induction of MUC5AC expression by tumor necrosis factor-A. Understanding the molecular mechanism of cytokine-enhanced MUC5AC expression in horses may lead to better treatment options and understanding of the pathogenesis of equine pulmonary diseases.


Asunto(s)
Obstrucción de las Vías Aéreas/veterinaria , Enfermedades de los Caballos/metabolismo , Mucina 5AC/biosíntesis , Tráquea/metabolismo , Obstrucción de las Vías Aéreas/genética , Animales , Células Epiteliales , Enfermedades de los Caballos/genética , Caballos , Microscopía Electrónica de Rastreo/veterinaria , Mucina 5AC/genética , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Sesquiterpenos/farmacología , Tráquea/citología , Transcripción Genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
20.
Crit Rev Immunol ; 28(4): 269-79, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19166380

RESUMEN

IL-17 is a proinflammatory cytokine produced by immune cells. Its significance in host defense and disease development has been demonstrated in various infection and autoimmune models. Recently, additional studies have shown that IL-17 is also important in modulating airway immune response in several aspects. Along with the well-established Th1/Th2 model, new discoveries regarding the Th17 lineage and IL-17 functions have added an extra twist to the already complicated cytokine network that regulates airway immunity. The IL-17 receptor is expressed on blood cells, as well as on structural cells such as the epithelial cells in the airway. Therefore, the effect of IL-17 on airway immunity is very broad, covering both the innate and the adaptive aspects. In this review, we summarize the findings of recent studies on IL-17 signaling and function in pulmonary immune response, and the implications of IL-17 in disease pathogenesis.


Asunto(s)
Inmunidad Innata , Interleucina-17/fisiología , Pulmón/inmunología , Animales , Asma/inmunología , Infecciones Bacterianas/inmunología , Humanos , Receptores de Interleucina-17/análisis , Receptores de Interleucina-17/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA