Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Curr Mol Med ; 24(1): 123-133, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37138491

RESUMO

BACKGROUND: Extensive deposition of extracellular matrix (ECM) in idiopathic pulmonary fibrosis (IPF) is due to hyperactivation and proliferation of pulmonary fibroblasts. However, the exact mechanism is not clear. OBJECTIVE: This study focused on the role of CTBP1 in lung fibroblast function, elaborated its regulation mechanism, and analyzed the relationship between CTBP1 and ZEB1. Meanwhile, the antipulmonary fibrosis effect and its molecular mechanism of Toosendanin were studied. METHODS: Human IPF fibroblast cell lines (LL-97A and LL-29) and normal fibroblast cell lines (LL-24) were cultured in vitro. The cells were stimulated with FCS, PDGF-BB, IGF-1, and TGF-ß1, respectively. BrdU detected cell proliferation. The mRNA expression of CTBP1 and ZEB1 was detected by QRT-PCR. Western blotting was used to detect the expression of COL1A1, COL3A1, LN, FN, and α-SMA proteins. An animal model of pulmonary fibrosis was established to analyze the effects of CTBP1 silencing on pulmonary fibrosis and lung function in mice. RESULTS: CTBP1 was up-regulated in IPF lung fibroblasts. Silencing CTBP1 inhibits growth factor-driven proliferation and activation of lung fibroblasts. Overexpression of CTBP1 promotes growth factor-driven proliferation and activation of lung fibroblasts. Silencing CTBP1 reduced the degree of pulmonary fibrosis in mice with pulmonary fibrosis. Western blot, CO-IP, and BrdU assays confirmed that CTBP1 interacts with ZEB1 and promotes the activation of lung fibroblasts. Toosendanin can inhibit the ZEB1/CTBP1protein interaction and further inhibit the progression of pulmonary fibrosis. CONCLUSION: CTBP1 can promote the activation and proliferation of lung fibroblasts through ZEB1. CTBP1 promotes lung fibroblast activation through ZEB1, thereby increasing excessive deposition of ECM and aggravating IPF. Toosendanin may be a potential treatment for pulmonary fibrosis. The results of this study provide a new basis for clarifying the molecular mechanism of pulmonary fibrosis and developing new therapeutic targets.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Camundongos , Animais , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/farmacologia , Fibrose Pulmonar Idiopática/genética , Pulmão , Fibroblastos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
2.
Ecotoxicol Environ Saf ; 269: 115816, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38091678

RESUMO

Autophagy mediates PM2.5-related lung injury (LI) and is tightly linked to inflammation and apoptosis processes. IL-37 has been demonstrated to regulate autophagy. This research aimed to examine the involvement of IL-37 in the progression of PM2.5-related LI and assess whether autophagy serves as a mediator for its effects.To create a model of PM2.5-related LI, this research employed a nose-only PM2.5 exposure system and utilized both human IL-37 transgenic mice and wild-type mice. The hIL-37tg mice demonstrated remarkable reductions in pulmonary inflammation and pathological LI compared to the WT mice. Additionally, they exhibited activation of the AKT/mTOR signaling pathway, which served to regulate the levels of autophagy and apoptosis.Furthermore, in vitro experiments revealed a dose-dependent upregulation of autophagy and apoptotic proteins following exposure to PM2.5 DMSO extraction. Simultaneously, p-AKT and p-mTOR expression was found to decrease. However, pretreatment with IL-37 demonstrated a remarkable reduction in the levels of autophagy and apoptotic proteins, along with an elevation of p-AKT and p-mTOR. Interestingly, pretreatment with rapamycin, an autophagy inducer, weakened the therapeutic impact of IL-37. Conversely, the therapeutic impact of IL-37 was enhanced when treated with 3-MA, a potent autophagy inhibitor. Moreover, the inhibitory effect of IL-37 on autophagy was successfully reversed by administering AKT inhibitor MK2206. The findings suggest that IL-37 can inhibit both the inflammatory response and autophagy, leading to the alleviation of PM2.5-related LI. At the molecular level, IL-37 may exert its anti autophagy and anti apoptosis effects by activating the AKT/mTOR signaling pathway.


Assuntos
Lesão Pulmonar , Material Particulado , Proteínas Proto-Oncogênicas c-akt , Animais , Humanos , Camundongos , Autofagia/efeitos dos fármacos , Interleucinas/farmacologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/tratamento farmacológico , Material Particulado/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
Immunobiology ; 228(3): 152358, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37003140

RESUMO

Air pollution consisting of fine particulate matter (PM2.5) can induce or aggravate pulmonary inflammatory injury. Irisin has been shown to inhibit inflammation and help to protect against acute kidney, lung or brain injury. However, the role of irisin in lung inflammation after exposure to PM2.5 remains unclear. The aim of this study was to investigate the effect and molecular mechanism of irisin supplementation on in vitro and in vivo models of PM2.5-induced acute lung injury(ALI). C57BL/6 mice and alveolar macrophage cell line (MH-S) were treated with PM2.5. Histopathological examination and FNDC5/ irisin immunofluorescence staining was performed on lung tissue sections. MH-S cell viability was determined by CCK-8 assay. The levels of Nod2, NF-κB p65 and NLRP3 were detected by qRT-PCR and western blotting. The levels of cytokines (IL-1ß, IL-18 and TNF-α) were detected by ELISA. PM2.5 exposure induced increased secretion of pro-inflammatory factors and activation of Nod2, NF-κB p65 and NLRP3 as well as endogenous levels of irisin. In vivo and in vitro inflammation was alleviated by irisin supplementation. Irisin significantly decreased IL-1ß, IL-18, and TNF-α production at both mRNA and protein level. Expression levels of Nod2, NF-κB p65, and NLRP3 were all significantly affected by irisin. In vivo the degree of pulmonary injury and inflammatory infiltration was weakened after irisin administration. In vitro, irisin could inhibit the activation of the NLRP3 inflammasome for a sustained period of 24 h, and its inhibitory ability was gradually enhanced. In conclusion, our findings indicate that irisin can modulate the inflammatory injury of lung tissue caused by PM2.5 through the Nod2/NF-κB signaling pathway, suggesting that irisin can be a candidate for the therapeutic or preventive intervention in acute lung inflammation.


Assuntos
Lesão Pulmonar Aguda , Pneumonia , Camundongos , Animais , NF-kappa B/metabolismo , Material Particulado/efeitos adversos , Interleucina-18 , Fibronectinas/efeitos adversos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais , Inflamação/metabolismo
4.
Aging (Albany NY) ; 14(16): 6796-6808, 2022 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-36040387

RESUMO

OBJECTIVE: To explore the roles of macrophages' miR-217-5p in the process of PM2.5 induced acute lung injury. METHODS: GEO database and KEGG pathway enrichment analysis as well as GSEA were used to predicted the miRNA and associated target signals. And then mice and RAW246.7 macrophages treated with PM2.5 to imitate PM2.5 induced acute lung injury environment and then transfected with miR-217-5p NC or miR-217-5p mimic. The levels of inflammatory factors TNF-α and anti-inflammatory factor IL-10 of mice serum were tested by ELISA. And the pathological changes and ROS level of mouse lung tissues were stained by HE and DHE staining. The proteins expression of phosphorylated-STAT1, total-STAT1, TNF-α, IFN-γ as well as p47, gp91, NOX4 in mice or RAW264.7 cells were tested by western blot or immunofluorescence of RAW264.7 cell slides. RESULTS: The results of bioinformatics analysis indicated the miR-217 as well as STAT1 were involved PM2.5 associated lung injury. After exposure to PM2.5, the decreased levels of serum TNF-α but not IL-10, consistent with reduced macrophages' accumulation as well as decreased ROS levels in lung tissues in miR-217-5p mimic group vs miR-217-5p NC group mice, and moreover, the protein expression levels of phosphorylated--STAT1, total-STAT1, TNF-α, IFN-γ, p47, gp91 and NOX4 in mouse lung tissues and RTAW246.7 macrophages cells were all significantly reduced with miR-217-5p mimic administration. The above phenomena were reversed by specific STAT1-inhibitor HY-N8107. CONCLUSIONS: miR-217-5p suppressed the activated STAT1-signal induced inflammation and oxidative stress trigged by PM2.5 in macrophages and resulted in the decreased lung injure caused by PM2.5.


Assuntos
Lesão Pulmonar Aguda , MicroRNAs , Lesão Pulmonar Aguda/metabolismo , Animais , Inflamação/metabolismo , Pulmão/metabolismo , Macrófagos/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Smog , Fator de Necrose Tumoral alfa/metabolismo
5.
Int J Pediatr Otorhinolaryngol ; 159: 111205, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35700689

RESUMO

OBJECTIVES: Cuffed endotracheal intubation and stent implantation were employed to simulate two types of benign airway stenosis and further to analysis the different features between them from trachecscopic characteristics, gross anatomy to histopathological changes. In addition, our study explored the therapeutic effect of mitomycin C at different concentrations on granulation tissue caused by stent implantation in order to provide a new therapeutic strategy for clinical treatment of benign airway stenosis. METHODS: Twelve beagle dogs were randomly divided into four groups, with three dogs in each group. Group A: Three beagle dogs were intubated through oral trachea after general anesthesia and cuff pressure maintained at 200 mmHg for 24 h. Group B, Group C and Group D: endotracheal coated self-expanding metal stents were placed after general anesthesia under the guidance of bronchoscope. On the Day7 after stent implantation, Group B, as control group, was injected phosphate buffer solution of 1 ml into granulation tissue at the end of stent; Group C was injected mitomycin C of 1 ml at 0.4 mg/ml and Group D was injected mitomycin C of 1 ml at 0.8 mg/ml into granulation tissue at the end of metal airway stent respectively, the same method as Group B. Bronchoscopy was used to observe tracheal lumen on the seventh day, fourteenth day and twenty-first day after modeling and pathological changes were examined on twenty-first day. RESULTS: Two models of benign airway stenosis can be established by cuffed endotracheal intubation and stent implantation. There was tracheal rupture in the trachea cartiage ring in the cuffed endotracheal intubation group, but was't in stent implantation group. Histopathological characteristics were different between cuffed endotracheal intubation and stent implantation groups. In stent placement groups, we found that the stenosis degree of mitomycin C at 0.4 mg/ml was approximately 19%-32%, mitomycin C at 0.8 mg/ml was approximately 16%-21% and the control group was approximately 36%-47%. CONCLUSION: The two models of canine benign tracheal stenosis induced by cuffed endotracheal intubation and stent implantation are relatively simple, reliable and reproducible and have different characteristics. Mitomycin C could inhibit proliferation of granulation tissue and attenuate the degree of airway stenosis caused by stent implantation.


Assuntos
Traqueia , Estenose Traqueal , Animais , Cães , Constrição Patológica/complicações , Intubação Intratraqueal/efeitos adversos , Intubação Intratraqueal/métodos , Mitomicina/farmacologia , Mitomicina/uso terapêutico , Ruptura/complicações , Ruptura/tratamento farmacológico , Ruptura/patologia , Stents/efeitos adversos , Traqueia/patologia , Estenose Traqueal/etiologia , Estenose Traqueal/terapia
6.
Biotechnol Appl Biochem ; 68(2): 356-365, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32357262

RESUMO

Chronic obstructive pulmonary disease (COPD) is a chronic and devastating condition characterized by poor airflow and breath. Smoking and other environmental factors-caused inflammations triggered excessive autophagy of normal lung epithelial cells, eventually leading to impaired lung functions. Previous studies showed that ghrelin exhibited beneficial effects on patients with COPD. However, the mechanisms underlying this impact remained largely unknown. In this study, in vitro and in vivo models of COPD-associated inflammation were established, and we found that inflammation and autophagy were abonormally activated through nuclear factor kappa b (NF-κB) and activator protein-1 (AP-1) signaling pathways. Interestingly, ghrelin could inhibit the excessive inflammation pathways and autophagy induced by particle matter and/or cigarette extract in bronchial epithelial cells. Furthermore, NF-κB and AP-1 signaling were both inhibited while lung functions were significantly improved. Taken together, identification of downstream signaling of ghrelin in inflammation provided a new avenue in the treatment of COPD.


Assuntos
Autofagia/efeitos dos fármacos , Grelina/farmacologia , Doença Pulmonar Obstrutiva Crônica , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Camundongos , NF-kappa B/metabolismo , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Fator de Transcrição AP-1/metabolismo
7.
Int Immunopharmacol ; 77: 105939, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31718930

RESUMO

BACKGROUND: Ambient fine particulate matter (PM2.5) could induce lung injury. Aryl hydrocarbon receptor (AhR) is involved in the molecular mechanisms of prooxidative and pro-inflammatory effect of PM2.5. Molecular hydrogen has antioxidant properties. The protective effect and mechanism of hydrogen on PM2.5-induced lung injury remain unclear. OBJECTIVES: This study aimed to determine whether hydrogen could alleviate lung injury in a rat model of subacute exposure to concentrated ambient PM2.5, and explore the mechanism related to AhR. METHODS: Male Wastar rats were exposed to either concentrated ambient particles (CAPs) (diameter: ≤2.5 µm, average concentration: 1328 ±â€¯730 µg/m3) or filtered air (FA) by nose-only inhalation (5 h/day, 5 days/week for 4 weeks). Hydrogen-treated rats inhaled 66.7% hydrogen from water electrolysis for 2 h after each exposure to CAPs or FA. RESULTS: CAPs inhalation induced lung injury, as demonstrated by pulmonary function decrease, histopathological damage, mucus hypersecretion [Periodic acid-Schiff (PAS) staining for mucins, immunohistochemistry and quantitative real-time PCR (RT-qPCR) for mucin 5AC (MUC5AC) expression], increased pro-inflammatory cytokines (TNF-α, IL-8 and IL-1ß) and oxidative damage indexes [malondialdehyde (MDA) and 8-isoprostane F2α (8-iso-PG)]. While, hydrogen inhalation significantly alleviated the damages mentioned above. In addition, low expression of AhR in lung tissues determined by Western Blot was found after CAPs exposure, whereas hydrogen inhibited AhR decline induced by CAPs. CONCLUSIONS: High concentrations of hydrogen could ameliorate pulmonary dysfunction, airway mucus hypersecretion, oxidation damage, and inflammation response in rats exposed to concentrated ambient PM2.5. Additionally, hydrogen alleviates lung injury induced by PM2.5 possibly through AhR-dependent mechanisms.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hidrogênio/farmacologia , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/metabolismo , Material Particulado/efeitos adversos , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lesão Pulmonar/induzido quimicamente , Masculino , Tamanho da Partícula , Ratos , Ratos Wistar
8.
Artigo em Inglês | MEDLINE | ID: mdl-27199554

RESUMO

BACKGROUND: Systemic inflammation and steroid resistance are the hallmarks of COPD. We examined the impact of p38 inhibitor (SB203580) in in vitro assays of systemic inflammation using pulmonary cells and patients' sera. OBJECTIVE AND METHODS: Data from 66 COPD patients and 15 age-/sex-matched healthy controls were compared. Interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α), and CCL5 were measured in serum samples and culture media from peripheral blood mononuclear cells. The impact of sera on IL-10 and CCL5 expression in alveolar macrophage cell line (MH-S) was examined. The in vitro effects of SB203580 on lipopolysaccharide-induced inflammation were investigated. RESULTS: Peripheral blood mononuclear cells from Global initiative for chronic Obstructive Lung Disease (GOLD) D patients produced more CCL5 and TNF-α, and less IL-10 compared to GOLD A-C patients. SB203580 treatment suppressed CCL5 and TNF-α and stimulated IL-10 production; however, the effect of SB203580 on IL-10 was lower in the COPD group. Culture of MH-S cells with COPD serum showed a significant increase in CCL5 and a significant decrease in IL-10 compared to healthy serum. This effect was not suppressed with SB203580 treatment. CONCLUSION: COPD serum has a potent proinflammatory effect on pulmonary cells. Inhibition of p38 phoshorylation had a limited effect in restoring impaired lymphocyte function and suppressing inflammation induced by COPD serum, implying important p38-independent inflammatory mechanisms in COPD.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/sangue , Imidazóis/farmacologia , Mediadores da Inflamação/sangue , Macrófagos Alveolares/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Doença Pulmonar Obstrutiva Crônica/sangue , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Idoso , Estudos de Casos e Controles , Linhagem Celular , Quimiocina CCL5/sangue , Feminino , Humanos , Interleucina-10/sangue , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/imunologia , Masculino , Pessoa de Meia-Idade , Fosforilação , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/imunologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Inflammation ; 39(2): 881-90, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26899569

RESUMO

UNLABELLED: Transmembrane protein 16A (TMEM16A) regulates a wide variety of cellular activities, including epithelial fluid secretion and maintenance of ion homeostasis. Lipopolysaccharide (LPS), an outer membrane component of Gram-negative bacteria, is one of the major causes of acute lung injury (ALI). In this study, we investigated the effects of LPS on the expression of TMEM16A in LA795 cells and mouse lung tissue and the potential mechanism. RESULT: We detected the expression of TMEM16A in LA795 cells and mouse lung tissue by RT-PCR, Western blot, and RNA interference techniques. TMEM16A expression was significantly increased by LPS stimulation in LA795 cells and in mouse lung tissue. Moreover, the LPS-induced TMEM16A expression enhancement in lung tissue was much more prominent in the alveolar epithelial region than in bigger airway epithelial cells. The typical TMEM16A current was recorded, and LPS treatment significantly enhances the current amplitude in LA795 cells. TMEM16A shRNA or TMEM16A inhibitor (T16Ainh-A01) did not affect alveolar fluid clearance (AFC), while co-application of T16Ainh-A01 induced a stronger AFC inhibition than LPS alone. LPS notably and synchronously enhanced Akt phosphorylation (p-Akt) and TMEM16A expression in a time-dependent manner in LA795 cells. Taken together, our results suggest that TMEM16A maybe plays an important role in pathological conditions of LPS-induced ALI as a protective protein.


Assuntos
Lesão Pulmonar Aguda/patologia , Canais de Cloreto/metabolismo , Lipopolissacarídeos/toxicidade , Edema Pulmonar/patologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Anoctamina-1 , Líquido da Lavagem Broncoalveolar/química , Linhagem Celular Tumoral , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Modelos Animais de Doenças , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Alvéolos Pulmonares/metabolismo , Edema Pulmonar/induzido quimicamente , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
10.
Int J Mol Med ; 37(1): 182-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26531002

RESUMO

Sulforaphane (1-isothiocyanate-4-methyl sulfonyl butane) is a plant extract (obtained from cruciferous vegetables, such as broccoli and cabbage) and is known to exert anticancer, antioxidant and anti-inflammatory effects. It stimulates the generation of human or animal cells, which is beneficial to the body. The aim of the current study was to determine whether sulforaphane protects against lipopolysaccharide (LPS)­induced acute lung injury (ALI) through its anti-inflammatory effects, and to investigate the signaling pathways involved. For this purpose, male BALB/c mice were treated with sulforaphane (50 mg/kg) and 3 days later, ALI was induced by the administration of LPS (5 mg/kg) and we thus established the model of ALI. Our results revealed that sulforaphane significantly decreased lactate dehydrogenase (LDH) activity (as shown by LDH assay), the wet-to-dry ratio of the lungs and the serum levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) (measured by ELISA), as well as nuclear factor-κB protein expression in mice with LPS-induced ALI. Moreover, treatment with sulforaphane significantly inhibited prostaglandin E2 (PGE2) production, and cyclooxygenase-2 (COX-2), matrix metalloproteinase-9 (MMP-9) protein expression (as shown by western blot analysis), as well as inducible nitric oxide synthase (iNOS) activity in mice with LPS-induced ALI. Lastly, we noted that pre-treatment with sulforaphane activated the nuclear factor-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway in the mice with LPS-induced ALI. These findings demonstrate that sulforaphane exerts protective effects against LPS-induced ALI through the Nrf2/ARE pathway. Thus, sulforaphane may be a potential a candidate for use in the treatment of ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Elementos de Resposta Antioxidante/efeitos dos fármacos , Isotiocianatos/uso terapêutico , Pulmão/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/imunologia , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Anti-Inflamatórios/farmacologia , Interleucina-6/sangue , Interleucina-6/imunologia , Isotiocianatos/farmacologia , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Sulfóxidos , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia
11.
Tumour Biol ; 36(1): 259-69, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25238880

RESUMO

Thymoquinone (TQ) is the primary bioactive component of Nigella sativa Linn seed oil and used as anti-inflammatory, anti-oxidant, and anti-neoplastic agent. Previous studies have shown that TQ exhibits inhibitory effects on multiple cancers. However, the detailed antineoplastic effects and its molecular mechanisms of TQ on lung cancer are not entirely elucidated yet. In the present study, we aimed to investigate the effects of TQ on cell proliferation, migration, and invasion as well as its underlying anti-metastatic mechanisms in A549 cells. Lung cancer cell line A549 cells were treated with different concentration of TQ for different period of time, and the growth-inhibitory effects of TQ was measured by MTT and cell count assays; cell cycle was determined by flow cytometry; wound healing and transwell assays were used to assess the cell migration and invasion activities; Western blot and real-time quantitative RT-PCR were used to determine the expression of proliferation and invasion associated genes as well as MAPKs pathway molecules; gelatinase activity was estimated using gelatin zymography assay. The results show that TQ played a role in inhibiting the proliferation, migration, and invasion of A549 lung cancer cells, it also inhibited the expression level of PCNA, cyclin D1, MMP2, and MMP9 mRNA and protein in a dose- and time-dependent manner especially at 10, 20, 40 µmol/L concentrations. The cell cycle inhibitor P16 expression and the gelatinase activities of MMP2 and MMP9 were also inhibited by TQ dramatically. TQ reduced phosphorylation of ERK1/2; however, the proliferation and invasion inhibitory effects of TQ on A549 cells were neutralized by ERK1/2 inhibitor PD98059. In conclusion, our study confirmed that TQ could inhibit A549 cell proliferation, migration, and invasion through ERK1/2 pathway, as proposed the therapeutic potential of TQ as an anti-metastatic agent in human lung cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Benzoquinonas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica
12.
Oncol Lett ; 8(2): 948-952, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25013520

RESUMO

The present study sought to characterize the role of macrophage inflammatory protein-3α (MIP-3α) in non-small cell lung cancer (NSCLC) patients with early recurrence or metastasis after primary pulmonary resection. Follow-up examinations were conducted for 203 NSCLC patients with primary pulmonary resection for two years post-operatively, and data was also collected for 20 healthy subjects. Serum MIP-3α levels were determined prior to surgery and at post-operative days (PODs) 30, 90 and 180, and the relevant clinical and operative variables were collected. Serum MIP-3α was measured using a commercially available enzyme-linked immunosorbent assay. There were no significant differences in age, gender and histological type among all groups (P>0.05). Serum MIP-3α levels on POD 180 were significantly higher in the recurrence group than in the non-recurrence group and healthy subjects (P=0.001). There was no significant difference in the serum MIP-3α level at PODs 90 and 180 in the patients with or without adjuvant chemotherapy (P>0.05). The recurrence rate in the high serum MIP-3α level group was 41.67%, much higher than the 23.53% observed in the low level group (P=0.006). The patients with high serum levels of MIP-3α had a significantly shorter overall recurrence-free time compared with those with low levels (P=0.004). Multivariate Cox's regression analyses showed that only serum MIP-3α level was significant, with a hazard ratio of 1.061, a 95% confidence interval of 1.044-1.078 and a P-value of 0.001. The serum MIP-3α level in the patients with liver and bone metastases were remarkably higher than those with recurrence at other sites. The high post-operative serum MIP-3α levels were associated with an increased risk of post-operative early recurrence or metastasis in the lung cancer patients, specifically in those with bone or liver metastases.

13.
Exp Lung Res ; 40(5): 237-50, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24784799

RESUMO

OBJECTIVE: To observe the expression of endogenous TMEM16A in rat alveolar type II epithelial cells (AT-II) and A549, and study the effect of TMEM16A on lipopolysaccharide (LPS)-induced proinflammatory cytokine secretion. METHODS: Rat AT-II cells were isolated and TMEM16A protein expression in rat AT-II cells was measured by Western blot. TMEM16A mRNA and protein expressions in A549 were measured by real-time quantitative polymerase chain reaction (PCR) and Western blot, respectively. TMEM16A gene was transfected into A549 using Lipofectamine 2000. Transfected cells were selected in the presence of G418 to create a stable TMEM16A overexpression A549 cell line. The expression of TMEM16A in A549 was knocked down by lentiviral vector-mediated RNA interference. TNF-α and IL-8 levels were determined by enzyme-linked immunosorbent assay (ELISA). A dual-luciferase reporter assay system was used to measure the transcriptional activity of NF-κB. RESULTS: (1) Endogenous TMEM16A was expressed in rat AT-II and A549. (2) TMEM16A expression in A549 significantly increased at 24 hours and 36 hours, and then decreased at 48 hours after LPS treatment. (3) TMEM16A mRNA and protein expressions were increased in the stable TMEM16A overexpression A549 cell line. (4) TMEM16A overexpression decreased the LPS-induced TNF-α and IL-8 secretions. (5) TMEM16A mRNA and protein expressions were knocked down in TMEM16A-siRNA lentivirus transfected A549. (6) TMEM16A knockdown increased the LPS-induced TNF-α and IL-8 secretions. (7) TMEM16A overexpression inhibited LPS-induced NF-κB activation. CONCLUSIONS: TMEM16A is expressed in AT-II. TMEM16A in A549 inhibits LPS-induced NF-κB activation and decreases proinflammatory cytokines release, protecting A549 from acute LPS-mediated damage.


Assuntos
Canais de Cloreto/fisiologia , Inflamação/prevenção & controle , Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Animais , Anoctamina-1 , Células Cultivadas , Canais de Cloreto/análise , Canais de Cloreto/genética , Citocinas/biossíntese , Células Epiteliais/imunologia , Humanos , Inflamação/imunologia , Pulmão/citologia , Masculino , NF-kappa B/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley
14.
J Asthma ; 51(3): 232-42, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24303994

RESUMO

BACKGROUND: There is a lack of information on the prevalence, severity and risk factors of asthma, rhinitis and eczema in Chinese children. OBJECTIVE: To establish baseline data for a major longitudinal study of factors affecting asthma, rhinitis and eczema in a large group of children from the industrialized city of Shijiazhuang, China. METHODS: We used the International Study of Asthma and Allergies in Childhood questionnaire and studied 10 338 children, ages 6-18, from Shijiazhuang. RESULTS: The prevalence of childhood asthma, rhinitis and eczema is 1.2%, 13.5% and 11.8%, respectively. Boys had higher prevalence of these conditions than girls and younger children had higher prevalence of asthma and eczema but lower prevalence of rhinitis than older children. Breastfed children had lower prevalence of asthma and rhinitis, but not eczema, than non-breastfed children. Overweight children had higher prevalence of asthma and rhinitis than those who were not overweight. Children exposed to paternal smoking had higher prevalence of rhinitis and eczema than those not exposed; children exposed to pets had higher prevalence of asthma and rhinitis than those not exposed. CONCLUSIONS: The prevalence of asthma in this study group is low, but the prevalence of rhinitis is high, and could be considered a major public health problem. The prevalence of asthma, rhinitis and eczema is generally higher in boys and younger children generally have higher prevalence of asthma and eczema but lower prevalence of rhinitis. Exposure to pets is a risk factor for rhinitis, being overweight is a risk factor for asthma and rhinitis, and exposure to parental smoking is a risk factor for asthma, rhinitis and eczema in these children.


Assuntos
Asma/epidemiologia , Eczema/epidemiologia , Rinite/epidemiologia , Adolescente , Distribuição por Idade , Animais , Pesos e Medidas Corporais , Aleitamento Materno/estatística & dados numéricos , Criança , China/epidemiologia , Feminino , Humanos , Masculino , Animais de Estimação , Prevalência , Características de Residência , Fatores de Risco , Índice de Gravidade de Doença , Distribuição por Sexo , Fatores Socioeconômicos , Poluição por Fumaça de Tabaco/estatística & dados numéricos
15.
Mol Med Rep ; 8(3): 903-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23900639

RESUMO

Hypoxia is an important mechanism involved in dermal scar formation. Post-intubation tracheal stenosis (PTS) has similar pathological characteristics and formation mechanisms to skin hypertrophic scars. Hypoxia-inducible factor­1α (HIF­1α) is a nuclear transcription factor that facilitates the adaptation of human cells to compromised oxygen tension under hypoxic conditions. The aim of this study was to investigate whether hypoxia and HIF-1α were involved in PTS. In the first part of our study, we observed the effects of tracheal mucosal pressure exerted by the endotracheal tube cuff and duration of intubation on tracheal stenosis using prospective methods. In the second part of our study, 24 patients were divided into three groups, according to the characteristics observed using bronchoscopy: granulation phase group, proliferative phase group and mature phase group. Tissues showing dysplasia were obtained using bronchoscopy forceps, and western blotting was performed to detect the protein expression of HIF-1α and its target genes, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF) and transforming growth factor (TGF)-ß. In experiment one, we observed that the tracheal mucosal pressure exerted by the endotracheal tube cuff in the PTS group was significantly higher compared with that in the group without PTS, and the duration of intubation was longer compared with that in the group without PTS (P<0.05). In experiment two, we observed that the expression of HIF-1α was highest in the granulation phase, showed a decrease in the proliferative phase and in the mature phase was significantly reduced compared with the other two phases. The results were similar for VEGF and bFGF. TGF­ß protein expression was highest in the proliferative phase, significantly reduced in the mature phase and the same trends were observed for collagen type Ⅰ and III, and α-smooth muscle actin. HIF-1α is involved in the pathogenesis of PTS and may be a potential key regulator in the initiation and facilitation of this process.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Estenose Traqueal/metabolismo , Actinas/metabolismo , Adolescente , Adulto , Idoso , Broncoscopia , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Matriz Extracelular/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Hipóxia , Masculino , Pessoa de Meia-Idade , Estenose Traqueal/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA