Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Filtros aplicados
Base de dados
Intervalo de ano de publicação
1.
Allergol. immunopatol ; 52(2): 16-22, mar. 2024. graf, ilus
Artigo em Inglês | IBECS | ID: ibc-231084

RESUMO

Background: Sepsis is a life-threatening condition characterized by acute organ dysfunction, which frequently leads to acute lung injury (ALI) in approximately 40% of cases. Isoegomaketone (IK) is a constituent of essential oil found in P. frutescens, known for its diverse biological properties, including anti-inflammatory and antitumor effects. However, the regulatory impact of IK on ALI in the context of sepsis remains poorly understood. Methods: Pathological alterations in lung tissues were assessed using hematoxylin and eosin staining. Enumeration of total leukocytes and neutrophils in bronchoalveolar lavage fluid (BALF) was performed using a hematocytometer, while the levels of interleukin (IL)-6, IL-1β, IL-10, and IL-17 in BALF were quantified using enzyme-linked immunosorbent serological assay. In addition, the levels of malondialdehyde (MDA), myeloperoxidase (MPO), superoxide dismutase (SOD), and glutathione (GSH) in lung tissues were assessed using respective commercial kits; cell apoptosis was evaluated using the terminal deoxynucleotide transferase--mediated dUTP nick end-labeling assay, and protein expressions were determined through Western blot analysis. Results: Our findings revealed that cecal ligation and puncture (CLP) treatment in mice induced severe lung injury, characterized by increased lung injury scores, significant bleeding, neutrophil infiltration, and alveolar edema. However, treatment with IK at a dose of 10 mg/kg ameliorated CLP-induced lung injury, while IK dose of 5 mg/kg showed no significant effect. Additionally, IK treatment at 10 mg/kg reduced CLP-induced inflammation by decreasing levels of IL-6, IL-1β, IL-10, and IL-17. Furthermore, IK at 10 mg/kg attenuated CLP-induced oxidative stress by modulating levels of MDA, MPO, SOD, and GSH... (AU)


Assuntos
Ratos , Sepse , Lesão Pulmonar Aguda , Óleos Voláteis , Perilla frutescens , Anti-Inflamatórios , Ensaios de Seleção de Medicamentos Antitumorais , Coloração e Rotulagem , Hematoxilina
2.
Allergol. immunopatol ; 52(2): 60-67, mar. 2024. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-231086

RESUMO

Objective: To explore the role of Y-box binding protein 1 (YBX-1) in the lipopolysaccharide (LPS)-stimulated inflammation and oxidative stress of BEAS-2B cell line and clarify the underlying mechanism. Methods: LPS-stimulated BEAS-2B cells were used as a cell model of sepsis-stimulated acute lung injury (ALI). Immunoblot and quantitative polymerase chain reaction assays were used to detect the expression of YBX-1 in LPS-stimulated BEAS-2B cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide, TdT-mediated dUTP nick end labeling, and immunoblot assays were conducted to determine the effects of YBX-1 on cell survival. JC-1 staining and adenosine triphosphate production were used to detect the effects of YBX-1 on mitochondrial function. Immunostaining and enzyme-linked immunosorbent serologic assay were performed to examine the effects of YBX-1 on the inflammation and oxidative stress of cells. Immunoblot assay was conducted to confirm the mechanism. Results: YBX-1 was lowly expressed in LPS-stimulated BEAS-2B cells and enhanced the survival of LPS-stimulated lung epithelial cells. In addition, YBX-1 improved mitochondrial function of LPS-stimulated BEAS-2B cells. YBX-1 inhibited the inflammation and oxidative stress of LPS-stimulated BEAS-2B cells. Mechanically, YBX-1 inhibited mitogen-activated protein kinase (MAPK) axis, thereby alleviating sepsis-stimulated ALI. Conclusion: YBX-1 alleviated inflammation and oxidative stress of LPS-stimulated BEAS-2B cells via MAPK axis. (AU)


Assuntos
Humanos , Proteína 1 de Ligação a Y-Box , Inflamação , Lipopolissacarídeos , Lesão Pulmonar Aguda , Sepse , Sobrevivência Celular , Células Epiteliais Alveolares
3.
Allergol. immunopatol ; 51(5)01 sept. 2023. tab, graf
Artigo em Inglês | IBECS | ID: ibc-225047

RESUMO

Objective: To investigate the regulatory mechanism of pleckstrin homology-like domain, family A, member 1 (PHLDA1) in sepsis-induced acute lung injury (ALI). Method: Mice model of sepsis were established by cecal ligation and puncture (CLP). The expression of PHLDA1 was reduced by injecting short hairpin RNA (shRNA)–PHLDA1 into the tail vein. The levels of PHLDA1, pro-inflammatory cytokines, such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), IL-1β, IL-18, super-oxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH), molecular mechanism related to pyroptosis, such as caspase 1, adaptor apoptosis-associated speck-like protein containing a CARD (ASC), and gasdermin D (GSDMD)-N, and nucleotide oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) were tested by Western blot analysis, quantitative real-time polymerase chain reaction, and enzyme-linked-immunosorbent serologic assay. Pathological changes in lung tissues were examined by hematoxylin and eosin staining. Wet–dry weight ratio of lung tissues was observed. Results: The expression of PHLDA1 was up-regulated in lung tissues from CLP-induced septic mice. Knockdown of PHLDA1 could reduce lung injury and wet–dry weight ratio in mice with sepsis-induced ALI. Moreover, silencing of PHLDA1 decreased the expressions of IL-1β, TNF-α, IL-18, IL-6, and MDA but increased SOD and GSH expressions in CLP-induced septic mice. The expressions of NLRP3, GSDMD-N, ASC, and caspase 1 were decreased by PHLDA1 silencing. Conclusion: Knockdown of PHLDA1 inhibited lung inflammation and pyroptosis in mice with sepsis-induced ALI by down-regulating NLRP3 (AU)


Assuntos
Animais , Masculino , Camundongos , Sepse/complicações , Sepse/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Regulação para Baixo
4.
Allergol. immunopatol ; 51(3): 56-67, 01 mayo 2023. tab
Artigo em Inglês | IBECS | ID: ibc-219814

RESUMO

Objective: Acute lung injury (ALI) is a common complication of critical diseases with high morbidity and mortality. This study explored the regulatory role and mechanism of high mobility histone box 1 protein (HMGB1) on pulmonary fibrosis (PF) after ALI in rats through nucleotide oligomerization domain-like receptor protein-3 (NLRP3) inflammasome. Methods: PF rat models after ALI were established by induction of bleomycin. Degree of fibrosis was assessed by Masson staining and Ashcroft scoring. Hydroxyproline (Hyp) contents in lung tissues and rat lung tissue morphology were detected by enzyme-linked-immunosorbent serologic assay (ELISA) and hematoxylin and eosin staining. The levels of NLRP3, major proteins of NLRP3 inflammasome (NLRP3/ASC/caspase-1), and downstream inflammatory cytokines interleukin (IL)-1 and IL-18 were determined using immunohistochemistry, Western blotting analysis, and ELISA. The nuclear/cytoplasmic nuclear factor erythroid 2-related factor 2 (Nrf2) levels and HO-1 levels were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis. Rats was injected with lentivirus carrying short hairpin (sh)-HMGB1 and zinc protoporphyria (ZNPP) (HO-1 inhibitor) to assess the effects of HMGB1 and HO-1 on PF and NLRP3 inflammasome activation. Results: Bleomycin induced PF after ALI in rats, manifested as patchy fibrosis, atelectasis, and excessive expansion, and increased Aschcroft score and Hyp content. Bleomycin treatment enhanced levels of NLRP3, ASC, caspase-1, IL-1, and IL-18 in rat lung tissues, which promoted activation of NLRP3 inflammasome. HMGB1 was up-regulated in bleomycin-induced rats. HMGB1 knockdown partially reversed NLRP3 inflammasome activation and PF progression (AU)


Assuntos
Humanos , Masculino , Ratos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Proteína HMGB1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Lesão Pulmonar Aguda , Modelos Animais de Doenças , Ratos Wistar , Bleomicina
5.
Allergol. immunopatol ; 51(3): 143-152, 01 mayo 2023. ilus
Artigo em Inglês | IBECS | ID: ibc-219822

RESUMO

Background: Acute lung injury (ALI) is a complex disease with a high mortality. Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) is a protein tyrosine phosphatase that participates in pathogenesis of multiple diseases. Nevertheless, the role of SHP2 in ALI remains unknown. Methods: The in vivo and in vitro lipopolysaccharide (LPS)-induced ALI models were successfully established. The histopathological changes were evaluated by hematoxylin and eosin staining. The vascular permeability of lungs was assessed by Evans blue assay. The expression of ACSL4 and SHP2 was detected by western blot and qRT-PCR assay. The lactate dehydrogenase (LDH) activity, malondialdehyde (MDA), iron, and glutathione (GSH) levels were measured by commercial kits. Results: The SHP2 was upregulated in LPS-induced ALI mice and LPS-stimulated MLE-12 cells. In loss-of function experiment, the knockdown of SHP2 attenuated LPS-induced lung injury, microvessels damage, pulmonary edema, and increase of lung vascular permeability in vivo. Mechanically, shSHP2-rescued LPS induced increase in LDH activity, MDA, and iron levels, and decrease in GSH levels, as well as the accumulation of reactive oxygen species in vivo and in vitro, leading to an alleviation of LPS-induced ferroptosis. Notably, shSHP2 reduced the expression of Acyl-CoA synthetase long-chain 4 (ACSL4). In the rescued experiments, overexpression of ACSL4 abolished the shSHP2-induced reduction of LDH activity, MDA, and iron levels, and increase in GSH levels, thereby aggravating the LPS-induced ferroptosis. Conclusion: These findings concluded that the knockdown of SHP2 attenuated LPS-induced ferroptosis via downregulation of ACSL4 expression in ALI, providing a novel sight for ALI treatment (AU)


Assuntos
Animais , Masculino , Camundongos , Lesão Pulmonar Aguda/metabolismo , Regulação para Baixo , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos BALB C
6.
Allergol. immunopatol ; 51(1): 37-43, ene. 2023. graf
Artigo em Inglês | IBECS | ID: ibc-214020

RESUMO

Objective: To reveal the possible effects of decursin on viability, oxidative stress, and inflammatory response in lipopolysaccharide (LPS)-treated human bronchial epithelial cells-2B (BEAS-2B) and human pulmonary artery endothelial cells (HPAEC) cells, and revealed the potential mechanisms. Methods: LPS was used to induce acute lung injury (ALI) in normal human lung epithelial cells, including BEAS-2B and HPAEC cells. Cell viability and apoptosis in response to LPS and decursin in BEAS-2B and HPAEC cells were, respectively, evaluated by MTT colorimetric and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. The oxidative stress and inflammatory response in LPS-treated BEAS-2B and HPAEC cells were detected by enzyme-linked-immunosorbent serologic assay. In addition, the role of decursin in nuclear -factor-kappa B (NF-κB) activation was analyzed by immunoblot and immunofluorescence assays. Results: Our data revealed that decursin could alleviate the viability of LPS-induced BEAS-2B and HPAEC cells. Decursin could also reduce LPS-induced oxidative stress in BEAS-2B and HPAEC cells. In addition, it could reduce LPS-induced inflammation in BEAS-2B and HPAEC cells. Mechanically, decursin suppressed the activation of NF-κB pathway. Conclusion: Decursin suppressed NF-κB pathway, and therefore alleviated ALI (AU)


Assuntos
Humanos , Lesão Pulmonar Aguda/metabolismo , Lipopolissacarídeos , NF-kappa B/metabolismo , Progressão da Doença , Transdução de Sinais
7.
Allergol. immunopatol ; 51(1): 44-53, ene. 2023. tab, graf
Artigo em Inglês | IBECS | ID: ibc-214021

RESUMO

Background/objective: Acute lung injury (ALI) is a critical clinical syndrome with high rates of incidence and mortality. However, its molecular mechanism remains unclear. The current work aimed to explore the molecular mechanisms of ALI by identifying different expression genes (DEGs) and candidate drugs using a combination of chip analysis and experimental validation. Methods: Three microarray datasets were downloaded from Gene Expression Omnibus (GEO) database to obtain DEGs. We conducted a Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway-enrichment analyses of overlapping DEGs among three databases. The expression level of key gene was verified by Western blotting analysis in LPS-treated ALI cell models. Finally, we predicted the candidate drugs targeting the key gene that might be effective for ALI treatment, and the role of candidate drug in treating ALI was verified by investigation. Results: A total 29 overlapping DEGs were up-regulated in LPS-induced ALI groups. They were enriched in inflammation and inflammation-related pathways. Serpin family A member 3 (SERPINA3) was defined as a key gene because it was associated with inflammation pathway and up-regulated in microarray datasets in LPS-induced ALI. In LPS-induced human bronchial epithelial cells transformed with Ad12-SV40-2B (BEAS-2B) cells, SERPINA3 was enhanced. Pyridoxal phosphate as an upstream drug of SERPINA3 could improve cell viability and reduce expression inflammatory factors in LPS-treated BEAS-2B cells. Conclusion: Our study suggested that pyridoxal phosphate could be a candidate drug targeting SERPINA3 gene in LPS-induced ALI. It has protective and anti-inflammatory effects in BEAS-2B cells, and may become a potential novel treatment for ALI (AU)


Assuntos
Humanos , Biologia Computacional/métodos , Lesão Pulmonar Aguda/diagnóstico , Lipopolissacarídeos , Biomarcadores , Células Cultivadas , Expressão Gênica , Serpinas
8.
Allergol. immunopatol ; 51(1): 54-62, ene. 2023. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-214022

RESUMO

Acute lung injury causes severe inflammation and oxidative stress in lung tissues. In this study, we analyzed the potential regulatory role of nuclear factor erythroid-2-related factor 2 (Nrf2) on NADPH oxidase 1 (NOX1) in tumor necrosis factor-α (TNF-α)-induced inflammation and oxidative stress in human type II alveolar epithelial cells. In this study, A549 cells were transfected with Nrf2 siRNA and overexpression vectors for 6 h before being induced by TNF-α for 24 h. TNF-α upregulated the expression of NOX1 and Nrf2 in A549 cells. Furthermore, overexpression of Nrf2 could reduce TNF-α-induced NF-κB mRNA and protein expression after transfection with the Nrf2 siRNA vector, and the levels of IL-6, IL-8, ROS, and malondialdehyde (MDA) in TNF-α-induced A549 cells increased, while the level of total antioxidation capability (T-AOC) decreased. On the other hand, the overexpression of Nrf2 decreased the levels of IL-6, IL-8, ROS, and MDA, while increasing T-AOC. The mRNA and protein levels of NOX1 were dramatically increased by TNF-α, while those changes were notably suppressed by Nrf2 overexpression. Further studies demonstrated that Nrf2 suppressed NOX1 transcription by binding to the -1199 to -1189 bp (ATTACACAGCA) region of the NOX1 promoter in TNF-α-stimulated A549 cells. Our study suggests that Nrf2 may bind to and regulate NOX1 expression to antagonize TNF-α-induced inflammatory reaction and oxidative stress in A549 cells (AU)


Assuntos
Humanos , Lesão Pulmonar Aguda/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , NADPH Oxidase 1/metabolismo , Estresse Oxidativo , Células Cultivadas
9.
Allergol. immunopatol ; 51(6): 1-7, 2023. graf
Artigo em Inglês | IBECS | ID: ibc-227302

RESUMO

Background: Acute lung injury (ALI) causes severe and uncontrolled pulmonary inflammation and has high morbidity in dying patients. Objective: This study aimed to evaluate the potential function of Kaempferitrin (Kae) and uncover its mechanisms in ALI. Material and Methods: We evaluated the role of Kae in ALI through the lipopolysaccharide (LPS)-induced histopathological changes, lung wet/dry (W/D) ratio, total bronchoalveolar lavage fluid (BALF) cells count, pulmonary inflammation, and the levels of interleukin (IL)-6, tumor necrosis factor-α (TNF-α), and IL-1β. The effect of Kae on NF-κB signaling pathway was discovered through the protein expression levels of transcription factors p65, p-p65, IκBα, and p-IκBα by Western blot analysis. Results: The results showed that Kae could improve lung injury by reducing apoptosis, histopathological changes, and lung W/D ratio; more importantly, Kae enhanced the survival of ALI mice. Moreover, Kae relieved inflammation, as it reduced total BALF cells count, and deceased the levels of TNF-α, IL-6, and IL-1β in serum. In addition, Western blot analysis data suggested that Kae could decrease the protein expression levels of transcription factors p65, p-p65, IκB-α, and p-IκB-α, which were promoted by LPS. Conclusion: The results of this study suggested that Kae could relieve LPS-induced ALI in mice and reduce inflammation and apoptosis through NF-κB pathway (AU)


Assuntos
Animais , Masculino , Camundongos , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , NF-kappa B/metabolismo , Pneumonia/patologia , Sepse , Inflamação/patologia , Lipopolissacarídeos/efeitos adversos , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Endogâmicos C57BL
10.
Allergol. immunopatol ; 51(6): 30-36, 2023. graf
Artigo em Inglês | IBECS | ID: ibc-227306

RESUMO

Objective: This study aimed to investigate the functioning and mechanism of coptisine in acute lung injury (ALI). Methods: Murine Lung Epithelial 12 (MLE-12) cells were stimulated with lipopolysaccharide (LPS) to construct an in vitro pulmonary injury model to study the functioning of coptisine in sepsis-induced ALI. The viability of MLE-12 cells was assessed by the cell counting kit-8 assay. The cytokine release of tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), and IL-1β was measured by enzyme-linked-immunosorbent serologic assay. The relative expression levels of TNF-α, IL-6, and IL-1β mRNA were examined by reverse transcription-quantitative polymerase chain reaction. The cell apoptosis of MLE-12 cells was determined by Annexin V/propidium iodide staining and analyzed by flow cytometry. The expressions of apoptosis-related proteins Bax and cleaved Caspase-3 were observed by Western blot analysis. The activation of nuclear factor kappa B (NF-κB) signaling pathway was discovered by the determination of phospho-p65, p65, phospho-nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα), and IκBα through Western blot analysis. Results: Coptisine treatment could significantly restore decrease in MLE-12 cell viability caused by LPS stimulation. The release of TNF-α, IL-6, and IL-1β was significantly inhibited by coptisine treatment. Coptisine treatment inhibited MLE-12 cell apoptosis induced by LPS, and also inhibited the expression levels of Bax and cleaved Caspase-3. Coptisine treatment along with LPS stimulation, significantly reduced the protein level of phospho-IκBα, increased the level of IκBα, and reduced phospho-p65–p65 ratio. Conclusion: These results indicated that coptisine attenuated sepsis lung injury by suppressing lung epithelial cell inflammation and apoptosis through NF-κB pathway. Therefore, coptisine may have potential to treat sepsis-induced ALI (AU)


Assuntos
Humanos , Animais , Lesão Pulmonar Aguda/tratamento farmacológico , Pneumonia/tratamento farmacológico , Apoptose , Caspase 3 , Células Epiteliais/metabolismo , /metabolismo , Lipopolissacarídeos/efeitos adversos , NF-kappa B/metabolismo , Sepse/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo , Proteína X Associada a bcl-2
11.
Allergol. immunopatol ; 51(6): 60-66, 2023. graf
Artigo em Inglês | IBECS | ID: ibc-227310

RESUMO

Background: Severe pneumonia is a kind of disease that develops from lung inflammation, and new drugs are still required to treat the same. Erythropoietin (EPO) is widely used to treat anemia in patients. However, there are fewer studies on the role of EPO in neutrophil extracellular trappings (NETs) and pneumonia, and the mechanism is unclear. Objective: To investigate the possible effects of EPO on the formation of NETs and progression of pneumonia. Methods: Mice pneumonia model was induced by tracheal lipopolysaccharide (LPS) administration. Hematoxylin and eosin (H&E) staining and automatic blood cell analysis were performed in this model to confirm the effects of EPO on lung injury. Flow cytometry, enzyme-linked immunosorbent serological assay, and immunostaining assay were conducted to detect the effects of EPO on the inflammation as well as formation of NETs in mice. Immunoblot was further conducted to confirm the mechanism. Results: EPO alleviated LPS-induced lung injury. EPO reduced the release of inflammatory factors induced by LPS. In addition, EPO inhibited the formation of NETs. Mechanically, EPO inhibited tumor necrosis factor (TNF) receptor associated factor 2 (TRAF2)/nuclear factor kappa-B (NF-κB) activity in mouse models, and therefore suppressed the progression of pneumonia. Conclusion: EPO inhibited formation of NETs to ameliorate lung injury in a pneumonia model, and could serve as a drug of pneumonia (AU)


Assuntos
Animais , Camundongos , Lesão Pulmonar Aguda/tratamento farmacológico , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Armadilhas Extracelulares , Pneumonia/induzido quimicamente , Modelos Animais de Doenças , Lipopolissacarídeos/efeitos adversos
12.
Allergol. immunopatol ; 50(6): 53-59, 01 nov. 2022. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-211506

RESUMO

extracellular traps formation contributes to inflammatory lung injury in sepsis. C1q/tumor necrosis factor–related protein-6 (CTRP6) is a paralog of adiponectin and exerts anti- inflammatory and antioxidant properties. The role of CTRP6 in sepsis-associated inflammatory lung injury was investigated in this study. Methods: Mice were injected with lipopolysaccharides (LPS) intraperitoneally to establish the mouse sepsis model. They were first tail-vein injected with adenovirus-mediated overexpression CTRP6 (Ad-CTRP6) and then subjected to the LPS injection. Pathological changes in lungs were detected by hematoxylin and eosin staining. Inflammation cytokine levels in bronchoalveolar lavage fluid were assessed by qRT-PCR and ELISA. Flow cytometry was used to detect the number of neutrophils in bronchoalveolar lavage fluid, and immunofluorescence was performed to assess neutrophil extracellular traps. Results: Lipopolysaccharides induced pulmonary congestion, interstitial edema, and alveolar wall thickening in the lungs, as well as upregulated lung histology score and wet/dry weight ratio. CTRP6 was reduced in lung tissues of septic mice. Injection with Ad-CTRP6 ameliorated extensive histopathological changes in LPS-induced mice and decreased lung histology score and wet/dry weight ratio. Overexpression of CTRP6 reduced the levels of TNF-α, IL-6, and IL-1β in septic mice. Injection with Ad-CTRP6 also decreased the number of neutrophils and downregulated Cit-H3 and myeloperoxidase polymers in septic mice. Protein expression of p-ERK in septic mice was reduced by overexpression of CTRP6. Conclusion: CTRP6 attenuated septic lung injury, exerted anti-inflammatory effect, and suppressed neutrophil extracellular traps formation against sepsis through inactivation of extracellular signal-regulated kinase signaling (AU)


Assuntos
Animais , Masculino , Camundongos , Lesão Pulmonar Aguda , Armadilhas Extracelulares/metabolismo , Sepse , Camundongos Endogâmicos C57BL , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Adipocinas/metabolismo , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/uso terapêutico , Sistema de Sinalização das MAP Quinases , Sepse/complicações , Sepse/metabolismo , Sepse/patologia
13.
J. physiol. biochem ; 78(4): 721–737, nov. 2022. ilus
Artigo em Inglês | IBECS | ID: ibc-216167

RESUMO

Pyroptosis is commonly induced by the gasdermin (GSDM) family and is accompanied by the release of inflammatory cytokines such as IL-1β and IL-18. Recently, increasing evidence suggests that pyroptosis plays a role in respiratory diseases. This review aimed to summarize the roles and mechanisms of pyroptosis in inflammation-related respiratory diseases. There are several pathways involved in pyroptosis, such as the canonical inflammasome-induced pathway, non-canonical inflammasome-induced pathway, caspase-1/3/6/7/GSDMB pathway, caspase-8/GSDMC pathway, caspase-8/GSDMD pathway, and caspase-3/GSEME pathway. Pyroptosis may be involved in asthma, chronic obstructive pulmonary disease (COPD), lung cancer, acute lung injury (ALI), silicosis, pulmonary hypertension (PH), and tuberculosis (TB), in which the NLRP3 inflammasome-induced pathway is mostly highlighted. Pyroptosis contributes to the deterioration of asthma, COPD, ALI, silicosis, and PH. In addition, pyroptosis has dual effects on lung cancer and TB. Additionally, whether pyroptosis participates in cystic fibrosis (CF) and sarcoidosis or not is largely unknown, though the activation of NLRP3 inflammasome is found in CF and sarcoidosis. In conclusion, pyroptosis may play a role in inflammation-related respiratory diseases, providing new therapeutic targets. (AU)


Assuntos
Humanos , Lesão Pulmonar Aguda , Doença Pulmonar Obstrutiva Crônica , Neoplasias Pulmonares , Piroptose , Proteínas de Ligação a DNA , Inflamassomos/metabolismo , Caspases
14.
Allergol. immunopatol ; 50(5): 121-128, sept. 2022. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-208632

RESUMO

Background: Sepsis-induced acute lung injury (ALI) is a syndrome associated with inflamma-tion. Cornus iridoid glycoside (CIG), a bioactive component isolated from Corni Fructus, exhibits anti-inflammatory activities. However, the function and underlying mechanisms of CIG in mice with sepsis-induced ALI remain elusive.Methods: The sepsis-elicited ALI model of mice was established by the induction of cecal ligation and puncture (CLP). The wet/dry (W/D) ratio of lung tissues was examined, and the pathological alterations were determined by hematoxylin and eosin staining. The messenger RNA (mRNA) expressions and serum levels of Interleukin (IL)-1Beta IL-6, and tumor necrosis factor-alfa(TNF-alpha) were measured by reverse transcription-quantitative polymerase chain reaction and enzyme-linked immunosorbent serologic assay, respectively. The concentrations of malondial-dehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were assessed by biochemical kits. In addition, the relative protein levels of p-p65, p65, phosphorylated-nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (p-IκBalpha), IκBalpha, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) gene were analyzed by Western blotting analysis.Results: CLP enhanced W/D ratio and aggravated pathological changes and scores in mice, which were obviously alleviated by the two concentrations of CIG treatment. CIG treatment notably decreased the CLP-induced mRNA expressions and serum levels of IL-1Beta, IL-6, TNF-alpha, and MDA, but enhanced the decreased concentrations (caused by CLP) of SOD and GSH-Px. Moreover, CIG treatment significantly decreased the ratios of p65/p-p65 and IκBα/p-IκBalpha caused by CLP, but...(AU)


Assuntos
Animais , Masculino , Camundongos , Lesão Pulmonar Aguda , Cornus , Sepse , Modelos Animais , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/etiologia , Cornus/genética , Cornus/metabolismo , Inflamação/complicações , Interleucina-6 , Glucosídeos Iridoides/efeitos adversos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , RNA Mensageiro , Sepse/complicações , Sepse/tratamento farmacológico , Sepse/patologia , Superóxido Dismutase/efeitos adversos
15.
Allergol. immunopatol ; 50(5): 138-147, sept. 2022. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-208633

RESUMO

Background: The development of acute lung injury (ALI) into a severe stage leads to acute respi-ratory distress syndrome (ARDS). The morbidity and mortality of ALI and ARDS are very high.Objective: This study is aimed to explore the effect of Krüppel-like factor 6 (KLF6) on lipopoly-saccharide (LPS)-induced type II alveolar epithelial cells in ALI by interacting with cysteine-rich angiogenic inducer 61 (CYR61).Material and Methods: ALI mice model and LPS-induced type II alveolar epithelial cells were conducted to simulate ALI in vivo and in vitro. The messenger RNA (mRNA) and protein expres-sion of KLF6 in lung tissues were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Pathological changes in lung tissues were observed by hematoxylin and eosin (H&E) staining. The viability and KLF6 expression of A549 cells treated with different concentrations of LPS were detected by cell counting kit-8 (CCK-8) assay, RT-qPCR, and Western blot analysis. After indicated treatment, the viability and apoptosis of A549 cells were analyzed by CCK-8 and TUNEL assays, and the inflammation fac-tors of A549 cells were detected by Enzyme-linked-immunosorbent serologic assay, RT-qPCR, and Western blot analysis. The combination of KLF6 and CYR61 was determined by chromatin immunoprecipitation (ChIP)-PCR and dual-luciferase reporter assay.Results: KLF6 expression was increased in lung tissues of ALI mice and LPS-induced A549 cells. Interference with KLF6 improved the viability, reduced the inflammatory damage, and promoted the apoptosis of LPS-induced A549 cells. In addition, KLF6 could bind to CYR61. Interference with KLF6 could decrease CYR61 expression in LPS-induced A549 cells. LPS also enhanced the TLR4/MYD88 signaling pathway, which was reversed by KLF6 interference. The above phenomena in LPS-induced A549 cells transfected with Si-KLF6 could be (AU)


Assuntos
Animais , Masculino , Camundongos , Lesão Pulmonar Aguda , Fator 6 Semelhante a Kruppel , Síndrome do Desconforto Respiratório , Modelos Animais de Doenças , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Células Epiteliais Alveolares/metabolismo , Apoptose , Inflamação/metabolismo , Fator 6 Semelhante a Kruppel/genética , Fator 6 Semelhante a Kruppel/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Sincalida/efeitos adversos , Sincalida/metabolismo , Camundongos Endogâmicos C57BL
16.
Allergol. immunopatol ; 50(4): 10-16, jul. 2022. graf
Artigo em Inglês | IBECS | ID: ibc-208889

RESUMO

Background: CXCL3 (C-X-C motif chemokine ligand 3) is a member of chemokines family, which binds to the receptor to recruit neutrophils to lungs, thus participating in the pathogenesis of asthmatic lung. The role of CXCL3 in sepsis-induced acute lung injury is investigated here.Methods: Human lung epithelial cell line (BEAS-2B) and human pulmonary artery endothelial cell line (HPAEC) were treated with lipopolysaccharides (LPS). MTT and flow cytometry were performed to detect cell viability and apoptosis, respectively. Enzyme-linked immunosorbent assay (ELISA) and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to assess the levels of inflammatory factors.Results: Treatment with LPS resulted in the decrease of cell viability in BEAS-2B and HPAEC. CXCL3 was particularly upregulated in LPS-treated BEAS-2B and HPAE cells. Knockdown of CXCL3 enhanced viability and suppressed apoptosis i006E LPS-treated BEAS-2B and HPAE cells. Knockdown of CXCL3 also upregulated TNF-α, I L-1β, and IL-18 in LPS-treated BEAS-2B and HPAE cells. Moreover, knockdown of CXCL3 suppressed the activation of mitogen-activated protein kinases (MAPKs) signaling in LPS-treated BEAS-2B and HPAE cells through downregulation of p-ERK1/2, p-p38, and p-JNK. On the other hand, overexpression of CXCL3 caused completely opposite results in LPS-treated BEAS-2B and HPAE cells.Conclusion: Knockdown of CXCL3 exerted antiapoptotic and anti-inflammatory effects against LPS-treated BEAS-2B and HPAE cells, at least partially, through inactivation of MAPKs signaling, suggesting a potential strategy for the intervention of sepsis-induced acute lung injury (AU)


Assuntos
Humanos , Lesão Pulmonar Aguda/metabolismo , Quimiocinas CXC/metabolismo , Sepse/metabolismo , Apoptose , Células Epiteliais/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo
18.
Allergol. immunopatol ; 50(3): 38-46, 2022. graf
Artigo em Inglês | IBECS | ID: ibc-203448

RESUMO

Dedicator of cytokinesis protein 2 (DOCK2) is a member of the cytoskeletal dynamics pro-tein family, and is ubiquitously expressed in hematopoietic cells according to previous stud-ies. This paper was intended to explore the underlying mechanism that DOCK2 might involve in the progression of acute lung injury (ALI). Following lipopolysaccharide (LPS) induction in the purchased A549 cells, the expression level of DOCK2 was determined and its knockdown was performed by transfection. Subsequently, the viability, inflammation, oxidative stress barrier, and apoptosis of transfected A549 cells were measured to observe the alterations. Inflammation-related and apoptosis-related proteins were measured by western blot analy-sis. Finally, 8-Chlorophenylthio-cyclic monophosphate (8-CPT), ras-related C3 botulinum toxin substrate (Rac) 1 agonist, was applied to treat cells for investigating the underlying mecha-nism regarding the role of DOCK2. According to the results, DOCK2 was upregulated in LPS-induced A549 cells. Following the knockdown of DOCK2, the release of inflammatory cytokines was alleviated, accompanied by attenuated oxidative stress, barrier injury, and apoptosis of LPS-induced A549 cells. Nonetheless, this trend was reversed by further treatment of 8-CPT. In summary, DOCK2 knockdown alleviates inflammation, oxidative stress, barrier injury, and apoptosis of LPS-induced A549 cells by associating with Rac1/Rac2. These findings highlighted the therapeutic potential of DOCK2 for the treatment of ALI.© 2022 Codon Publications. Published by Codon Publications (AU)


Assuntos
Humanos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lipopolissacarídeos , Apoptose , Citocinese , Inflamação , Proteínas rac1 de Ligação ao GTP , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas
19.
Allergol. immunopatol ; 50(3): 71-76, 2022. graf
Artigo em Inglês | IBECS | ID: ibc-203452

RESUMO

Background: Acute lung injury (ALI) is a clinical syndrome characterized by hyperosmotic ­pulmonary­edema­and­ increased­alveolar­fluid.­Phospholipase­C­ epsilon-1­(PLCE1),­identified­as a member of phospholipase family, and the relationship between PLCE1 and lung injury is n ot clear.Objective: To assess the possible role of Phospholipase C Epsilon 1 (PLCE1) in Acute lung injury (ALI) progression and related mechanisms. Materials and methods:­ The­ effects­of­ LPS­ and­ PLCE1­on­ cell­ viability­and­ apoptosis­were­examined­by­ MTT­ and­ flow­ cytometry.­Also,­ the­ level­ of­ PLCE1­was­ controlled­by­ transfection­of­ its­ plasmid­and­ shRNA.­The­ inflammatory­response­in­ response­to­ PLCE1­overexpression­or­ablation was analyzed by quantitative PCR and ELISA assay. And the involvement of PKC and NF-κB­signal­pathway­were­detected­by­Immunoblot.­Results: In this study, we developed a LPS-induced ALI cell model. We found PLCE1 was upreg-ulated­in­ LPS-induced­pneumonia­cells­ and­ affected­cell­ viability.­Also,­knockdown­of­ PLCE1­reduced LPS-induced apoptosis of pneumonia cells. In addition, depletion of PLCE1 suppressed LPS-induced­secretion­of­ proinflammatory­cytokines­in­ pneumonia­cells.­Mechanically,­we­found­depletion­of­ PLCE1­inhibited­PKC­ and­ NF-κB­signal­pathway,­and­ therefore­alleviated­LPS-induced ALI.Conclusion: We therefore thought PLCE1 co (AU)uld serve as a promising drug for ALI


Assuntos
Humanos , Lesão Pulmonar Aguda/induzido quimicamente , Lipopolissacarídeos/efeitos adversos , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , NF-kappa B/metabolismo , Fosfolipases Tipo C , Transdução de Sinais
20.
Allergol. immunopatol ; 49(5): 117-124, sept. 2021. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-214775

RESUMO

Background Sepsis is a systemic inflammatory response syndrome and leads to patient’s death. Objective: To investigate the effect of myocyte enhancer factor 2 (MEF2C) on acute lung injury (ALI) with sepsis and its possible mechanism. Material and Methods The cecal ligation and puncture (CLP)-induced sepsis rat model was established. The lung injury was determined by lung wet–dry weight ratio, the concentration of inflammatory cytokines, including tumor necrosis factor-α (TNF-α), Interlukin (IL)-6, IL-1β, and IL-10, were measured by the enzyme-linked-immunosorbent serologic assay kit. The cell apoptosis was detected by TUNEL staining assay. Results Interestingly, MEF2C was down-regulated in this model. Moreover, adeno-associated virus (AAV)-MEF2C treatment markedly suppressed TNF-α, IL-1β, and IL-6 concentrations but promoted IL-10 concentration in serum in CLP-challenged rats. Besides, overexpression of MEF2C alleviates CLP-induced lung injury. Interestingly, AAV-MEF2C treatment was confirmed to suppress apoptosis in CLP-induced sepsis rats as well as promote aquaporin APQ1 expression. Mechanistically, the rescue experiments indicated that MEF2C alleviated CLP-induced lung inflammatory response and apoptosis via up-regulating AQP1. Conclusion In summary, overexpression of MEF2C suppressed CLP-induced lung inflamma-tory response and apoptosis via up-regulating AQP1, providing a novel therapeutic target for sepsis-induced ALI (AU)


Assuntos
Animais , Masculino , Ratos , Lesão Pulmonar Aguda/tratamento farmacológico , Fatores de Transcrição MEF2/uso terapêutico , Punções , Sepse , Ratos Sprague-Dawley , Aquaporinas , Modelos Animais de Doenças , Interleucina-10 , Fator de Necrose Tumoral alfa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...