Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Postgrad Med J ; 100(1182): 209-218, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38147883

ABSTRACT

This article reviews the correlation between presepsin and sepsis and the resulting acute respiratory distress syndrome (ARDS). ARDS is a severe complication of sepsis. Despite the successful application of protective mechanical ventilation, restrictive fluid therapy, and neuromuscular blockade, which have effectively reduced the morbidity and mortality associated with ARDS, the mortality rate among patients with sepsis-associated ARDS remains notably high. The challenge lies in the prediction of ARDS onset and the timely implementation of intervention strategies. Recent studies have demonstrated significant variations in presepsin (PSEP) levels between patients with sepsis and those without, particularly in the context of ARDS. Moreover, these studies have revealed substantially elevated PSEP levels in patients with sepsis-associated ARDS compared to those with nonsepsis-associated ARDS. Consequently, PSEP emerges as a valuable biomarker for identifying patients with an increased risk of sepsis-associated ARDS and to predict in-hospital mortality.


Subject(s)
Respiratory Distress Syndrome , Sepsis , Humans , Sepsis/complications , Sepsis/therapy , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/therapy , Respiration, Artificial/methods , Biomarkers , Hospital Mortality , Peptide Fragments , Lipopolysaccharide Receptors
2.
FASEB J ; 36(8): e22475, 2022 08.
Article in English | MEDLINE | ID: mdl-35899478

ABSTRACT

Recent findings suggest that extracellular heat shock protein 90α (eHSP90α) promotes pulmonary fibrosis, but the underlying mechanisms are not well understood. Aging, especially cellular senescence, is a critical risk factor for idiopathic pulmonary fibrosis (IPF). Here, we aim to investigate the role of eHSP90α on cellular senescence in IPF. Our results found that eHSP90α was upregulated in bleomycin (BLM)-induced mice, which correlated with the expression of senescence markers. This increase in eHSP90α mediated fibroblast senescence and facilitated mitochondrial dysfunction. eHSP90α activated TGF-ß signaling through the phosphorylation of the SMAD complex. The SMAD complex binding to p53 and p21 promoters triggered their transcription. In vivo, the blockade of eHSP90α with 1G6-D7, a specific eHSP90α antibody, in old mice attenuated the BLM-induced lung fibrosis. Our findings elucidate a crucial mechanism underlying eHSP90α-induced cellular senescence, providing a framework for aging-related fibrosis interventions.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis , Animals , Bleomycin/toxicity , Cellular Senescence , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta/metabolism
3.
Technol Cult ; 63(3): 921-923, 2022.
Article in English | MEDLINE | ID: mdl-35848278
4.
FASEB J ; 36(6): e22359, 2022 06.
Article in English | MEDLINE | ID: mdl-35621121

ABSTRACT

Asthma is a disease characterized by airway epithelial barrier destruction, chronic airway inflammation, and airway remodeling. Repeated damage to airway epithelial cells by allergens in the environment plays an important role in the pathophysiology of asthma. Ferroptosis is a novel form of regulated cell death mediated by lipid peroxidation in association with free iron-mediated Fenton reactions. In this study, we explored the contribution of ferroptosis to house dust mite (HDM)-induced asthma models. Our in vivo and in vitro models showed labile iron accumulation and enhanced lipid peroxidation with concomitant nonapoptotic cell death upon HDM exposure. Treatment with ferroptosis inhibitors deferoxamine (DFO) and ferrostatin-1 (Fer-1) illuminated the role of ferroptosis and related damage-associated molecular patterns in HDM-treated airway epithelial cells. Furthermore, DFO and Fer-1 reduced HDM-induced airway inflammation in model mice. Mechanistically, NCOA4-mediated ferritin-selective autophagy (ferritinophagy) was initiated during ferritin degradation in response to HDM exposure. Together, these data suggest that ferroptosis plays an important role in HDM-induced asthma and that ferroptosis may be a potential treatment target for HDM-induced asthma.


Subject(s)
Asthma , Ferroptosis , Animals , Epithelial Cells/metabolism , Ferritins/metabolism , Inflammation , Iron/metabolism , Mice , Pyroglyphidae
5.
Int Immunopharmacol ; 104: 108504, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35026657

ABSTRACT

Pulmonary fibrosis is a fatal lung disease for which no effective treatment is available. Previous studies have shown that the expression of programmed cell death-Ligand (PD-L1) is significantly increased in pulmonary fibrosis, and that this is related to the occurrence of this disease. However, the underlying mechanism is not clear. To clarify the efficacy and mechanism of an anti-PD-L1 monoclonal antibody (anti-PD-L1 mAb) as a treatment for pulmonary fibrosis, we conducted histopathological, molecular, and functional analyses in a mouse model of bleomycin-induced pulmonary fibrosis and a cell model of fibrosis induced by transforming growth factor-beta 1 (TGF-ß1). Our results indicate that PD-L1 is highly expressed in the lung fibrosis model. The anti-PD-L1 mAb significantly alleviated bleomycin-induced lung structural disorders and collagen deposition in mice and inhibited the proliferation, migration, activation and extracellular matrix deposition of TGF-ß1-induced lung fibroblasts. Interestingly, the anti-PD-L1 mAb could also alleviate the autophagy impairment observed in pulmonary fibrosis. The potential mechanism is through the downregulation of the PI3K/Akt/mTOR signaling pathway. Our study provides evidence of the crucial ability of anti-PD-L1 mAbs to activate autophagy in the context of pulmonary fibrosis, providing a new strategy for the treatment of this disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Autophagy/drug effects , B7-H1 Antigen/metabolism , Bleomycin , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1
6.
Front Immunol ; 13: 1090773, 2022.
Article in English | MEDLINE | ID: mdl-36685596

ABSTRACT

Introduction: Catecholamines such as norepinephrine or epinephrine have been reported to participate in the development of acute respiratory distress syndrome (ARDS) by activating adrenergic receptors (ARs). But the role of α1-AR in this process has yet to be elucidated. Methods: In this study, ARDS mouse model was induced by intratracheal instillation of lipopolysaccharide. After treatment with α1-AR agonist phenylephrine or antagonist prazosin, lung pathological injury, alveolar barrier disruption and inflammation, and haemodynamic changes were evaluated. Cytokine levels and cell viability of alveolar macrophages were measured in vitro. Nuclear factor κB (NF-κB), mitogen-activated protein kinase, and Akt signalling pathways were analysed by western blot. Results: It showed that α1-AR activation alleviated lung injuries, including reduced histopathological damage, cytokine expression, and inflammatory cell infiltration, and improved alveolar capillary barrier integrity of ARDS mice without influencing cardiovascular haemodynamics. In vitro experiments suggested that α1-AR stimulation inhibited secretion of TNF-α, IL-6, CXCL2/MIP-2, and promoted IL-10 secretion, but did not affect cell viability. Moreover, α1-AR stimulation inhibited NF-κB and enhanced ERK1/2 activation without significantly influencing p38, JNK, or Akt activation. Discussion: Our studies reveal that α1-AR stimulation could ameliorate lipopolysaccharide-induced lung injury by inhibiting NF-κB and promoting ERK1/2 to suppress excessive inflammatory responses of alveolar macrophages.


Subject(s)
Lung Injury , Respiratory Distress Syndrome , Mice , Animals , NF-kappa B/metabolism , Macrophages, Alveolar/metabolism , MAP Kinase Signaling System , Lung Injury/chemically induced , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Distress Syndrome/metabolism , Cytokines/metabolism , Receptors, Adrenergic/metabolism
7.
Front Pharmacol ; 12: 739220, 2021.
Article in English | MEDLINE | ID: mdl-34880752

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive fatal disease characterized by interstitial remodeling, with high lethality and a lack of effective medical therapies. Tetrandrine has been proposed to present anti-fibrotic effects, but the efficacy and mechanisms have not been systematically evaluated. We sought to study the potential therapeutic effects and mechanisms of tetrandrine against lung fibrosis. The anti-fibrotic effects of tetrandrine were evaluated in bleomycin-induced mouse models and TGF-ß1-stimulated murine lung fibroblasts. We performed Chromatin Immunoprecipitation (ChIP), Immunoprecipitation (IP), and mRFP-GFP-MAP1LC3B adenovirus construct to investigate the novel mechanisms of tetrandrine-induced autophagy. Tetrandrine decreased TGF-ß1-induced expression of α-smooth muscle actin, fibronectin, vimentin, and type 1 collagen and proliferation in fibroblasts. Tetrandrine restored TGF-ß1-induced impaired autophagy flux, accompanied by enhanced interaction of SQSTM1 and MAP1LC3-Ⅱ. ChIP studies revealed that tetrandrine induced autophagy via increasing binding of NRF2 and SQSTM1 promoter. Furthermore, tetrandrine inhibited TGF-ß1-induced phosphorylation of mTOR by reducing activation of Rheb. In vivo tetrandrine suppressed the bleomycin-induced expression of fibrotic markers and improved pulmonary function. Our data suggest that protective effect of tetrandrine against lung fibrosis might be through promoting Rheb-mTOR and NRF2-SQSTM1 mediated autophagy. Tetrandrine may thus be potentially employed as a novel therapeutic medicine against IPF.

8.
Front Pharmacol ; 12: 744826, 2021.
Article in English | MEDLINE | ID: mdl-34603058

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal disease in which the normal alveolar network is gradually replaced by fibrotic scars. Current evidence suggests that metabolic alterations correlate with myofibroblast activation in IPF. Anlotinib has been proposed to have antifibrotic effects, but the efficacy and mechanisms of anlotinib against lung fibrosis have not been systematically evaluated. The antifibrotic effects of anlotinib were evaluated in bleomycin-induced mouse models and transforming growth factor-beta 1 (TGF-ß1)-stimulated lung fibroblasts. We measured lactate levels, 2-NBDG glucose uptake and the extracellular acidification rate (ECAR) to assess glycolysis in fibroblasts. RNA-protein coimmunoprecipitation (RIP) and polysome analyses were performed to investigate novel mechanisms of glycolytic reprogramming in pulmonary fibrosis. We found that anlotinib diminished myofibroblast activation and inhibited the augmentation of glycolysis. Moreover, we show that PCBP3 posttranscriptionally increases PFKFB3 expression by promoting its translation during myofibroblast activation, thus promoting glycolysis in myofibroblasts. Regarding mechanism, anlotinib exerts potent antifibrotic effects by downregulating PCBP3, reducing PFKFB3 translation and inhibiting glycolysis in myofibroblasts. Furthermore, we observed that anlotinib had preventative and therapeutic antifibrotic effects on bleomycin-induced pulmonary fibrosis. Therefore, we identify PCBP3 as a protein involved in the regulation of glycolysis reprogramming and lung fibrogenesis and propose it as a therapeutic target for pulmonary fibrosis. Our data suggest that anlotinib has antifibrotic effects on the lungs, and we provide a novel mechanism for this effect. Anlotinib may constitute a novel and potent candidate for the treatment of pulmonary fibrosis.

9.
Front Pharmacol ; 12: 708462, 2021.
Article in English | MEDLINE | ID: mdl-34497513

ABSTRACT

Pulmonary fibrosis is characterized by alveolar epithelial cell injury, lung fibroblast proliferation, differentiation, and extracellular matrix (ECM) deposition. Our previous study indicated that extracellular HSP90α (eHSP90α) promotes pulmonary fibrosis by activating the MAPK signaling pathway. Thus, treatment with 1G6-D7 (a selective HSP90α monoclonal antibody) to antagonize eHSP90α could effectively ameliorate fibrosis. This study aimed to elucidate the mechanism underlying the effects of eHSP90α in pulmonary fibrosis by focusing on its link with endoplasmic reticulum (ER) stress. Our results showed that eHSP90α promoted lung fibroblast differentiation by activating ER stress. Treatment with the ER stress inhibitor tauroursodeoxycholate (TUDCA) or glucose-regulated protein 78 kDa (GRP78) depletion significantly abrogated the effect of eHSP90α on ER stress and fibroblast activation. In addition, eHSP90α induced ER stress in fibroblasts via the phosphoinositide-4,5-bisphosphate 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway, which could be blocked by the PI3K/AKT inhibitor LY294002, and blockade of eHSP90α by 1G6-D7 markedly inhibited ER stress in the model, indicating preventive and therapeutic applications. Intriguingly, we observed that TUDCA effectively reduced the secretion of eHSP90α in vitro and in vivo. In conclusion, this study shows that the interaction between eHSP90α and ER stress plays a crucial role in pulmonary fibrosis, indicating a positive feedback in lung fibroblasts. Targeting eHSP90α and alleviating fibroblast ER stress may be promising therapeutic approaches for pulmonary fibrosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...