Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 879
Filter
1.
Gen Physiol Biophys ; 43(5): 399-409, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140680

ABSTRACT

Acute lung injury (ALI) is a significant health condition with notable rates of morbidity and mortality globally. Long non-coding ribose nucleic acids (lncRNAs) play vital roles in mitigating various inflammation-related diseases, including ALI. The study aimed to investigate the functional role and molecular mechanisms of lncRNA SNHG1 on ALI in lipopolysaccharide (LPS)-treated A549 cells and in LPS-induced ALI mice. The expression of SNHG1 was initially examined in LPS-treated A549 cells. We further demonstrated the critical function of SNHG1 through various cellular assessments following SNHG1 knockdown, including cell counting kit (CCK)-8 assay, flow cytometry analysis, as well as enzyme-linked immunosorbent assay (ELISA). Reducing SNHG1 levels hindered the negative effects of LPS on cell viability, apoptosis, and inflammation. Moreover, SNHG1 acted as a negative regulator for miR-199a-3p, which targeted downstream ROCK2. Depletion of miR-199a-3p or enhanced expression of ROCK2 abolished the protective effects of SNHG1 knockdown on LPS-induced apoptosis and inflammation. Consistently, silencing SNHG1 alleviated LPS-induced lung injury in mice, demonstrating its potential therapeutic benefits in managing ALI. Overall, this study sheds light on the role of SNHG1 in modulating inflammation and apoptosis in ALI through the miR-199a-3p/ROCK2 pathway, offering new insights for the treatment of this condition.


Subject(s)
Acute Lung Injury , Gene Knockdown Techniques , Lipopolysaccharides , MicroRNAs , RNA, Long Noncoding , rho-Associated Kinases , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Mice , Humans , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Male , A549 Cells , Signal Transduction , Mice, Inbred C57BL , Apoptosis/genetics
2.
Nat Commun ; 15(1): 7241, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174557

ABSTRACT

Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid ß-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.


Subject(s)
Acute Lung Injury , Alveolar Epithelial Cells , Carnitine O-Palmitoyltransferase , Fatty Acids , Mitochondria , Oxidation-Reduction , Respiratory Distress Syndrome , Animals , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Mitochondria/metabolism , Alveolar Epithelial Cells/metabolism , Fatty Acids/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/immunology , Acute Lung Injury/genetics , Mice , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/genetics , Male , Humans , Chemokine CXCL2/metabolism , Chemokine CXCL2/genetics , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Mice, Knockout , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Inflammation/metabolism , Inflammation/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/immunology , Adenosine Triphosphate/metabolism , Pneumonia/metabolism , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/genetics
3.
Sci Rep ; 14(1): 20006, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39198493

ABSTRACT

Acute lung injury (ALI) is a severe clinical respiratory condition characterized by high rates of mortality and morbidity, for which effective treatments are currently lacking. In this study, lipopolysaccharide (LPS) was used to induce ALI mice, demonstrating the efficacy of tetramethylpyrazine (TMP) in ameliorating ALI. Subsequent we perfored high-throughput sequencing analysis and used Targetscan 8.0 and miRWalk 3.0 databases to predict the interaction between microRNAs and destrin (DSTN), ultimately identifying miR-369-3p as the focus of the investigation. The adenovirus carrying miR-369-3p was administered one week prior to LPS-induced in order to assess its potential efficacy in ameliorating ALI in mice. The findings indicated that the overexpression of miR-369-3p resulted in enhanced lung function, reduced pulmonary edema, inflammation, and permeability in LPS-induced ALI mice, while the suppression of miR-369-3p exacerbated the damage in these mice. Furthermore, the beneficial effects of TMP on LPS-induced ALI were negated by the downregulation of miR-369-3p. The results of our study demonstrate that TMP mitigates LPS-induced ALI through upregulation of miR-369-3p. Consequently, the findings of this study advocate for the clinical utilization of TMP in ALI treatment, with miR-369-3p emerging as a promising target for future ALI interventions.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , MicroRNAs , Pyrazines , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Pyrazines/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Male , Disease Models, Animal , Mice, Inbred C57BL
4.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3837-3847, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39099357

ABSTRACT

The study investigates the therapeutic effects and mechanisms of ginsenoside Rg_1(GRg_1) on sepsis-induced acute lung injury(SALI). A murine model of SALI was created using cecal ligation and puncture(CLP) surgery, and mice were randomly assigned to groups for GRg_1 intervention. Survival and body weight changes were recorded, lung function was assessed with a non-invasive lung function test system, and lung tissue damage was evaluated through HE staining. The content and expression of inflammatory factors were measured by ELISA and qRT-PCR. Apoptosis was examined using flow cytometry and TUNEL staining. The activation and expression of apoptosis-related molecules cysteinyl aspartate specific proteinase 3(caspase-3), B-cell lymphoma-2(Bcl-2), Bcl-2 associated X protein(Bax), and endoplasmic reticulum stress-related molecules protein kinase R-like endoplasmic reticulum kinase(PERK), eukaryotic initiation factor 2α(eIF2α), activating transcription factor 4(ATF4), and C/EBP homologous protein(CHOP) were studied using Western blot and qRT-PCR. In addition, an in vitro model of lipopolysaccharide(LPS)-induced lung alveolar epithelial cell injury was used, with the application of the endoplasmic reticulum stress inducer tunicamycin to validate the action mechanism of GRg_1. RESULTS:: indicated that, when compared to the model group, GRg_1 intervention significantly enhanced the survival time of CLP mice, mitigated body weight loss, and improved impaired lung function indices. The GRg_1-treated mice also displayed reduced lung tissue pathological scores, a reduced lung tissue wet-to-dry weight ratio, and lower protein content in the bronchoalveolar lavage fluid. Serum levels of interleukin-6(IL-6), interleukin-1ß(IL-1ß), and tumor necrosis factor-α(TNF-α), as well as the mRNA expressions of these cytokines in lung tissues, were decreased. There was a notable decrease in the proportion of apopto-tic alveolar epithelial cells, and down-regulated expressions of caspase-3, Bax, PERK, eIF2α, ATF4, and CHOP and up-regulated expression of Bcl-2 were observed. In vitro findings showed that the apoptosis-lowering and apoptosis-related protein down-regulating effects of GRg_1 were significantly inhibited with the co-application of tunicamycin. Altogether, GRg_1 reduces apoptosis of alveolar epithelial cells, inhibits inflammation in the lungs, alleviates lung injury, and enhances lung function, possibly through the PERK/eIF2α/ATF4/CHOP pathway.


Subject(s)
Activating Transcription Factor 4 , Acute Lung Injury , Alveolar Epithelial Cells , Apoptosis , Eukaryotic Initiation Factor-2 , Ginsenosides , Sepsis , Transcription Factor CHOP , eIF-2 Kinase , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Ginsenosides/pharmacology , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Mice , Apoptosis/drug effects , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Sepsis/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Male , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Humans , Endoplasmic Reticulum Stress/drug effects , Mice, Inbred C57BL
5.
Respir Res ; 25(1): 263, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956592

ABSTRACT

BACKGROUND: Aberrant activation of macrophages is associated with pathogenesis of acute lung injury (ALI). However, the potential pathogenesis has not been explored. OBJECTIVES: We aimed to identify whether histone deacetylase (HDAC) 10 is involved in lipopolysaccharide (LPS)-exposed ALI and reveal the underlying pathogenesis by which it promotes lung inflammation in LPS-exposed ALI via modifying P62 with deacetylation. METHODS: We constructed an ALI mice model stimulated with LPS to determine the positive effect of Hdac10 deficiency. Moreover, we cultured murine alveolar macrophage cell line (MH-S cells) and primary bone marrow-derived macrophages (BMDMs) to explore the pro-inflammatory activity and mechanism of HDAC10 after LPS challenge. RESULTS: HDAC10 expression was increased both in mice lung tissues and macrophage cell lines and promoted inflammatory cytokines production exposed to LPS. Hdac10 deficiency inhibited autophagy and inflammatory response after LPS stimulation. In vivo, Hdac10fl/fl-LysMCre mice considerably attenuated lung inflammation and inflammatory cytokines release exposed to LPS. Mechanistically, HDAC10 interacts with P62 and mediates P62 deacetylation at lysine 165 (K165), by which it promotes P62 expression and increases inflammatory cytokines production. Importantly, we identified that Salvianolic acid B (SAB), an HDAC10 inhibitor, reduces lung inflammatory response in LPS-stimulated ALI. CONCLUSION: These results uncover a previously unknown role for HDAC10 in regulating P62 deacetylation and aggravating lung inflammation in LPS-induced ALI, implicating that targeting HDAC10 is an effective therapy for LPS-exposed ALI.


Subject(s)
Acute Lung Injury , Histone Deacetylases , Lipopolysaccharides , Lysine , Mice, Inbred C57BL , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Lipopolysaccharides/toxicity , Mice , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/deficiency , Lysine/metabolism , Mice, Knockout , Male , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Myeloid Cells/metabolism
6.
Respir Res ; 25(1): 276, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010105

ABSTRACT

BACKGROUND: The pathogenesis of acute lung injury (ALI) involves a severe inflammatory response, leading to significant morbidity and mortality. N6-methylation of adenosine (m6A), an abundant mRNA nucleotide modification, plays a crucial role in regulating mRNA metabolism and function. However, the precise impact of m6A modifications on the progression of ALI remains elusive. METHODS: ALI models were induced by either intraperitoneal injection of lipopolysaccharide (LPS) into C57BL/6 mice or the LPS-treated alveolar type II epithelial cells (AECII) in vitro. The viability and proliferation of AECII were assessed using CCK-8 and EdU assays. The whole-body plethysmography was used to record the general respiratory functions. M6A RNA methylation level of AECII after LPS insults was detected, and then the "writer" of m6A modifications was screened. Afterwards, we successfully identified the targets that underwent m6A methylation mediated by METTL3, a methyltransferase-like enzyme. Last, we evaluated the regulatory role of METTL3-medited m6A methylation at phosphatase and tensin homolog (Pten) in ALI, by assessing the proliferation, viability and inflammation of AECII. RESULTS: LPS induced marked damages in respiratory functions and cellular injuries of AECII. The m6A modification level in mRNA and the expression of METTL3, an m6A methyltransferase, exhibited a notable rise in both lung tissues of ALI mice and cultured AECII cells subjected to LPS treatment. METTL3 knockdown or inhibition improved the viability and proliferation of LPS-treated AECII, and also reduced the m6A modification level. In addition, the stability and translation of Pten mRNA were enhanced by METTL3-mediated m6A modification, and over-expression of PTEN reversed the protective effect of METTL3 knockdown in the LPS-treated AECII. CONCLUSIONS: The progression of ALI can be attributed to the elevated levels of METTL3 in AECII, as it promotes the stability and translation of Pten mRNA through m6A modification. This suggests that targeting METTL3 could offer a novel approach for treating ALI.


Subject(s)
Acute Lung Injury , Alveolar Epithelial Cells , Cell Proliferation , Methyltransferases , Mice, Inbred C57BL , PTEN Phosphohydrolase , RNA, Messenger , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/pathology , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Mice , Cell Proliferation/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Male , RNA, Messenger/metabolism , Cell Survival/physiology , Cell Survival/drug effects , Methylation , Adenosine/analogs & derivatives , Adenosine/metabolism , Lipopolysaccharides/toxicity , RNA Stability , Cells, Cultured
7.
Free Radic Biol Med ; 222: 588-600, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996820

ABSTRACT

Emerging evidence has reported that acute lung injury (ALI), characterized by inflammation and oxidative stress in airway epithelium, is regulated by programmed cell death. Ferroptosis, a regulated form of cell death spurred by uncontrolled lipid peroxidation, has been proven to implicate various diseases. Inhibiting ferroptosis represents a feasible strategy for ALI through the suppression of lipid peroxidation, while the mechanism remains to be further elucidated. Here, we identified Sequestosome 1 (SQSTM1) as a negative regulator of airway epithelium ferroptosis during ALI. SQSTM1 knockdown cells manifested higher sensitivity to ferroptosis. Mechanistically, SQSTM1 was found to directly interact with vitamin D receptor (VDR) through its nuclear receptor (NR) box motif, facilitating its nuclear translocation and initiating autophagy at the transcriptional level. To further validate these findings, an in vivo preventive model utilizing spermidine, a proven inducer of SQSTM1 was established. The results consistently demonstrated that spermidine supplementation significantly induced SQSTM1 and ameliorated ALI by mitigating airway epithelial ferroptosis. Notably, these effects were abrogated in the absence of SQSTM1. Taken together, this study identified SQSTM1 as a negative regulator of airway epithelium ferroptosis in a VDR-mediated autophagy manner, making it a potential therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury , Autophagy , Ferroptosis , Receptors, Calcitriol , Sequestosome-1 Protein , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/genetics , Ferroptosis/genetics , Ferroptosis/drug effects , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Animals , Humans , Mice , Male , Mice, Inbred C57BL , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Oxidative Stress , Lipid Peroxidation/drug effects
8.
Free Radic Biol Med ; 222: 638-649, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39019096

ABSTRACT

BACKGROUND: Acute lung injury (ALI) can cause multiple organ dysfunction and a high mortality rate. Inflammatory responses, oxidative stress, and immune damage contribute to their pathogenic mechanisms. We studied the role of the newly discovered lncRNA, Lncmir155hg, in ALI. METHODS: The levels of Lncmir155hg and miR-450b-5p from mice with ALI were detected via polymerase chain reaction analysis (qRT-PCR) and Fluorescence in situ hybridization (FISH). Pathological changes of lung were detected by HE (hematoxylin and eosin) staining, and HIF-1α, NOD-like receptor 3 (NLRP3) and caspase-1 protein changes were detected by immunohistochemistry. MLE-12 cells proliferation was detected by Cell-Counting Kit 8 analysis, and reactive oxygen species (ROS) was detected via flow cytometry. NLRP3, apoptosis-associated speck-like protein (ASC), and caspase-1 were measured via western blotting, and enzyme-linked immunosorbent assays detected the expression of Inflammatory factors. Lncmir155hg, miR-450b-5p, miR-450b-5p, and HIF-1α targets were predicted using LncTar and miRWalk and confirmed in dual-luciferase reporter assays. RESULTS: In mice with ALI and MLE-12 cells induced by lipopolysaccharide (LPS), Lncmir155hg was high-expressed and miR-450b-5p was low-expressed. sh-Lncmir155hg reduced the damage of lung tissue, the production of inflammatory cytokines and oxidative stress reaction induced by LPS,miR-450b-5p reverses the effect of Lncmir155hg in mice. sh-Lncmir155hg decreased the protein levels of HIF-1α, NLRP3 and caspase-1 in LPS-induced lung tissues. sh-Lncmir155hg + miR-450b-5p inhibitor transfection reversed the effect of sh-Lncmir155hg on the expression of HIF-1α, NLRP3 and caspase-1. Lncmir155hg knockdown induced proliferation and inhibited NLRP3-inflammasome activation and oxidative stress in MLE-12 cells of ALI. miR-450b-5p was identified to have binding with Lncmir155hg, and inhibition of miR-450b-5p eliminated the effect of si-Lncmir155hg in MLE-12 cells of ALI. More importantly, miR-450b-5p was directly combined with HIF-1α, miR-450b-5p mimic promoted proliferation and inhibited activation of inflammasome associated proteins and reaction of oxidative stress, and HIF-1α overexpression abolished these effects. CONCLUSION: Lncmir155hg aggravated ALI via the miR-450b-5p/HIF-1α axis.


Subject(s)
Acute Lung Injury , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammasomes , MicroRNAs , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , RNA, Long Noncoding , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/chemically induced , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammasomes/metabolism , Inflammasomes/genetics , Male , Gene Expression Regulation , Lipopolysaccharides/toxicity , Apoptosis/genetics , Mice, Inbred C57BL , Cell Proliferation , Reactive Oxygen Species/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Cell Line , Disease Models, Animal , Humans
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167354, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39004378

ABSTRACT

Acute lung injury (ALI) is a serious disorder characterized by the release of pro-inflammatory cytokines and cascade activation of macrophages. Ferroptosis, a form of iron-dependent cell death triggered by intracellular phospholipid peroxidation, has been implicated as an internal mechanism underlying ALI. In this study, we investigated the effects of m6A demethylase fat mass and obesity-associated protein (FTO) on the inhibition of macrophage ferroptosis in ALI. Using a mouse model of lipopolysaccharide (LPS)-induced ALI, we observed the induction of ferroptosis and its co-localization with the macrophage marker F4/80, suggesting that ferroptosis might be induced in macrophages. Ferroptosis was promoted during LPS-induced inflammation in macrophages in vitro, and the inflammation was counteracted by the ferroptosis inhibitor ferrostatin-1 (fer-1). Given that FTO showed lower expression levels in the lung tissue of mice with ALI and inflammatory macrophages, we further dissected the regulatory capacity of FTO in ferroptosis. The results demonstrated that FTO alleviated macrophage inflammation by inhibiting ferroptosis. Mechanistically, FTO decreased the stability of ACSL4 mRNA via YTHDF1, subsequently inhibiting ferroptosis and inflammation by interrupting polyunsaturated fatty acid consumption. Moreover, FTO downregulated the synthesis and secretion of prostaglandin E2, thereby reducing ferroptosis and inflammation. In vivo, the FTO inhibitor FB23-2 aggravated lung injury, the inflammatory response, and ferroptosis in mice with ALI; however, fer-1 therapy mitigated these effects. Overall, our findings revealed that FTO may function as an inhibitor of the inflammatory response driven by ferroptosis, emphasizing its potential as a target for ALI treatment.


Subject(s)
Acute Lung Injury , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Coenzyme A Ligases , Ferroptosis , Inflammation , Macrophages , Animals , Male , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Cyclohexylamines , Disease Models, Animal , Ferroptosis/drug effects , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Lipopolysaccharides , Macrophages/metabolism , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred C57BL , Phenylenediamines/pharmacology , RAW 264.7 Cells
10.
Clin Sci (Lond) ; 138(13): 797-815, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38840498

ABSTRACT

IGFBP7 has been found to play an important role in inflammatory diseases, such as acute lung injury (ALI). However, the role of IGFBP7 in different stages of inflammation remains unclear. Transcriptome sequencing was used to identify the regulatory genes of IGFBP7, and endothelial IGFBP7 expression was knocked down using Aplnr-Dre mice to evaluate the endothelial proliferation capacity. The expression of proliferation-related genes was detected by Western blotting and RT-PCR assays. In the present study, we found that knockdown of IGFBP7 in endothelial cells significantly decreases the expression of endothelial cell proliferation-related genes and cell number in the recovery phase but not in the acute phase of ALI. Mechanistically, using bulk-RNA sequencing and CO-IP, we found that IGFBP7 promotes phosphorylation of FOS and subsequently up-regulates YAP1 molecules, thereby promoting endothelial cell proliferation. This study indicated that IGFBP7 has diverse roles in different stages of ALI, which extends the understanding of IGFBP7 in different stages of ALI and suggests that IGFBP7 as a potential therapeutic target in ALI needs to take into account the period specificity of ALI.


Subject(s)
Acute Lung Injury , Cell Proliferation , Endothelial Cells , Insulin-Like Growth Factor Binding Proteins , Animals , Humans , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor Binding Proteins/genetics , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , YAP-Signaling Proteins/metabolism
11.
J Toxicol Sci ; 49(7): 289-299, 2024.
Article in English | MEDLINE | ID: mdl-38945840

ABSTRACT

BACKGROUND: Endothelial barrier dysfunction is critical for the pathogenesis of sepsis-induced acute lung injury (ALI). Lipopolysaccharide (LPS)-stimulated human pulmonary microvascular endothelial cells (HPMECs) are widely used as the cell model of sepsis-associated ALI for exploration of endothelial barrier dysfunction. Dickkopf (DKK) family proteins were reported to mediate endothelial functions in various diseases. The present study explored the effect of Dickkopf-3 (DKK3) on endothelial barrier permeability, angiogenesis, and tight junctions in LPS-stimulated HPMECs. METHODS: RT-qPCR was required for detecting DKK3 and miR-98-3p expression. The angiogenesis of HPMECs was evaluated by tube formation assays. Monolayer permeability of HPMECs was examined by Transwell rhodamine assays. The protein expression of DKK3 and tight junctions in HPMECs was measured via western blotting. Luciferase reporter assay was used to verify the interaction between miR-98-3p and DKK3. RESULTS: LPS treatment inhibited angiogenetic ability while increasing the permeability of HPMECs. DKK3 expression was upregulated while miR-98-3p level was reduced in LPS-treated HPMECs. DKK3 knockdown alleviated HPMEC injury triggered by LPS stimulation. MiR-98-3p targeted DKK3 in HPMECs. Overexpression of miR-98-3p protects HPMECs from the LPS-induced endothelial barrier dysfunction, and the protective effect was reversed by DKK3 overexpression. CONCLUSIONS: MiR-98-3p ameliorates LPS-evoked pulmonary microvascular endothelial barrier dysfunction in sepsis-associated ALI by targeting DKK3.


Subject(s)
Acute Lung Injury , Adaptor Proteins, Signal Transducing , Endothelial Cells , Lipopolysaccharides , MicroRNAs , Sepsis , MicroRNAs/genetics , MicroRNAs/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Humans , Sepsis/complications , Sepsis/metabolism , Endothelial Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lung/blood supply , Cells, Cultured , Tight Junctions/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Capillary Permeability/drug effects
12.
Redox Biol ; 74: 103194, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852200

ABSTRACT

Elevated lactate levels are a significant biomarker of sepsis and are positively associated with sepsis-related mortality. Sepsis-associated lung injury (ALI) is a leading cause of poor prognosis in clinical patients. However, the underlying mechanisms of lactate's involvement in sepsis-associated ALI remain unclear. In this study, we demonstrate that lactate regulates N6-methyladenosine (m6A) modification levels by facilitating p300-mediated H3K18la binding to the METTL3 promoter site. The METTL3-mediated m6A modification is enriched in ACSL4, and its mRNA stability is regulated through a YTHDC1-dependent pathway. Furthermore, short-term lactate stimulation upregulates ACSL4, which promotes mitochondria-associated ferroptosis. Inhibition of METTL3 through knockdown or targeted inhibition effectively suppresses septic hyper-lactate-induced ferroptosis in alveolar epithelial cells and mitigates lung injury in septic mice. Our findings suggest that lactate induces ferroptosis via the GPR81/H3K18la/METTL3/ACSL4 axis in alveolar epithelial cells during sepsis-associated ALI. These results reveal a histone lactylation-driven mechanism inducing ferroptosis through METTL3-mediated m6A modification. Targeting METTL3 represents a promising therapeutic strategy for patients with sepsis-associated ALI.


Subject(s)
Coenzyme A Ligases , Ferroptosis , Methyltransferases , Sepsis , Methyltransferases/metabolism , Methyltransferases/genetics , Animals , Sepsis/metabolism , Sepsis/complications , Mice , Humans , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Lung Injury/metabolism , Lung Injury/etiology , Lung Injury/pathology , Lung Injury/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/genetics , Male , Disease Models, Animal , Lactic Acid/metabolism
13.
Free Radic Biol Med ; 222: 199-210, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38901501

ABSTRACT

BACKGROUND: Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is intricately involved in modulating the inflammatory response in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Nevertheless, the myeloid PTEN governing Hippo-YAP pathway mediated oxidative stress and inflammation in lipopolysaccharide (LPS)-induced ALI remains to be elucidate. METHODS: The floxed Pten (PtenFL/FL) and myeloid-specific Pten knockout (PtenM-KO) mice were intratracheal instill LPS (5 mg/kg) to establish ALI, then Yap siRNA mix with the mannose-conjugated polymers was used to knockdown endogenous macrophage YAP in some PtenM-KO mice before LPS challenged. The bone marrow-derived macrophages (BMMs) from PtenFL/FL and PtenM-KO mice were obtained, and BMMs were transfected with CRISPR/Cas9-mediated glycogen synthase kinase 3 Beta (GSK3ß) knockout (KO) or Yes-associated protein (YAP) KO vector subjected to LPS (100 ng/ml) challenged or then cocultured with MLE12 cells. RESULTS: Here, our findings demonstrate that myeloid-specific PTEN deficiency exerts a protective against LPS-induced oxidative stress and inflammation dysregulated in ALI model. Moreover, ablation of the PTEN-YAP axis in macrophages results in reduced nuclear factor-E2-related factor-2 (NRF2) expression, a decrease in antioxidant gene expression, augmented levels of free radicals, lipid and protein peroxidation, heightened generation of pro-inflammatory cytokines, ultimately leading to increased apoptosis in MLE12 cells. Mechanistically, it is noteworthy that the deletion of myeloid PTEN promotes YAP translocation and regulates NRF2 expression, alleviating LPS-induced ALI via the inhibition of GSK3ß and MST1 binding. CONCLUSIONS: Our study underscores the crucial role of the myeloid PTEN-YAP-NRF2 axis in governing oxidative stress and inflammation dysregulated in ALI, indicating its potential as a therapeutic target for ALI.


Subject(s)
Acute Lung Injury , Adaptor Proteins, Signal Transducing , Inflammation , Lipopolysaccharides , Macrophages , Mice, Knockout , Oxidative Stress , PTEN Phosphohydrolase , YAP-Signaling Proteins , Animals , Male , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Lipopolysaccharides/toxicity , Macrophages/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Signal Transduction , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics
14.
Int Immunopharmacol ; 137: 112510, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38897130

ABSTRACT

Acute lung injury (ALI), a critical complication observed in various clinical disorders, is characterized by widespread inflammation, neutrophil infiltration, and proinflammatory cytokine production. This study showed that the recently identified non-coding RNA ISIR and its human homolog gene AK131315 played a role in regulating lipopolysaccharide (LPS)-induced inflammatory responses. ISIR and AK131315 increased the production of inflammatory cytokines in LPS-stimulated macrophages, and exogenous ISIR aggravated LPS-induced lung inflammation in an animal model of ALI. Mechanistically, ISIR promoted LPS-triggered NF-κB and MAPK signaling and the transcription of proinflammatory cytokines by enhancing TAK1 activation. Furthermore, a significant correlation was observed between AK131315 expression and pulmonary infectious caused by Gram-negative bacteria, suggesting that AK131315 plays an important role in bacterial infections. Altogether, these findings indicate that ISIR regulates LPS-induced inflammation and AK131315 is involved in the pathogenesis of bacterial infections.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , MAP Kinase Kinase Kinases , NF-kappa B , Lipopolysaccharides/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/immunology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , NF-kappa B/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Male , Cytokines/metabolism , Cytokines/genetics , RAW 264.7 Cells , Inflammation/genetics , Inflammation/chemically induced , MAP Kinase Signaling System/drug effects , Signal Transduction , Disease Models, Animal
15.
Cells ; 13(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38727303

ABSTRACT

Small interfering RNA (siRNA) holds significant therapeutic potential by silencing target genes through RNA interference. Current clinical applications of siRNA have been primarily limited to liver diseases, while achievements in delivery methods are expanding their applications to various organs, including the lungs. Cholesterol-conjugated siRNA emerges as a promising delivery approach due to its low toxicity and high efficiency. This study focuses on developing a cholesterol-conjugated anti-Il6 siRNA and the evaluation of its potency for the potential treatment of inflammatory diseases using the example of acute lung injury (ALI). The biological activities of different Il6-targeted siRNAs containing chemical modifications were evaluated in J774 cells in vitro. The lead cholesterol-conjugated anti-Il6 siRNA after intranasal instillation demonstrated dose-dependent therapeutic effects in a mouse model of ALI induced by lipopolysaccharide (LPS). The treatment significantly reduced Il6 mRNA levels, inflammatory cell infiltration, and the severity of lung inflammation. IL6 silencing by cholesterol-conjugated siRNA proves to be a promising strategy for treating inflammatory diseases, with potential applications beyond the lungs.


Subject(s)
Acute Lung Injury , Cholesterol , Interleukin-6 , RNA, Small Interfering , Animals , Mice , Acute Lung Injury/therapy , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Cell Line , Cholesterol/chemistry , Cholesterol/pharmacology , Disease Models, Animal , Interleukin-6/metabolism , Interleukin-6/genetics , Lipopolysaccharides , Lung/pathology , Lung/metabolism , Mice, Inbred C57BL , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
16.
Sci Rep ; 14(1): 11160, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750066

ABSTRACT

Sepsis is a systemic inflammatory response syndrome resulting from the invasion of the human body by bacteria and other pathogenic microorganisms. One of its most prevalent complications is acute lung injury, which places a significant medical burden on numerous countries and regions due to its high morbidity and mortality rates. MicroRNA (miRNA) plays a critical role in the body's inflammatory response and immune regulation. Recent studies have focused on miR-21-5p in the context of acute lung injury, but its role appears to vary in different models of this condition. In the LPS-induced acute injury model of A549 cells, there is differential expression, but the specific mechanism remains unclear. Therefore, our aim is to investigate the changes in the expression of miR-21-5p and SLC16A10 in a type II alveolar epithelial cell injury model induced by LPS and explore the therapeutic effects of their targeted regulation. A549 cells were directly stimulated with 10 µg/ml of LPS to construct a model of LPS-induced cell injury. Cells were collected at different time points and the expression of interleukin 1 beta (IL-1ß), tumor necrosis factor-α (TNF-α) and miR-21-5p were measured by RT-qPCR and western blot. Then miR-21-5p mimic transfection was used to up-regulate the expression of miR-21-5p in A549 cells and the expression of IL-1ß and TNF-α in each group of cells was measured by RT-qPCR and western blot. The miRDB, TargetScan, miRWalk, Starbase, Tarbase and miR Tarbase databases were used to predict the miR-21-5p target genes and simultaneously, the DisGeNet database was used to search the sepsis-related gene groups. The intersection of the two groups was taken as the core gene. Luciferase reporter assay further verified SLC16A10 as the core gene with miR-21-5p. The expression of miR-21-5p and SLC16A10 were regulated by transfection or inhibitors in A549 cells with or without LPS stimulation. And then the expression of IL-1ß and TNF-α in A549 cells was tested by RT-qPCR and western blot in different groups, clarifying the role of miR-21-5p-SLC16A10 axis in LPS-induced inflammatory injury in A549 cells. (1) IL-1ß and TNF-α mRNA and protein expression significantly increased at 6, 12, and 24 h after LPS stimulation as well as the miR-21-5p expression compared with the control group (P < 0.05). (2) After overexpression of miR-21-5p in A549 cells, the expression of IL-1ß and TNF-α was significantly reduced after LPS stimulation, suggesting that miR-21-5p has a protection against LPS-induced injury. (3) The core gene set, comprising 51 target genes of miR-21-5p intersecting with the 1448 sepsis-related genes, was identified. This set includes SLC16A10, TNPO1, STAT3, PIK3R1, and FASLG. Following a literature review, SLC16A10 was selected as the ultimate target gene. Dual luciferase assay results confirmed that SLC16A10 is indeed a target gene of miR-21-5p. (4) Knocking down SLC16A10 expression by siRNA significantly reduced the expression of IL-1ß and TNF-α in A549 cells after LPS treatment (P < 0.05). (5) miR-21-5p inhibitor increased the expression levels of IL-1ß and TNF-α in A549 cells after LPS stimulation (P < 0.05). In comparison to cells solely transfected with miR-21-5p inhibitor, co-transfection of miR-21-5p inhibitor and si-SLC6A10 significantly reduced the expression of IL-1ß and TNF-α (P < 0.05). MiR-21-5p plays a protective role in LPS-induced acute inflammatory injury of A549 cells. By targeting SLC16A10, it effectively mitigates the inflammatory response in A549 cells induced by LPS. Furthermore, SLC16A10 holds promise as a potential target for the treatment of acute lung injury.


Subject(s)
Acute Lung Injury , Alveolar Epithelial Cells , Amino Acid Transport Systems, Neutral , MicroRNAs , Humans , A549 Cells , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Gene Expression Regulation , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/toxicity , MicroRNAs/genetics , MicroRNAs/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism
18.
J Cell Mol Med ; 28(10): e18280, 2024 May.
Article in English | MEDLINE | ID: mdl-38758159

ABSTRACT

Acute lung injury (ALI) is featured with a robust inflammatory response. Angiopoietin-like protein 2 (ANGPTL2), a pro-inflammatory protein, is complicated with various disorders. However, the role of ANGPTL2 in ALI remains to be further explored. The mice and MH-S cells were administrated with lipopolysaccharide (LPS) to evoke the lung injury in vivo and in vitro. The role and mechanism of ANGPTL was investigated by haematoxylin-eosin, measurement of wet/dry ratio, cell count, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling, reverse transcription quantitative polymerase chain reaction, immunofluorescence, enzyme-linked immunosorbent assay, detection of autophagic flux and western blot assays. The level of ANGPTL2 was upregulated in lung injury. Knockout of ANGPTL2 alleviated LPS-induced pathological symptoms, reduced pulmonary wet/dry weight ratio, the numbers of total cells and neutrophils in BALF, apoptosis rate and the release of pro-inflammatory mediators, and modulated polarization of alveolar macrophages in mice. Knockdown of ANGPTL2 downregulated the level of pyroptosis indicators, and elevated the level of autophagy in LPS-induced MH-S cells. Besides, downregulation of ANGPTL2 reversed the LPS-induced the expression of leukocyte immunoglobulin (Ig)-like receptor B2 (LILRB2) and triggering receptor expressed on myeloid cells 2 (TREM2), which was reversed by the overexpression of LILRB2. Importantly, knockdown of TREM2 reversed the levels of autophagy- and pyroptosis-involved proteins, and the contents of pro-inflammatory factors in LPS-induced MH-S cells transfected with si ANGPTL2, which was further inverted with the treatment of rapamycin. Therefore, ANGPTL2 silencing enhanced autophagy to alleviate alveolar macrophage pyroptosis via reducing LILRB2-mediated inhibition of TREM2.


Subject(s)
Acute Lung Injury , Angiopoietin-Like Protein 2 , Autophagy , Macrophages, Alveolar , Pyroptosis , Receptors, Immunologic , Animals , Male , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Autophagy/genetics , Gene Knockdown Techniques , Lipopolysaccharides , Macrophages, Alveolar/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Pyroptosis/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 296-302, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710513

ABSTRACT

Objective To evaluate the effects of heme oxygenase-1 (HO-1) gene deletion on immune cell composition and inflammatory injury in lung tissues of mice with lipopolysaccharide (LPS)-induced acute lung injury (ALI). Methods C57BL/6 wild-type (WT) mice and HO-1 conditional-knockout (HO-1-/-) mice on the same background were randomly divided into four groups (n=5 in every group): WT control group, LPS-treated WT group, HO-1-/- control group and LPS-treated HO-1-/- group. LPS-treated WT and HO-1-/- groups were injected with LPS (15 mg/kg) through the tail vein to establish ALI model, while WT control group and HO-1-/- control group were injected with an equivalent volume of normal saline through the tail vein, respectively. Twelve hours later, the mice were sacrificed and lung tissues from each group were collected for analysis. Histopathological alterations of lung tissues were assessed by HE staining. The levels of mRNA expression of tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), and IL-6 were determined by PCR. The percentages of neutrophils (CD45+CD11b+Ly6G+Ly6C-), total monocytes (CD45+CD11b+Ly6Chi), pro-inflammatory monocyte subsets (CD45+CD11b+Ly6ChiCCR2hi) and total macrophages (CD45+CD11b+F4/80+), M1 macrophage (CD45+CD11b+F4/80+CD86+), M2 macrophage (CD45+CD11b+F4/80+CD206+), total T cells (CD45+CD3+), CD3+CD4+ T cells, CD3+CD8+ T cells and myeloid suppressor cells (MDSCs, CD45+CD11b+Gr1+) were detected by flow cytometry. Results Compared with the corresponding control groups, HE staining exhibited increased inflammation in the lung tissues of both LPS-treated WT and HO-1-/- model mice; mRNA expression levels of TNF-α, IL-1ß and IL-6 were up-regulated; the proportions of neutrophils, total monocytes, pro-inflammatory monocyte subsets, MDSCs and total macrophages increased significantly. The percentage of CD3+, CD3+CD4+ and CD3+CD8+ T cells decreased significantly. Under resting-state, compared with WT control mice, the proportion of neutrophils, monocytes and pro-inflammatory monocyte subset increased in lung tissues of HO-1-/- control mice, while the proportion of CD3+ and CD3+CD8+ T cells decreased. Compared with LPS-treated WT mice, the mRNA expression levels of TNF-α and IL-1ß were up-regulated in lung tissues of LPS-treated HO-1-/- mice; the proportion of total monocytes, pro-inflammatory monocyte subsets, M1 macrophages and M1/M2 ratio increased greatly; the percentage of CD3+CD8+ T cells decreased significantly. Conclusion The deletion of HO-1 affects the function of the lung immune system and aggravates the inflammatory injury after LPS stimulation in ALI mice.


Subject(s)
Acute Lung Injury , Heme Oxygenase-1 , Lipopolysaccharides , Lung , Mice, Inbred C57BL , Mice, Knockout , Animals , Male , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/genetics , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Lung/immunology , Lung/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(4): 430-434, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38813641

ABSTRACT

Hyperoxia-induced acute lung injury (HALI) is an important complication of clinical oxygen therapy, which is mainly characterized by acute respiratory distress syndrome (ARDS) in adults and broncho-pulmonary dysplasia (BPD) in infants. HALI seriously affects the prognosis and quality of life of patients, so it has received more and more attention. However, the pathogenesis of HALI is complex and unclear, and there is no clear treatment method at present. Non-coding RNA (ncRNA) is an important type of functional RNA transcriptome. Due to the lack of effective open reading frame, ncRNA does not have the function of coding proteins. However, ncRNA can still regulate gene expression at multiple levels and affect the occurrence and development of many diseases. In recent years, a large number of in vitro and in vivo studies have shown that ncRNA is involved in the pathogenesis of HALI and is of great significance. This article reviews the expression and significance of ncRNA in HALI, in order to provide new diagnosis and treatment ideas for the prevention and treatment of HALI.


Subject(s)
Acute Lung Injury , Hyperoxia , RNA, Untranslated , Humans , Hyperoxia/complications , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Acute Lung Injury/therapy , RNA, Untranslated/genetics , Animals
SELECTION OF CITATIONS
SEARCH DETAIL