Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cytokine ; 173: 156441, 2024 01.
Article in English | MEDLINE | ID: mdl-37995394

ABSTRACT

Macrophages have recently been discovered to assume a significant role in the progression of cryptococcosis. However, the potential involvement of macrophage-derived exosomes in the pathogenesis of cryptococcosis remains uncertain. In this study, we investigated the changes of microRNAs in macrophage exosomes (exo-miRNAs) in cryptococcal infections and the role of markedly altered exo-miRNAs in the modulation of Human Umbilical Vein Endothelial Cells (HUVEC) permeability and ROS accumulation and pyroptosis in Human Bronchial Epithelioid Cells (BEAS-2B). Techniques such as microarray analysis and real-time quantitative PCR were used to detect different exo-miRNAs and to screen for the most highly expressed exo-miRNAs. Then its mimics were transfected into HUVEC to study its effect on the monolayer permeability of HUVEC. Finally, the relationship between this exo-miRNAs and the ROS accumulation and pyroptosis was verified by bioinformatics analysis. The results showed that five exo-miRNAs were overexpressed and two exo-miRNAs were reduced, among which, exo-miR-4449 was expressed at the highest level. Exo-miR-4449 could be internalized by HUVEC and enhanced its monolayer permeability. Moreover, exo-miR-4449 was found to promote ROS accumulation and pyroptosis in BEAS-2B through HIC1 pathway. Thus, exo-miR-4449 plays an important role in the pathogenesis of cryptococcosis and holds promise as a significant biomarker for treatment.


Subject(s)
Cryptococcosis , Cryptococcus , MicroRNAs , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Pyroptosis/genetics , Cryptococcus/metabolism , Reactive Oxygen Species/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Macrophages/metabolism , Cryptococcosis/metabolism , Cryptococcosis/pathology , Kruppel-Like Transcription Factors
2.
J Biochem Mol Toxicol ; 36(7): e23060, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35355364

ABSTRACT

In this study, we explored to detect the effects and mechanism of bone-marrow-derived mesenchymal stem cells (BMSCs) on ventilator-induced lung injury (VILI). We transplanted BMSCs in mice and then induced VILI using mechanical ventilation (MV) treatment. The pathological changes, the content of PaO2 and PaCO2 , wet/dry weight ratio (W/D) of the lung, levels of tumor necrosis factor-α and interleukin-6 in bronchoalveolar lavage fluid, and apoptosis were detected. The autophagy-associated factor p62, LC3, and Beclin-1 expression were analyzed by western blot. The quantitative polymerase chain reaction was applied to detect abnormally expressed microRNAs, including miR-155-5p. Subsequently, we overexpressed miR-155-5p in VILI mice to detect the effects of miR-155-5p on MV-induced lung injury. Then, we carried out bioinformatics analysis to verify the BMSCs-regulated miR-155-5p that target messenger RNA. It was observed that BMSCs transplantation mitigated the severity of VILI in mice. BMSCs transplantation reduced lung inflammation, strengthened the arterial oxygen partial pressure, and reduced apoptosis and the W/D of the lung. BMSCs promoted autophagy of pulmonary endothelial cells accompanied by decreased p62 and increased LC3 II/I and Beclin-1. BMSCs increased the levels of miR-155-5p in VILI mice. Overexpression of miR-155-5p alleviated lung injury in VILI mice following reduced apoptosis and increased autophagy. Finally, TAB2 was identified as a downstream target of miR-155-5p and regulated by miR-155-5p. BMSCs may protect lung tissues from MV-induced injury, inhibit lung inflammation, promote autophagy through upregulating of miR-155-5p.


Subject(s)
Mesenchymal Stem Cell Transplantation , MicroRNAs , Ventilator-Induced Lung Injury , Animals , Autophagy , Beclin-1 , Endothelial Cells/metabolism , Mice , MicroRNAs/genetics , Ventilator-Induced Lung Injury/therapy
3.
Eur J Histochem ; 67(4)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38085254

ABSTRACT

This study aimed to explore the role and mechanism of umbilical cord mesenchymal stem cells (UCMSCs) in regulating inflammation of bronchial epithelial cells. Transforming growth factor beta-1 (TGF-ß1) was used to induce inflammation in human bronchial epithelial cells. Cell proliferation was detected through CCK8 and cell apoptosis was detected by Annexin V and propidium iodide double staining. E-cadherin and α-smooth muscle actin (α-SMA) were detected by immunofluorescence, and tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in culture medium supernatant were detected by ELISA. The expression of E-cadherin, α-SMA, Sonic hedgehog (Shh), Gli1 and Snail was detected by Western blot analysis. Compared with the control group, bronchial epithelial cells treated with TGF-ß1 showed significantly decreased proliferation, increased apoptosis, increased secretion of TNF-α and IL-6, increased expression of α-SMA, Shh, Gli1 and Snail and decreased E-cadherin expression. However, co-culture with UCMSCs inhibited TGF-ß1-induced changes in human bronchial epithelial cell proliferation, apoptosis, secretion of TNF-α and IL-6 and activation of the Hedgehog pathway. In conclusion, UCMSCs have protective effects on TGF-ß1-induced inflammation in human bronchial epithelial cells by regulating the Hedgehog pathway.


Subject(s)
Mesenchymal Stem Cells , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/pharmacology , Cadherins/metabolism , Epithelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism
4.
Clin Respir J ; 17(7): 663-671, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37406999

ABSTRACT

OBJECTIVES: To evaluate the outcomes of bronchial artery embolization (BAE) for the treatment of massive hemoptysis in patients with pulmonary tuberculosis and identify risk factors that influence recurrence. METHODS: A total of 81 patients with massive hemoptysis who underwent BAE between January 2014 and December 2017 were retrospectively reviewed. All of the patients had either a history of pulmonary tuberculosis or a current diagnosis of pulmonary tuberculosis. Follow-up ranged from 18 to 66 months. RESULTS: Hemoptysis was stopped or markedly decreased, with subsequent clinical improvement in 73 patients, while 11 patients experienced recurrence during the follow-up period. Systemic-pulmonary shunts and clinical failure showed a statistically significant correlation with the recurrence rate. The cumulative non-recurrence rate was 95.3% for 3 months and 81.9% for more than 24 months. Complications were common (12.5%), but self-limiting. CONCLUSIONS: BAE is a safe and effective treatment option for the control of massive hemoptysis in pulmonary tuberculosis patients. Systemic-pulmonary shunts and clinical failure are the risk factors for recurrence.


Subject(s)
Embolization, Therapeutic , Tuberculosis, Pulmonary , Humans , Hemoptysis/etiology , Hemoptysis/therapy , Retrospective Studies , Risk Factors , Treatment Outcome , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/therapy , Embolization, Therapeutic/adverse effects , Bronchial Arteries
5.
J Microbiol ; 60(4): 402-410, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35157222

ABSTRACT

Acute lung injury caused by Candida albicans could result in high mortality and morbidity. MicroRNA-155 (miR-155) and suppressor of cytokine signaling 1 (SOCS1) have been believed to play a key in the regulation of inflammatory response. Whether miR-155/SOCS1 axis could regulate the acute lung injury caused by C. albicans has not been reported. The acute lung injury animal model was established with acute infection of C. albicans. miR-155 inhibitor, miR-155 mimic, and sh-SOCS1 were constructed. The binding site between miR-155 and SOCS1 was identified with dual luciferase reporter assay. Knockdown of miR-155 markedly inhibited the germ tube formation of C. albicans. Knockdown of miR-155 significantly up-regulated the expression of SOCS1, and the binding site between miR-155 and SOCS1 was identified. Knockdown of miR-155 improved the acute lung injury, suppressed inflammatory factors and fungus loading through SOCS1. Knockdown of SOCS1 greatly reversed the influence of miR-155 inhibitor on the cell apoptosis in vitro. The improvement of acute lung injury caused by C. albicans, suppression of inflammatory response and C. albicans infection, and inhibitor of cell apoptosis were achieved by knocking down miR-155 through SOCS1. This research might provide a new thought for the prevention and treatment of acute lung injury caused by C. albicans through targeting miR-155/SOCS1 axis.


Subject(s)
Acute Lung Injury , Candida albicans , Candidiasis , MicroRNAs , Suppressor of Cytokine Signaling 1 Protein , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Animals , Candida albicans/genetics , Candida albicans/metabolism , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Down-Regulation , Inflammation/metabolism , Inflammation/microbiology , MicroRNAs/genetics , MicroRNAs/metabolism , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL