1
|
He W, Zhao L, Wang P, Ren M, Han Y. MiR-125b-5p ameliorates ox-LDL-induced vascular endothelial cell dysfunction by negatively regulating TNFSF4/TLR4/NF-κB signaling. BMC Biotechnol 2025; 25:11. [PMID: 39856662 PMCID: PMC11760099 DOI: 10.1186/s12896-025-00944-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Oxidized low-density lipoprotein (ox-LDL)-induced endothelial cell dysfunction plays a crucial role in the progression of atherosclerosis (AS). Although miR-125b-5p is known to be involved in cardiovascular and cerebrovascular disorders, its function in ox-LDL-induced endothelial injury is still not well understood. METHODS An in vitro AS cell model was established by exposing human umbilical vein endothelial cells (HUVECs) to 100 µg/mL ox-LDL for 24 h. A series of functional assays, including CCK-8 assay, flow cytometry, MDA and SOD kits, capillary-like network formation assay and ELISA assay were performed in vitro. TNFSF4/TLR4/NF-κB pathway-related protein expressions were measured by Western blot. Molecular mechanisms were elucidated through quantitative real-time PCR, western blot analysis, and luciferase reporter assays. RESULTS Our investigation revealed that exposure to ox-LDL led to a downregulation in miR-125b-5p, while upregulating the expression of tumor necrosis factor (ligand) superfamily, member 4 (TNFSF4), TLR4, p-p65 and p-IkBa in HUVECs in a dose-dependent manner. We confirmed TNFSF4 as a direct target of miR-125b-5p. Ox-LDL exposure led to decreased cell viability and angiogenic capacity, along with increased apoptosis, inflammation, and oxidative stress in HUVECs. These effects were reversed by overexpressing miR-125b-5p or knocking down TNFSF4. Overexpression of TNFSF4 significantly reversed the effects brought about by miR-125b-5p in HUVECs exposed to ox-LDL. Moreover, miR-125b-5p inactivated the TLR4/NF-κB signaling pathway by negatively regulating TNFSF4. CONCLUSIONS In summary, our findings demonstrate that miR-125b-5p possessed an anti-inflammatory and anti-apoptosis against ox-LDL-induced HUVEC injury by regulating the TNFSF4/TLR4/NF-κB signaling, indicating that miR-125b-5p may have an important therapeutic function for AS.
Collapse
Affiliation(s)
- Wenshuai He
- Department of Emergency Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot City, Inner Mongolia Autonomous Region, 010017, China.
| | - Limin Zhao
- Department of Emergency Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot City, Inner Mongolia Autonomous Region, 010017, China
| | - Pengfei Wang
- Department of Emergency Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot City, Inner Mongolia Autonomous Region, 010017, China
| | - Maojia Ren
- Department of Cardiology, Inner Mongolia People's Hospital, Inner Mongolia Autonomous, Region, 010017, China
| | - Yunfei Han
- Department of Emergency Medicine, Inner Mongolia People's Hospital, No. 20 Zhaowuda Road, Hohhot City, Inner Mongolia Autonomous Region, 010017, China
| |
Collapse
|
2
|
Han Y, Xi L, Leng F, Xu C, Zheng Y. Topical Delivery of microRNA-125b by Framework Nucleic Acids for Psoriasis Treatment. Int J Nanomedicine 2024; 19:2625-2638. [PMID: 38505169 PMCID: PMC10950082 DOI: 10.2147/ijn.s441353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024] Open
Abstract
Purpose Psoriasis is a chronic and recurrent inflammatory dermatitis characterized by T cell imbalance and abnormal keratinocyte proliferation. MicroRNAs (miRNAs) hold promise as therapeutic agents for this disease; however, their clinical application is hindered by poor stability and limited skin penetration. This study demonstrates the utilization of Framework Nucleic Acid (FNA) for the topical delivery of miRNAs in psoriasis treatment. Methods By utilizing miRNA-125b as the model drug, FNA-miR-125b was synthesized via self-assembly. The successful synthesis and stability of FNA-miR-125b in bovine fetal serum (FBS) were verified through gel electrophoresis. Subsequently, flow cytometry was employed to investigate the cell internalization on HaCaT cells, while qPCR determined the effects of FNA-miR-125b on cellular functions. Additionally, the skin penetration ability of FNA-miR-125b was assessed. Finally, a topical administration study involving FNA-miR-125b cream on imiquimod (IMQ)-induced psoriasis mice was conducted to evaluate its therapeutic efficacy. Results The FNA-miR-125b exhibited excellent stability, efficient cellular internalization, and potent inhibition of keratinocyte proliferation. In the psoriasis mouse model, FNA-miR-125b effectively penetrated the skin tissue, resulting in reduced epidermal thickness and PASI score, as well as decreased levels of inflammatory cytokines.
Collapse
Affiliation(s)
- Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Fang Leng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| |
Collapse
|
3
|
Chatterjee B, Sarkar M, Bose S, Alam MT, Chaudhary AA, Dixit AK, Tripathi PP, Srivastava AK. MicroRNAs: Key modulators of inflammation-associated diseases. Semin Cell Dev Biol 2024; 154:364-373. [PMID: 36670037 DOI: 10.1016/j.semcdb.2023.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/06/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
Inflammation is a multifaceted biological and pathophysiological response to injuries, infections, toxins, and inflammatory mechanisms that plays a central role in the progression of various diseases. MicroRNAs (miRNAs) are tiny, 19-25 nucleotides long, non-coding RNAs that regulate gene expression via post-transcriptional repression. In this review, we highlight the recent findings related to the significant roles of miRNAs in regulating various inflammatory cascades and immunological processes in the context of many lifestyle-related diseases such as diabetes, cardiovascular diseases, cancer, etc. We also converse on how miRNAs can have a dual impact on inflammatory responses, suggesting that regulation of their functions for therapeutic purposes may be disease-specific.
Collapse
Affiliation(s)
- Bilash Chatterjee
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mrinmoy Sarkar
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Subhankar Bose
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Md Tanjim Alam
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh, Saudi Arabia
| | | | - Prem Prakash Tripathi
- Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amit Kumar Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Gu Z, Sun M, Liu J, Huang Q, Wang Y, Liao J, Shu T, Tao M, Mao G, Pei Z, Meng W, Zhang X, Wei Y, Zhang S, Li S, Xiao K, Lu Y, Xu Q. Endothelium-Derived Engineered Extracellular Vesicles Protect the Pulmonary Endothelial Barrier in Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306156. [PMID: 38062916 PMCID: PMC10853733 DOI: 10.1002/advs.202306156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/17/2023] [Indexed: 02/10/2024]
Abstract
Acute lung injury (ALI) is a severe respiratory disease with a high mortality rate. The integrity of the pulmonary endothelial barrier influences the development and prognosis of ALI. Therefore, it has become an important target for ALI treatment. Extracellular vesicles (EVs) are promising nanotherapeutic agents against ALI. Herein, endothelium-derived engineered extracellular vesicles (eEVs) that deliver microRNA-125b-5p (miRNA-125b) to lung tissues exerting a protective effect on endothelial barrier integrity are reported. eEVs that are modified with lung microvascular endothelial cell-targeting peptides (LET) exhibit a prolonged retention time in lung tissues and targeted lung microvascular endothelial cells in vivo and in vitro. To improve the efficacy of the EVs, miRNA-125b is loaded into EVs. Finally, LET-EVs-miRNA-125b is constructed. The results show that compared to the EVs, miRNA-125b, and EVs-miRNA-125b, LET-EVs-miRNA-125b exhibit the most significant treatment efficacy in ALI. Moreover, LET-EVs-miRNA-125b is found to have an important protective effect on endothelial barrier integrity by inhibiting cell apoptosis, promoting angiogenesis, and protecting intercellular junctions. Sequencing analysis reveals that LET-EVs-miRNA-125b downregulates early growth response-1 (EGR1) levels, which may be a potential mechanism of action. Taken together, these findings suggest that LET-EVs-miRNA-125b can treat ALI by protecting the endothelial barrier integrity.
Collapse
Affiliation(s)
- Zhengyan Gu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
| | - Mingxue Sun
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Jihao Liu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Qi Huang
- School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyang110006P. R. China
| | - Yunqin Wang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Jun Liao
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
- School of MedicineShanghai UniversityShanghai200444P. R. China
| | - Tingbin Shu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Min Tao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Guanchao Mao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Zhipeng Pei
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Wenqi Meng
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinkang Zhang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Youheng Wei
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsFudan UniversityShanghai200433P. R. China
| | - Shanshan Zhang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Songling Li
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Kai Xiao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Marine Biomedical Science and Technology Innovation Platform of Lingang Special AreaShanghai200433P. R. China
| | - Ying Lu
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
| | - Qingqiang Xu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Basic Medical Center for Pulmonary DiseaseFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| |
Collapse
|
5
|
Peng W, Yang Y, Chen J, Xu Z, Lou Y, Li Q, Zhao N, Qian K, Liu F. Small Extracellular Vesicles Secreted by iPSC-Derived MSCs Ameliorate Pulmonary Inflammation and Lung Injury Induced by Sepsis through Delivery of miR-125b-5p. J Immunol Res 2023; 2023:8987049. [PMID: 37425491 PMCID: PMC10329558 DOI: 10.1155/2023/8987049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 12/07/2022] [Accepted: 05/18/2023] [Indexed: 07/11/2023] Open
Abstract
Background Sepsis-induced acute lung injury is a common critical illness in intensive care units with no effective treatment is currently available. Small extracellular vesicles, secreted by mesenchymal stem cells (MSCs), derived from human-induced pluripotent stem cells (iMSC-sEV), possess striking advantages when incorporated MSCs and iPSCs, which are considered extremely promising cell-free therapeutic agents. However, no studies have yet been conducted to systemically examine the effects and underlying mechanisms of iMSC-sEV application on attenuated lung injury under sepsis conditions. Method iMSC-sEV were intraperitoneally administered in a rat septic lung injury model induced by cecal ligation and puncture (CLP). The efficacy of iMSC-sEV was assessed by histology, immunohistochemistry, and pro-inflammatory cytokines of bronchoalveolar lavage fluid. We also evaluated the in vitro effects of iMSC-sEV on the activation of the inflammatory response in alveolar macrophages (AMs). Small RNA sequencing was utilized to detect changes in the miRNA expression profile in lipopolysaccharide (LPS)-treated AMs after iMSC-sEV administration. The effects of miR-125b-5p on the function of AMs were studied. Results iMSC-sEV were able to attenuate pulmonary inflammation and lung injury following CLP-induced lung injury. iMSC-sEV were internalized by AMs and alleviated the release of inflammatory factors by inactivating the NF-κB signaling pathway. Moreover, miR-125b-5p showed a fold-change in LPS-treated AMs after iMSC-sEV administration and was enriched in iMSC-sEV. Mechanistically, iMSC-sEV transmitted miR-125b-5p into LPS-treated AMs to target TRAF6. Conclusion Our findings demonstrated that iMSC-sEV treatment protects against septic lung injury and exerts anti-inflammatory effects on AMs at least partially through miR-125b-5p, suggesting that iMSC-sEV may provide a novel cell-free strategy for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Wei Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yun Yang
- Department of Critical Care Medicine, The People's Hospital of Fengcheng City, Yichun, Jiangxi, China
| | - Jiaquan Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zeyao Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yuanlei Lou
- Institute of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qi Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ning Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kejian Qian
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Ge C, Liu J, Fu Y, Jia L, Long L, Dong S. MicroRNA-21 protects against sepsis-induced acute lung injury by targeting phosphatase and tensin homolog in mice. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221120978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: Sepsis can cause acute lung injury (ALI), one of the leading causes of death in critically ill patients. The underlying mechanisms of sepsis-induced acute lung injury include excessive inflammation, oxidative stress, cell apoptosis, pulmonary edema, and lung tissue dysfunction. Recent studies have shown that miRNA-21 (miR-21) plays a vital role in sepsis-induced acute kidney injury. Relatively few studies have focused on the protective effects of ALI. This study aimed to determine the potential role of miR-21 in sepsis-induced ALI. Methods: We performed quantitative real-time polymerase chain reaction in a septic mouse model induced by cecal ligation and puncture (CLP) and found that miR-21 expression was upregulated. We then transfected the miR-21 precursor to upregulate miR-21 expression and miR-21 inhibitor to downregulate miR-21 expression. The sham group was exposed only to the cecum. ALI was induced by CLP, and the pre-miR-21+ALI and anti-miR-21+ALI groups were treated with miR-21 precursor or miR-21 inhibitor in the caudal vein before CLP. Pre-miR-21+ALI+PTEN inhibition (Pre-miR-21+ALI+PI) and anti-miR-21+ALI+PTEN inhibition (Anti-miR-21+ALI+PI) groups were treated with PTEN inhibition into the caudal vein after miR-21 transfection. Inflammatory cytokines, oxidative stress indicators, lung tissue cell apoptosis, oxygenation index (OI), lung wet/dry weight ratio, and lung pathological changes in the lung were observed in each group. Results: Compared with ALI mice, inflammatory response, oxidative stress indicators, lung tissue cell apoptosis, and the degree of lung injury were remarkably alleviated in Pre-miR-21+ALI mice and aggravated in Anti-miR-21+ALI mice. Western blot analysis showed that phosphatase and tensin homolog (PTEN) protein expression was decreased in CLP-treated mics. PTEN protein expression was decreased in the Pre-miR-21+ALI group but increased in the Anti-miR-21+ALI group. Moreover, the effect of miR-21 on anti-inflammatory, anti-oxidative stress, and anti-apoptosis enhanced after PTEN inhibition. Conclusion: This study revealed that miR-21 has a protective effect in sepsis-induced ALI by regulating PTEN in mice.
Collapse
Affiliation(s)
- Chen Ge
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Junhang Liu
- Department of Orthopaedics Surgery, Children’s Hospital of Hebei, Shijiazhuang, P.R. China
| | - You Fu
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Lijing Jia
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Ling Long
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Shimin Dong
- Department of Emergency, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, P.R. China
| |
Collapse
|
7
|
Yao W, Xu L, Jia X, Li S, Wei L. MicroRNA‑129 plays a protective role in sepsis‑induced acute lung injury through the suppression of pulmonary inflammation via the modulation of the TAK1/NF‑κB pathway. Int J Mol Med 2021; 48:139. [PMID: 34080641 PMCID: PMC8175065 DOI: 10.3892/ijmm.2021.4972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 05/07/2021] [Indexed: 12/18/2022] Open
Abstract
Excessive inflammatory response and apoptosis play key roles in the pathogenic mechanisms of sepsis-induced acute lung injury (ALI); however, the molecular pathways linked to ALI pathogenesis remain unclear. Recently, microRNAs (miRNAs/miRs) have emerged as important regulators of inflammation and apoptosis in sepsis-induced ALI; however, the exact regulatory mechanisms of miRNAs remain poorly understood. In the present study, the gene microarray dataset GSE133733 obtained from the Gene Expression Omnibus database was analyzed and a total of 38 differentially regulated miRNAs were identified, including 17 upregulated miRNAs and 21 downregulated miRNAs, in mice with lipopolysaccharide (LPS)-induced ALI, in comparison to the normal control mice. miR-129 was found to be the most significant miRNA, among the identified miRNAs. The upregulation of miR-129 markedly alleviated LPS-induced lung injury, as indicated by the decrease in lung permeability in and the wet-to-dry lung weight ratio, as well as the improved survival rate of mice with ALI administered miR-129 mimic. Moreover, the upregulation of miR-129 reduced pulmonary inflammation and apoptosis in mice with ALI. Of note, transforming growth factor activated kinase-1 (TAK1), a well-known regulator of the nuclear factor-κB (NF-κB) pathway, was directly targeted by miR-129 in RAW 264.7 cells. More importantly, miR-129 upregulation impeded the LPS-induced activation of the TAK1/NF-κB signaling pathway, as illustrated by the suppression of the nuclear phosphorylated-p65, p-IκB-α and p-IKKβ expression levels. Collectively, the findings of the present study indicate that miR-129 protects mice against sepsis-induced ALI by suppressing pulmonary inflammation and apoptosis through the regulation of the TAK1/NF-κB signaling pathway. This introduces the basis for future research concerning the application of miR-129 and its targets for the treatment of ALI.
Collapse
Affiliation(s)
- Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Lei Xu
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Saisai Li
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
8
|
Zhang W, Li J, Yao H, Li T. Restoring microRNA-499-5p Protects Sepsis-Induced Lung Injury Mice Via Targeting Sox6. NANOSCALE RESEARCH LETTERS 2021; 16:89. [PMID: 34019224 PMCID: PMC8140057 DOI: 10.1186/s11671-021-03534-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 04/20/2021] [Indexed: 05/05/2023]
Abstract
BACKGROUND MicroRNAs (miRs) are known to participate in sepsis; hence, we aim to discuss the protective effect of miR-499-5p targeting sex-determining region Y-related high-mobility-group box 6 (Sox6) on sepsis-induced lung injury in mice. METHODS The sepsis-induced lung injury model was established by cecal ligation and puncture. The wet/dry weight (W/D) ratio, miR-499-5p, Sox6, Caspase-3 and Caspase-9 expression in lung tissues of mice were tested. Lung injury score, collagen fibers and the degree of pulmonary fibrosis in lung tissues were determined. Further, the cell apoptosis in lung tissues was measured. The inflammatory factors contents and oxidative stress indices in bronchoalveolar lavage fluid (BALF) and lung tissues were detected via loss- and gain-of-function assays. The targeting relation between miR-499-5p and Sox6 was verified. RESULTS W/D ratio and Sox6 were increased while miR-499-5p was decreased in lung tissues of sepsis-induced lung injury mice. Restored miR-499-5p or depleted Sox6 alleviated lung tissues pathology, reduced lung injury score, collagen fibers, the degree of pulmonary fibrosis, TUNEL positive cells, Caspase-3 and Caspase-9 protein expression and inflammatory factors contents in BALF and lung tissues as well as oxidative stress response in lung tissues of sepsis-induced lung injury mice. miR-499-5p targeted Sox6. CONCLUSION High expression of miR-499-5p can attenuate cell apoptosis in lung tissues and inhibit inflammation of sepsis-induced lung injury mice via depleting Sox6, and it is a potential candidate marker and therapeutic target for sepsis-induced lung injury.
Collapse
Affiliation(s)
- Wenjie Zhang
- Intensive Care Unit (ICU), Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70, Heping Road, Weihai, 264200, Shandong, China
| | - Jing Li
- Preventive Medicine Ward, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, Shandong, China
| | - Hui Yao
- Intensive Care Unit (ICU), Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70, Heping Road, Weihai, 264200, Shandong, China
| | - Tianmin Li
- Intensive Care Unit (ICU), Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70, Heping Road, Weihai, 264200, Shandong, China.
| |
Collapse
|
9
|
Deng L, Zhao M, Wang Y, Wang X, Liu J. Dexmedetomidine Inhibits Acute Lung Injury by Upregulating miR-144 Expression in Mice. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The understanding of lung injury’s mechanisms at the molecular level is not fully completed. MicroR-NAs (miRNAs), which are part of different pathophysiological processes, are essential biological regulators that operate by suppressing target genes. A mouse model of acute lung
injury (ALI), which is triggered by lipopolysaccharide (LPS), was used to analyze miR-144 level in the ALI mice with or without dexmedetomidine treatment. Inflammation was investigated by the ratio of wet weight’s value to dry weight (W/D) of the lung, the release of cytokines TNF-α,
cytokines IL-6, and cytokines IL-1β, and MPO activity. To validate the effect of dexmedetomidine on miR-144, overex-pression and knockdown of miR-144 were applied to treat antagomir144 and agomir144. The result suggested that LPS-triggered ALI was alleviated by dexmedetomidine.
miR-144 was downregulated in ALI mice. The knockdown of miR-144 attenuated the protection of dexmedetomidine to acute lung injury. Overexpression of miR-144 attenuated the ALI, which was induced by LPS.
Collapse
Affiliation(s)
- Liqiang Deng
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| | - Min Zhao
- Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao 266000, China
| | - Yihao Wang
- Qingdao Municipal Hospital, Qingdao 266000, China
| | - Xujian Wang
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| | - Juan Liu
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| |
Collapse
|
10
|
Zheng Y, Liu J, Chen P, Lin L, Luo Y, Ma X, Lin J, Shen Y, Zhang L. RETRACTED: Exosomal miR-22-3p from human umbilical cord blood-derived mesenchymal stem cells protects against lipopolysaccharid-induced acute lung injury. Life Sci 2021; 269:119004. [PMID: 33417960 DOI: 10.1016/j.lfs.2020.119004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 2D/H/L, and 7B/F, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). Concerns were also raised over the provenance of the flow cytometry plots in Fig. 1C. The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yuanfang Zheng
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Jiyuan Liu
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Ping Chen
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Lu Lin
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Yinzhu Luo
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Xiaoying Ma
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Jincai Lin
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Ying Shen
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Liyan Zhang
- Department of Neonatology, the Affiliated Fuzhou Children Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China.
| |
Collapse
|
11
|
Liu C, Yin Z, Feng T, Zhang M, Zhou Z, Zhou Y. An integrated network pharmacology and RNA-Seq approach for exploring the preventive effect of Lonicerae japonicae flos on LPS-induced acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113364. [PMID: 32916233 PMCID: PMC7480403 DOI: 10.1016/j.jep.2020.113364] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lonicerae japonicae flos (LJF, the dried flower bud or newly bloomed flower of Lonicera japonica Thunb.), a typical herbal medicine, targets the lung, heart and stomach meridian with the function of clearing heat and detoxication. It ameliorated inflammatory responses and protected against acute lung inflammation in animal models. Acute lung injury (ALI) is a kind of inflammatory disease in which alveolar cells are damaged. However, a network pharmacology study to thoroughly investigate the mechanisms preventing ALI has not been performed. AIM OF THE STUDY In this study, we examined the main active ingredients in LJF and the protective effects of LJF on LPS-induced ALI in rats. MATERIALS AND METHODS First, the main active ingredients of LJF were screened in the TCMSP database, and the ALI-associated targets were collected from the GeneCards database. Then, we used compound-target and target-pathway networks to uncover the preventive mechanisms of LJF. Furthermore, we assessed the preventive effects of LJF in an LPS-induced rat model with the RNA-Seq technique to validate the possible molecular mechanisms of the effects of LJF in the treatment of ALI. RESULTS The network pharmacology results identified 28 main active compounds in LJF, and eight chemical components highly related to the potential targets, which were potential active compounds in LJF. In all, 94 potential targets were recognized, including IL6, TNF, PTGS2, APP, F2, and GRM5. The pathways revealed that the possible targets of LJF involved in the regulation of the IL-17 signalling pathway. Then, in vivo experiments indicated that LJF decreased the levels of proinflammatory cytokines (TNF-, IL-1, and IL-6) in serum and bronchoalveolar lavage fluid, decreased the levels of oxidative stress factors (MDA and MPO) and increased the activities of SOD and GSH-Px in lung tissue. The RNA-Seq results revealed that 7811, 775 and 3654 differentially expressed genes (DEGs) in Ctrl (control group), ALI-LJF (Lonicerae japonicae flos group) and ALI-DXSM (dexamethasone group), respectively. KEGG pathway analysis showed that the DEGs associated with immune response and inflammation signalling pathways and the IL-17 signalling pathway were significantly enriched in LJF. Compared with those in ALI, the expression of CXCL2, CXCL1, CXCL6, NFKBIA, IFNG, IL6, IL17A, IL17F, IL17C, MMP9 and TNFAIP3, which are involved in the IL-17 signalling pathway, were significantly decreased in the LJF group according to the qRT-PCR analyses. CONCLUSIONS In view of the network pharmacology and RNA-Seq results, the study identified the main active ingredient and potential targets of LJF involved in protecting against ALI, which suggests directions for further research on LJF.
Collapse
Affiliation(s)
- Chang Liu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Zhigang Yin
- College of Life Science, Guizhou University, Guiyang, Guizhou, 550025, China
| | - Tingting Feng
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Min Zhang
- Guizhou Engineering Center for Innovative Traditional Chinese Medicine and Ethnic Medicine, Guizhou University, Guiyang, Guizhou, 550025, China
| | - Zhi Zhou
- Guizhou Engineering Center for Innovative Traditional Chinese Medicine and Ethnic Medicine, Guizhou University, Guiyang, Guizhou, 550025, China
| | - Ying Zhou
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China; Guizhou Engineering Center for Innovative Traditional Chinese Medicine and Ethnic Medicine, Guizhou University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
12
|
Zhao J, Xie F, Chen R, Zhang Z, Dai R, Zhao N, Wang R, Sun Y, Chen Y. Transcription factor NF-κB promotes acute lung injury via microRNA-99b-mediated PRDM1 down-regulation. J Biol Chem 2020; 295:18638-18648. [PMID: 33109608 PMCID: PMC7939479 DOI: 10.1074/jbc.ra120.014861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/08/2020] [Indexed: 01/12/2023] Open
Abstract
Acute lung injury (ALI), is a rapidly progressing heterogenous pulmonary disorder that possesses a high risk of mortality. Accumulating evidence has implicated the activation of the p65 subunit of NF-κB [NF-κB(p65)] activation in the pathological process of ALI. microRNAs (miRNAs), a group of small RNA molecules, have emerged as major governors due to their post-transcriptional regulation of gene expression in a wide array of pathological processes, including ALI. The dysregulation of miRNAs and NF-κB activation has been implicated in human diseases. In the current study, we set out to decipher the convergence of miR-99b and p65 NF-κB activation in ALI pathology. We measured the release of pro-inflammatory cytokines (IL-1β, IL-6, and TNFα) in bronchoalveolar lavage fluid using ELISA. MH-S cells were cultured and their viability were detected with cell counting kit 8 (CCK8) assays. The results showed that miR-99b was up-regulated, while PRDM1 was down-regulated in a lipopolysaccharide (LPS)-induced murine model of ALI. Mechanistic investigations showed that NF-κB(p65) was enriched at the miR-99b promoter region, and further promoted its transcriptional activity. Furthermore, miR-99b targeted PRDM1 by binding to its 3'UTR, causing its down-regulation. This in-creased lung injury, as evidenced by increased wet/dry ratio of mouse lung, myeloperoxidase activity and pro-inflammatory cytokine secretion, and enhanced infiltration of inflammatory cells in lung tissues. Together, our findings indicate that NF-κB(p65) promotion of miR-99b can aggravate ALI in mice by down-regulating the expression of PRDM1.
Collapse
Affiliation(s)
- Jie Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China.
| | - Fei Xie
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Ruidong Chen
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Zhen Zhang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rujun Dai
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Na Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rongxin Wang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yanhong Sun
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yue Chen
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| |
Collapse
|
13
|
Dyamenahalli K, Garg G, Shupp JW, Kuprys PV, Choudhry MA, Kovacs EJ. Inhalation Injury: Unmet Clinical Needs and Future Research. J Burn Care Res 2020; 40:570-584. [PMID: 31214710 DOI: 10.1093/jbcr/irz055] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary and systemic insults from inhalation injury can complicate the care of burn patients and contribute to significant morbidity and mortality. However, recent progress in diagnosis and treatment of inhalation injury has not kept pace with the care of cutaneous thermal injury. There are many challenges unique to inhalation injury that have slowed advancement, including deficiencies in our understanding of its pathophysiology, the relative difficulty and subjectivity of bronchoscopic diagnosis, the lack of diagnostic biomarkers, the necessarily urgent manner in which decisions are made about intubation, and the lack of universal recommendations for the application of mucolytics, anticoagulants, bronchodilators, modified ventilator strategies, and other measures. This review represents a summary of critical shortcomings in our understanding and management of inhalation injury identified by the American Burn Association's working group on Cutaneous Thermal Injury and Inhalation Injury in 2018. It addresses our current understanding of the diagnosis, pathophysiology, and treatment of inhalation injury and highlights topics in need of additional research, including 1) airway repair mechanisms; 2) the airway microbiome in health and after injury; and 3) candidate biomarkers of inhalation injury.
Collapse
Affiliation(s)
- Kiran Dyamenahalli
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado School of Medicine, Aurora
| | - Gaurav Garg
- Department of Surgery, Georgetown University School of Medicine, Washington, District of Columbia
| | - Jeffrey W Shupp
- Department of Surgery, Georgetown University School of Medicine, Washington, District of Columbia
| | - Paulius V Kuprys
- Department of Surgery, Burn & Shock Trauma Research Institute, Health Sciences Division, Loyola University, Maywood, Illinois
| | - Mashkoor A Choudhry
- Department of Surgery, Burn & Shock Trauma Research Institute, Health Sciences Division, Loyola University, Maywood, Illinois
| | - Elizabeth J Kovacs
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado School of Medicine, Aurora
| |
Collapse
|
14
|
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate target gene expression by binding to sequences in messenger RNA processing. Inflammation is a protective reaction from harmful stimuli. MiRNAs can be biomarkers of diseases related to inflammation and are widely expressed in serum. However, overall changes in serum miRNA levels during inflammation have yet to be observed. Here, we selected studies published until 20 January 2020 that examined miRNAs in mouse models of inflammation. Serum microRNA, inflammation, inflammatory and mouse were used as search terms to select articles from PubMed and MEDLINE. Among the articles, sepsis and 18 related miRNAs were mainly examined. Eleven miRNAs were related to brain disease and 10 with fibrosis. Seventeen injury-induced inflammatory disease studies were included, as well as other inflammatory diseases, such as metabolic disease, vascular disease, arthritis, asthma, autoimmune disease, inflammatory bowel disease, and thyroiditis. The data described miRNA-associated downstream pathways associated with inflammation as well as mitochondrial responses, oxidative responses, apoptosis, cell signalling, and cell differentiation. We expect that the data will inform future animal inflammation-related miRNA studies.
Collapse
Affiliation(s)
- Areum Lee
- College of Korean Medicine, Dongguk University, Goyang, Republic of Korea
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, Goyang, Republic of Korea
| |
Collapse
|
15
|
Chen H, Zhang X, Zhang T, Li X, Li J, Yue Y, Wang M, Zheng Y, Fan H, Wang J, Yao M. Ambient PM Toxicity Is Correlated with Expression Levels of Specific MicroRNAs. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:10227-10236. [PMID: 32660239 DOI: 10.1021/acs.est.0c03876] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Uncertainties regarding optimized air pollution control remain as the underlying mechanisms of city-specific ambient particulate matter (PM)-induced health effects are unknown. Here, water-soluble extracts of PMs collected from four global cities via automobile air-conditioning filters were consecutively injected three times by an amount of 1, 2, and 2 mg into the blood circulation of Wistar rats after filtration by a 0.45 μm pore size membrane. Acute health effects, such as immune and inflammatory responses and hemorrhage in alveoli, were observed right after the PM extraction injection. Significant differences between cities in biomarker tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) levels were detected following the second and third PM injections. Rats' inflammatory responses varied substantially with the injections of city-specific PMs. Repeated PM extract exposure rendered the rats more vulnerable to subsequent challenges, and downregulation of certain microRNAs was observed in rats. Among the studied miRNAs, miR-125b, and miR-21 were most sensitive to the PM exposure, exhibiting a negative dose-response-type relationship with a source-specific PM (oxidative potential) toxicity (r2 = 0.63 and 0.57; p-values < 0.05). The results indicated that city-specific PMs could induce different health effects by selectively regulating different miRNAs, and that certain microRNAs, e.g., miR-125b and miR-21, may be externally mediated to neutralize PM-related health damages.
Collapse
Affiliation(s)
- Haoxuan Chen
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Xiangyu Zhang
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Ting Zhang
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Xinyue Li
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Jing Li
- Linde + Robinson Laboratories, California Institute of Technology, Pasadena, California 91125, United States
| | - Yang Yue
- Institute of Environmental Engineering, ETH Zurich, Zurich 8093, Switzerland
- Laboratory for Advanced Analytical Technologies, Empa, Swiss Federal Laboratories for Materials Science and Technology, Dubendorf 8600, Switzerland
| | - Minfei Wang
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Yunhao Zheng
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Hanqing Fan
- Department of Earth and Environmental Engineering, Columbia University, New York, New York 10027, United States
| | - Jing Wang
- Institute of Environmental Engineering, ETH Zurich, Zurich 8093, Switzerland
- Laboratory for Advanced Analytical Technologies, Empa, Swiss Federal Laboratories for Materials Science and Technology, Dubendorf 8600, Switzerland
| | - Maosheng Yao
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
16
|
Ding L, Gao X, Yu S, Sheng L. miR-128-3p enhances the protective effect of dexmedetomidine on acute lung injury in septic mice by targeted inhibition of MAPK14. J Bioenerg Biomembr 2020; 52:237-245. [PMID: 32594289 DOI: 10.1007/s10863-020-09842-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022]
Abstract
To investigate the role of miR-128-3p and MAPK14 in the dexmedetomidine treatment of acute lung injury in septic mice. SPF C57BL/6 mice were divided into 8 groups. The pathological changes and wet/dry weight ratio (W/D), PaO2, PaCO2, MDA, SOD and MPO levels in lung tissue and the serum levels of inflammation factors were observed. Dual luciferase reporter assay was used to detect the targeting relationship of miR-128-3p and MAPK14, and qPCR and WB were used to detect the expression of miR-128-3p and MAPK14. Compared with the Normal group, other groups had lower MDA, MPO, inflammatory factors levels and the expression level of MAPK14, while the content of SOD and the expression level of miR-128-3p was significantly decreased (all p < 0.05). Compared with the Model group, the contents of MDA, MPO, inflammatory factors in the DEX group and miR-128-3p mimic group were significantly decreased, and the content SOD was significantly increased, however, opposite results were occurred in oe-MAPK14 group (all p < 0.05). Compared with the DEX group, all the indicators in miR-128-3p mimic+DEX group showed significant improvement (all p < 0.05). Compared with the miR-128-3p mimic group, all the indicators were deteriorated in the miR-128-3p mimic+oe-MAPK14 group (all p < 0.05). The combination of DEX and oe-MAPK14 blocked the protective effect of dexmedetomidine on acute lung injury in septic mice. miR-128-3p can further enhance the protective effect of dexmedetomidine on acute lung injury in septic mice by targeting and inhibiting MAPK14 expression.
Collapse
Affiliation(s)
- Li Ding
- Department of Anesthesiology, The People's Hospital of Yinzhou, No.251 Baizhang East Road, Ningbo, Zhejiang Province, 315040, China.
| | - Xiang Gao
- Department of Anesthesiology, The People's Hospital of Yinzhou, No.251 Baizhang East Road, Ningbo, Zhejiang Province, 315040, China
| | - Shenghui Yu
- Department of Anesthesiology, The People's Hospital of Yinzhou, No.251 Baizhang East Road, Ningbo, Zhejiang Province, 315040, China
| | - Liufang Sheng
- Department of Anesthesiology, The People's Hospital of Yinzhou, No.251 Baizhang East Road, Ningbo, Zhejiang Province, 315040, China.
| |
Collapse
|
17
|
Li C, Han T, Li R, Fu L, Yue L. miR-26a-5p mediates TLR signaling pathway by targeting CTGF in LPS-induced alveolar macrophage. Biosci Rep 2020; 40:BSR20192598. [PMID: 32420583 PMCID: PMC7273919 DOI: 10.1042/bsr20192598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 01/08/2023] Open
Abstract
To explore the regulation mechanism of miR-26a-5p and connective tissue growth factor (CTGF) in lipopolysaccharide (LPS)-induced alveolar macrophages, which is a severe pneumonia cell model. MH-S cells were grouped into Normal group, Model group, negative control (NC) group, miR-26a-5p mimic group, oe-CTGF group, miR-26a-5p mimic + oe-CTGF group. The expression level of miR-26a-5p, CTGF and Toll-like receptor (TLR) signaling related molecules (TLR2, TLR4 and nuclear factor-κB p65) were detected by qRT-PCR and WB, respectively. The cell viability and apoptosis rate were detected by methyl thiazolyl tetrazolium (MTT) and flow cytometry, respectively. Compared with the Normal group, the expression level of miR-26a-5p was significantly decreased, while CTGF protein level was significantly increased in the Model group. Compared with the Model group, MH-S cells with miR-26a-5p overexpression showed enhanced cell viability, decreased apoptosis rate, declined expression level of TLR signaling related molecules and reduced level of tumor necrosis factor-α (TNF-α), interleukin (IL) 6 (IL-6) and IL-1β, while those with CTGF overexpression had an opposite phenotype. In conclusion, miR-26a-5p can inhibit the expression of CTGF and mediate TLR signaling pathway to inhibit the cell apoptosis and reduce the expression of proinflammatory cytokines in alveolar macrophages which is a cell model of severe pneumonia.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, He’nan Province, China
| | - Tingfeng Han
- Department of Gynecology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, He’nan Province, China
| | - Run Li
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, He’nan Province, China
| | - Liming Fu
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, He’nan Province, China
| | - Lei Yue
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, He’nan Province, China
| |
Collapse
|
18
|
Gao H, Xiao D, Gao L, Li X. MicroRNA‑93 contributes to the suppression of lung inflammatory responses in LPS‑induced acute lung injury in mice via the TLR4/MyD88/NF‑κB signaling pathway. Int J Mol Med 2020; 46:561-570. [PMID: 32468034 PMCID: PMC7307825 DOI: 10.3892/ijmm.2020.4610] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a severe inflammatory lung disease with a rapid onset. The anti-inflammatory functions of microRNA-93 (miRNA/miR-93) have been described in various types of tissue injury and disease. However, the biological role of miR-93 and its molecular mechanisms underlying the initiation and progression of ALI have not yet been reported, at least to the best of our knowledge. The present study aimed to investigate the regulatory effects exerted by miR-93 in ALI. Using an in vivo murine model of ALI induced by lipopolysaccharide (LPS), miR-93 expression was found to be downregulated in the lung tissues and bronchoalveolar lavage fluid (BALF) compared with the control group. Following agomiR-93 injection, it was observed that agomiR-93 attenuated lung injury, as evidenced by decreased lung permeability, a reduced lung wet/dry weight ratio and an increased survival rate of the mice. Concomitantly, agomiR-93 significantly reduced LPS-induced the interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α levels in BALF. Of note, Toll-like receptor 4 (TLR4), an upstream regulator of the nuclear factor (NF)-κB signaling pathway, was directly suppressed by miR-93 in RAW 264.7 cells. Importantly, agomiR-93 induced a significant suppression of the TLR4/myeloid differentiation primary response 88 (MyD88)/NF-κB signaling pathway, as demonstrated by the downregulation of MyD88, and the phosphorylation of IκB-α and p65 in the lung tissues of mice with ALI. Taken together, the findings of the present study indicate that miR-93 attenutes LPS-induced lung injury by regulating the TLR4/MyD88/NF-κB signaling pathway, suggesting that miR-93 may prove to be a potential therapeutic target for ALI.
Collapse
Affiliation(s)
- Hu Gao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Dongqiong Xiao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Linbo Gao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Xihong Li
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
19
|
Chen X, Hu J, Pan Y, Tang Z. Novel noncoding RNAs biomarkers in acute respiratory distress syndrome. Expert Rev Respir Med 2020; 14:299-306. [PMID: 31903804 DOI: 10.1080/17476348.2020.1711736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction: Acute respiratory distress syndrome (ARDS) is a very common condition associated with critically ill patients, which causes substantial morbidity and mortality. Currently, there is no effective clinical ARDS treatment strategy. Novel targets that effectively treat ARDS need to be found.Areas covered: Data sources were published articles through June 2019 in PubMed using the following keywords: 'acute respiratory distress syndrome,' 'miRNAs,' 'lncRNAs,' and 'biomarkers.' The selection of studies focused on in cellular model, animal model, and clinical studies of ARDS.Expert commentary: Accumulated evidence revealed that some specific miRNAs and lncRNAs could regulate the signaling pathways of the pathophysiology by targeting specific molecule in ARDS. The differentially expressed miRNAs exert a crucial role in apoptosis of neutrophil, antigen-presenting cells and lung epithelial cell, and the dysfunction of mitochondrial. Recently, the influence of lncRNAs upon miRNA function is also rapidly emerging. In some cases, lncRNA MALAT1 target TLR4 to mediate the p38 MAPK and NF-κB signaling pathway in ARDS rat model. In other cases, lncRNA CASC2 was found to act as a ceRNA of miR-144-3p which directly targeted AQP1 in LPS-induced A549 cell. In addition, other miRNA-lncRNA regulatory patterns in ARDS and novel biomarkers still require deeper research.
Collapse
Affiliation(s)
- Xianfeng Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Juntao Hu
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Yiping Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Zhanhong Tang
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| |
Collapse
|
20
|
Yang H, Lu Z, Huo C, Chen Y, Cao H, Xie P, Zhou H, Liu D, Liu J, Yu L. Liang-Ge-San, a Classic Traditional Chinese Medicine Formula, Attenuates Lipopolysaccharide-Induced Acute Lung Injury Through Up-Regulating miR-21. Front Pharmacol 2019; 10:1332. [PMID: 31803051 PMCID: PMC6868520 DOI: 10.3389/fphar.2019.01332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Acute lung injury (ALI) is a life-threatening disease without effective chemotherapy at present. Liang-Ge-San (LGS) is a famous traditional Chinese medicine formula, which is used to treat ALI in China. However, only a few studies have addressed the mechanisms of LGS in ALI. Purpose: To evaluate the anti-inflammatory effects of LGS on lipopolysaccharide (LPS)-induced ALI, and to explore its underlying molecular mechanism. Methods: Murine RAW264.7 cells were treated with LGS and LPS (1 μg/ml). The generation of IL-6, TNF-α, IL-1β was detected by ELISA. The protein expressions of STAT3 and P-STAT3 (Tyr705) were determined by Western blotting and fluorescence confocal microscopy. STAT3 transcriptional activity was investigated by luciferase reporter gene assay. qPCR was used to detect the expressions of microRNA-21 (miR-21), STAT3, and IL-6. DSS cross-linking assay was used to assess the change of STAT3 dimer. In vivo anti-inflammatory effects of LGS were evaluated in an ALI mouse model induced by tracheal instillation of LPS (3 mg/kg). The anti-ALI effects were evaluated by ELISA, qPCR, Western blotting, BCA, and H&E assays. Results: LGS suppressed LPS-stimulated IL-6, TNF-α, and IL-1β generation in murine macrophages RAW264.7. Moreover, LGS down-regulated protein levels of P-STAT3 (Tyr705) and STAT3, inhibited STAT3 transcriptional activity, and up-regulated miR-21. Furthermore, blockage of miR-21 antagonized the inhibitory effects of LGS on the production of IL-6 and the expressions of P-STAT3 (Tyr705) and STAT3 as well as the formation of STAT3 dimer. Critically, LGS up-regulated the expression of miR-21 and inhibited the protein expressions of STAT3 and P-STAT3 (Tyr705) to reduce the release of IL-6 and inflammatory cell infiltration as well as the degree of edema in LPS-induced ALI mice. Conclusion: LGS inhibited LPS-induced ALI through up-regulating miR-21 and subsequently inhibiting the STAT3 signaling pathway, thereby decreasing the release of IL-6.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Junshan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Linzhong Yu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Zhang X, Chu X, Gong X, Zhou H, Cai C. The expression of miR-125b in Nrf2-silenced A549 cells exposed to hyperoxia and its relationship with apoptosis. J Cell Mol Med 2019; 24:965-972. [PMID: 31713992 PMCID: PMC6933325 DOI: 10.1111/jcmm.14808] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/24/2019] [Accepted: 10/19/2019] [Indexed: 01/09/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects the quality of life of infants. At present, premature exposure to hyperoxia for extended periods of time is believed to affect the development of lung tissue and vascularity, resulting in BPD. The oxidative stress caused by hyperoxia exposure is an important risk factor for BPD in premature infants. Nuclear factor E2‐related factor 2 (Nrf2) is an important regulator of antioxidant mechanisms. As a microRNA, microRNA‐125b (miR‐125b) plays an important role in cell proliferation, differentiation and apoptosis. Although the Nrf2/ARE pathway has been extensively studied, little is known about the regulatory role of microRNAs in Nrf2 expression. In this study, the expression levels of Nrf2 and miR‐125b in the lung tissues of premature Sprague Dawley (SD) rats and A549 cells exposed to hyperoxia were detected by quantitative real‐time polymerase chain reaction (qRT‐PCR), and the apoptosis of A549 cells was detected by flow cytometry. The results showed that Nrf2 and miRNA‐125b in the lung tissues of premature rats increased significantly upon exposure to hyperoxia and played a protective role. Nrf2 was suppressed by small interfering RNA (siRNA) in A549 cells, miR‐125b was similarly inhibited, and apoptosis was significantly increased. These results suggest that miR‐125b helps protect against BPD as a downstream target of Nrf2.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyun Chu
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohui Gong
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huilin Zhou
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Cai
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Hoefel G, Tay H, Foster P. MicroRNAs in Lung Diseases. Chest 2019; 156:991-1000. [DOI: 10.1016/j.chest.2019.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
|
23
|
Hernández-Beeftink T, Guillen-Guio B, Villar J, Flores C. Genomics and the Acute Respiratory Distress Syndrome: Current and Future Directions. Int J Mol Sci 2019; 20:E4004. [PMID: 31426444 PMCID: PMC6721149 DOI: 10.3390/ijms20164004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/05/2019] [Accepted: 08/11/2019] [Indexed: 12/19/2022] Open
Abstract
The excessive hospital mortality associated with acute respiratory distress syndrome (ARDS) in adults mandates an urgent need for developing new therapies and tools for the early risk assessment of these patients. ARDS is a heterogeneous syndrome with multiple different pathogenetic processes contributing differently in different patients depending on clinical as well as genetic factors. Identifying genetic-based biomarkers holds the promise for establishing effective predictive and prognostic stratification methods and for targeting new therapies to improve ARDS outcomes. Here we provide an updated review of the available evidence supporting the presence of genetic factors that are predictive of ARDS development and of fatal outcomes in adult critically ill patients and that have been identified by applying different genomic and genetic approaches. We also introduce other incipient genomics approximations, such as admixture mapping, metagenomics and genome sequencing, among others, that will allow to boost this knowledge and likely reveal new genetic predictors of ARDS susceptibility and prognosis among critically ill patients.
Collapse
Affiliation(s)
- Tamara Hernández-Beeftink
- Research Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria 35010, Spain
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife 38010, Spain
| | - Beatriz Guillen-Guio
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife 38010, Spain
| | - Jesús Villar
- Research Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria 35010, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife 38010, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid 28029, Spain.
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife 38600, Spain.
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Santa Cruz de Tenerife 38200, Spain.
| |
Collapse
|
24
|
Wang T, Jiang L, Wei X, Dong Z, Liu B, Zhao J, Wang L, Xie P, Wang Y, Zhou S. Inhibition of miR-221 alleviates LPS-induced acute lung injury via inactivation of SOCS1/NF-κB signaling pathway. Cell Cycle 2019; 18:1893-1907. [PMID: 31208297 DOI: 10.1080/15384101.2019.1632136] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The role of inflammation response has been well documented in the development of acute lung injury (ALI). However, little is known about the functions of miRNAs in the regulation of inflammation in ALI. The aim of this study was to explore the effects of miRNAs in the regulation of inflammation in ALI and to elucidate the biomolecular mechanisms responsible for these effects. The expression profiles of miRNAs in lung tissues from lipopolysaccharide (LPS)-induced ALI mice model were analyzed using a microarray. It was observed that microRNA-221-3p (miR-221) was significantly increased in lung tissues in ALI mice. The inhibition of miR-221 attenuated lung injury including decreased lung W/D weight ratio and lung permeability and survival rates of ALI mice, as well as apoptosis, whereas its agomir-mediated upregulation exacerbated the lung injury. Concomitantly, miR-221 inhibition significantly reduced LPS-induced pulmonary inflammation, while LPS-induced pulmonary inflammation was aggravated by miR-221 upregulation. Of note, suppressor of cytokine signaling-1 (SOCS1), an effective suppressor of the NF-κB signaling pathway, was found to be a direct target of miR-221 in RAW264.7 cells. Overexpression of SOCS1 by pcDNA-SOCS1 plasmids markedly reversed the miR-221 inhibition-mediated inhibitory effects on inflammation and apoptosis in LPS-treated RAW264.7 cells. Finally, it was found that miR-221 inhibition suppressed LPS induced the activation of the NF-κB signaling pathway, as demonstrated by downregulation of phosphorylated-IκBα, p-p65 and upregulation of IκBα, whilst miR-221 overexpression had an opposite result in ALI mice. Our findings demonstrate that inhibition of miR-221 can alleviate LPS-induced inflammation via inactivation of SOCS1/NF-κB signaling pathway in ALI mice.
Collapse
Affiliation(s)
- Tao Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Lihua Jiang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Xiaoyong Wei
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Zhenghua Dong
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Bo Liu
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Junbo Zhao
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Lijuan Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Peilin Xie
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Yuxia Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Shangyou Zhou
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| |
Collapse
|
25
|
Wang Y, Zhang X, Tian J, Liu G, Li X, Shen D. Sevoflurane alleviates LPS‑induced acute lung injury via the microRNA‑27a‑3p/TLR4/MyD88/NF‑κB signaling pathway. Int J Mol Med 2019; 44:479-490. [PMID: 31173183 PMCID: PMC6605322 DOI: 10.3892/ijmm.2019.4217] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
Acute lung injury (ALI) is a critical syndrome that is associated with a high morbidity and mortality in patients. Sevoflurane has a lung protective effect in ALI as it reportedly has anti‑inflammatory and apoptotic‑regulating activity. However, the mechanism is still not entirely understood. The aim of the present study was to explore the effects of sevoflurane on lipopolysaccharide (LPS)‑induced ALI in mice and the possible mechanisms involved. The results revealed that sevoflurane treatment improved LPS‑induced lung injury, as evidenced by the reduction in mortality, lung permeability, lung wet/dry ratio and lung histopathological changes in mice. Total cell counts and the production of pro‑inflammatory cytokines [tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6] in bronchoalveolar fluid were also decreased following treatment with sevoflurane. Additionally, LPS‑triggered apoptosis in lung tissues, which was eliminated by sevoflurane. Furthermore, a miRCURY™ LNA array was employed to screen for differentially expressed microRNAs (miRs/miRNAs). Among these miRNAs, 6 were differentially expressed and were involved in the inflammatory response, but only miR‑27a‑3p (miR‑27a) was regulated by sevoflurane. Subsequently, the present study investigated whether sevoflurane exerts its function through the modulation of miR‑27a. The results demonstrated that the overexpression of miR‑27a via an injection with agomiR‑27a produced similar protections as sevoflurane, while the inhibition of miR‑27a suppressed the lung protective effects of sevoflurane in ALI mice. In addition, the present study identified that miR‑27a inhibited Toll‑like receptor 4 (TLR4) by binding to its 3'‑untranslated region. Western blot analysis demonstrated that sevoflurane may ameliorate the inflammatory response by blocking the miR‑27a/TLR4/MyD88/NF‑κB signaling pathway. The present results indicate that sevoflurane may be a viable therapeutic option in the treatment of patients with ALI.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xiaoran Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jianmin Tian
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guoze Liu
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xiaofang Li
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Dan Shen
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
26
|
Yu X, Li C. Protective effects of propofol on experimental neonatal acute lung injury. Mol Med Rep 2019; 19:4507-4513. [PMID: 30942421 DOI: 10.3892/mmr.2019.10113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/17/2018] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the effects of propofol on neonatal acute lung injury (ALI) in a rat model and to examine the molecular mechanisms underlying propofol function. A rat model of ALI was established by intraperitoneal injection of lipopolysaccharides (LPS). The neonatal rats were treated with various concentrations of propofol and a lung injury score was assessed. The protein expression levels of pro‑inflammatory cytokines was detected using ELISA. In the present study, oxidative stress was determined by measuring the level of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD) in lung tissues. Reverse transcription quantitative‑polymerase chain reaction and western blot analysis were used to examine the mRNA and protein expression levels of the factors downstream to LPS signaling pathway. Treatment with propofol significantly alleviated LPS‑induced lung injury in neonatal rats as suggested by the decreased lung injury score, increased partial pressure of oxygen and decreased lung wet‑dry weight ratio. LPS promoted the upregulation of tumor necrosis factor α (TNF‑α), interleukin (IL)‑6 and IL‑1β in lung tissues and bronchoalveolar lavage fluid from neonatal rats exhibiting ALI. Notably, treatment with propofol decreased the expression levels of these factors. Additionally, LPS caused an increase in the levels of MDA, and a decrease in SOD activity, and treatment with propofol suppressed these effects in a dose‑dependent manner. Furthermore, LPS induced the upregulation of phosphorylated (p‑)p38, nuclear factor κ‑light‑chain‑enhancer of activated B cells (NF‑κB), p‑p65, NLR family pyrin domain containing 3 (NLRP3), apoptosis‑associated speck‑like protein containing CARD and caspase‑1 in lung tissues of neonatal rats, and treatment with propofol was able to downregulate these factors in a dose‑dependent manner. Propofol alleviated lung injury in neonatal rats with LPS‑induced ALI by preventing inflammation and oxidative stress via the regulation of the activity of the p38 mitogen‑activated protein kinase/NF‑κB signaling pathway and the expression levels of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiongwu Yu
- Department of Pediatric Surgery, Maternal and Child Health Care Hospital of Qujing, Qujing, Yunnan 655000, P.R. China
| | - Chuanfeng Li
- Department of Pediatric Surgery, Maternal and Child Health Care Hospital of Qujing, Qujing, Yunnan 655000, P.R. China
| |
Collapse
|
27
|
Xiao P, Sun S, Cao J, Wang J, Li H, Hou S, Ding H, Liu Z, Fang Y, Bai S, Qin X, Yu F, Liu J, Wang X, Lv Q, Fan H. Expression profile of microRNAs in bronchoalveolar lavage fluid of rats as predictors for smoke inhalation injury. Burns 2018; 44:2042-2050. [DOI: 10.1016/j.burns.2018.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/08/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
|
28
|
Pattarayan D, Thimmulappa RK, Ravikumar V, Rajasekaran S. Diagnostic Potential of Extracellular MicroRNA in Respiratory Diseases. Clin Rev Allergy Immunol 2018; 54:480-492. [PMID: 27677501 DOI: 10.1007/s12016-016-8589-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lack of markers of subclinical disease state and clinical phenotype other than pulmonary function test has made the diagnosis and interventions of environmental respiratory diseases a major challenge. MicroRNAs (miRNAs), small non-coding single stranded RNAs, have emerged as potential disease-modifier in various environmental respiratory diseases. They can also be found in various body fluids and are remarkably stable. Because of their high stability, disease-specific expression, and the ease to detect and quantify them have raised the potential of miRNAs in body fluids to be useful clinical diagnostic biomarkers for lung disease phenotyping. In the present review, we provide a comprehensive overview of progress made in identifying miRNAs in various body fluids including blood, serum, plasma, bronchoalveolar lavage (BAL) fluid, and sputum as biomarkers for a wide range of human respiratory diseases such as acute lung injury/acute respiratory distress syndrome (ALI/ARDS), idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), and asthma. Finally, we discuss several challenges remain to be concerned and suggest few disease-specific and non-specific miRNAs to become part of future clinical practice.
Collapse
Affiliation(s)
- Dhamotharan Pattarayan
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Rajesh K Thimmulappa
- Department of Biochemistry, Jagadguru Sri Shivarathreeshwara University, Mysuru, Karnataka, India
| | - Vilwanathan Ravikumar
- Department of Biochemistry, School of Life Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Subbiah Rajasekaran
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India.
| |
Collapse
|
29
|
Zhu WD, Xu J, Zhang M, Zhu TM, Zhang YH, Sun K. MicroRNA-21 inhibits lipopolysaccharide-induced acute lung injury by targeting nuclear factor-κB. Exp Ther Med 2018; 16:4616-4622. [PMID: 30542412 PMCID: PMC6257314 DOI: 10.3892/etm.2018.6789] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 08/17/2018] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury (ALI) is a frequent, but severe complication following sepsis in patients with critical illness. The present study aimed to investigate the potential role of microRNA-21 (miR-21) in the regulation of inflammation in the ALI induced by lipopolysaccharide (LPS) in vitro and in vivo. The levels of inflammatory cytokines, tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β and IL-10, and the level of miR-21 expression were measured in the lungs of LPS-induced ALI rats and NR8383 alveolar macrophages (AMs). To confirm the regulatory effect of miR-21 in the inflammatory reactions of ALI, NR8383 cells were transfected with a mimic of miR-21 or an anti-miR-21 inhibitor, and the subsequent changes of the miR-21 level and the levels of inflammatory cytokines were detected. The underlying molecular mechanism was also investigated. LPS-induced ALI in rats resulted in significant overexpression of pro-inflammatory cytokines, TNF-α, IL-6 and IL-1β, and miR-21, but reduced the expression of the anti-inflammatory cytokine IL-10. LPS treatment also led to a higher expression level of miR-21 and increased secretion of pro-inflammatory cytokines in NR8383 cells in a time-dependent manner. Manipulation with the miR-21 mimic significantly suppressed the LPS-mediated induction of TNF-α, IL-6 and IL-1β in NR8383 cells, while that induction was upregulated when miR-21 expression was silenced via transfection with the anti-miR-21 inhibitor. Further mechanism experiments revealed that miR-21 regulates LPS-induced inflammation responses via the Toll-like receptor 4 and nuclear factor-κB (Nf-κB) signaling pathway. miR-21 negatively regulates inflammatory responses in LPS-induced ALI by targeting the NF-κB signaling pathway, providing further insight into the molecular mechanism of ALI progression.
Collapse
Affiliation(s)
- Wei-Dong Zhu
- Emergency Department, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Jia Xu
- Personnel Section, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Mao Zhang
- Emergency Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Tie-Ming Zhu
- Hepatobiliary Surgery Department, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Yun-Hua Zhang
- Intensive Care Unit, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Ke Sun
- Orthopedics Department, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| |
Collapse
|
30
|
Ju M, Liu B, He H, Gu Z, Liu Y, Su Y, Zhu D, Cang J, Luo Z. MicroRNA-27a alleviates LPS-induced acute lung injury in mice via inhibiting inflammation and apoptosis through modulating TLR4/MyD88/NF-κB pathway. Cell Cycle 2018; 17:2001-2018. [PMID: 30231673 DOI: 10.1080/15384101.2018.1509635] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Acute lung injury (ALI) is a critical clinical condition with a high mortality rate, characterized with excessive uncontrolled inflammation and apoptosis. Recently, microRNAs (miRNAs) have been found to play crucial roles in the amelioration of various inflammation-induced diseases, including ALI. However, it remains unknown the biological function and regulatory mechanisms of miRNAs in the regulation of inflammation and apoptosis in ALI. The aim of this study is to identify and evaluate the potential role of miRNAs in ALI and reveal the underlying molecular mechanisms of their effects. Here, we analyzed microRNA expression profiles in lung tissues from LPS-challenged mice using miRNA microarray. Because microRNA-27a (miR-27a) was one of the miRNAs being most significantly downregulated, which has an important role in regulation of inflammation, we investigated its function. Overexpression of miR-27a by agomir-27a improved lung injury, as evidenced by the reduced histopathological changes, lung wet/dry (W/D) ratio, lung microvascular permeability and apoptosis in the lung tissues, as well as ameliorative survival of ALI mice. This was accompanied by the alleviating of inflammation, such as the reduced total BALF cell and neutrophil counts, decreased levels of tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-6) interleukin-1β (IL-1β) and myeloperoxidase (MPO) activity in BAL fluid. Toll-like receptor 4 (TLR4), an important regulator of the nuclear factor kappa-B (NF-κB) signaling pathway, was identified as a novel target of miR-27a in RAW264.7 cells. Furthermore, our results showed that LPS stimulation increased the expression of MyD88 and NF-κB p65 (p-p65), but inhibited the expression of inhibitor of nuclear factor-κB-α (IκB-α), suggesting the activation of NF-κB signaling pathway. Further investigations revealed that agomir-miR-27a reversed the promoting effect of LPS on NF-κB signaling pathway. The results here suggested that miR-27a alleviates LPS-induced ALI in mice via reducing inflammation and apoptosis through blocking TLR4/MyD88/NF-κB activation.
Collapse
Affiliation(s)
- MinJie Ju
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - BoFei Liu
- b Department of Intensive Care Medicine , 1st People Hospital , ZhangjiaGang , China
| | - HongYu He
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - ZhunYong Gu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - YiMei Liu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - Ying Su
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - DuMing Zhu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - Jing Cang
- c Department of Anesthesiology , Zhongshan Hospital, Fudan University , Shanghai , China
| | - Zhe Luo
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| |
Collapse
|
31
|
Fu L, Zhu P, Qi S, Li C, Zhao K. MicroRNA-92a antagonism attenuates lipopolysaccharide (LPS)-induced pulmonary inflammation and injury in mice through suppressing the PTEN/AKT/NF-κB signaling pathway. Biomed Pharmacother 2018; 107:703-711. [PMID: 30138892 DOI: 10.1016/j.biopha.2018.08.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/10/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022] Open
Abstract
Overwhelming lung inflammation is a key feature of acute lung injury (ALI). MicroRNAs (miRNAs) have been implicated in the regulation diverse cellular processes including the inflammatory response. However, little is known about their functions and molecular involvement in regulating the inflammatory process in ALI. Herein, we established a lipopolysaccharide (LPS)-induced ALI mouse model and used miRNA microarray analysis to investigate and compare the miRNA expression profiles in mouse lung tissues. We found that miR-92a was markedly upregulated in the lung tissues of ALI mice compared with that in normal lung tissues. This upregulation of miR-92a in LPS-induced ALI mice was further confirmed in lung tissues, splenocytes and bronchoalveolar lavage fluid (BALF) by quantitative real-time PCR. Inhibition of miR-92a by injection with antagomir-92a markedly reduced LPS-induced pathological changes associated with lung inflammation, and reduces lung wet/dry ratio (W/D ratio), and Evans blue dye extravasation (an indicator of lung epithelial permeability). Moreover, inhibition of miR-92a ameliorated the inflammatory response by reducing the repression of proinflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6 in lung tissues. In addition, we identified that miR-92a inhibited the phosphatase and tensin homolog on chromosome ten (PTEN) by binding to its 3'-UTR in RAW264.7 murine macrophage cells. Western blot analysis demonstrated that inhibition of miR-92a may ameliorate inflammatory response through blocking PTEN/AKT/NF-κB signaling pathway in ALI mice. Collectively, these results have revealed a significant role of miR-92a in the lung inflammatory response associated with ALI in mice, and suggest that miR-92a may have potential as a prognostic indicator and novel therapeutic target for the treatment of ALI in future.
Collapse
Affiliation(s)
- Liming Fu
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, Henan, China.
| | - Ping Zhu
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, Henan, China
| | - Sanli Qi
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, Henan, China
| | - Chunyan Li
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, Henan, China
| | - Kunfang Zhao
- Department of Emergency, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, Henan, China
| |
Collapse
|
32
|
Wan QQ, Wu D, Ye QF. Candidate Genes as Biomarkers in Lipopolysaccharide-Induced Acute Respiratory Distress Syndrome Based on mRNA Expression Profile by Next-Generation RNA-Seq Analysis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4384797. [PMID: 29850515 PMCID: PMC5911337 DOI: 10.1155/2018/4384797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/09/2018] [Accepted: 01/22/2018] [Indexed: 01/04/2023]
Abstract
Up until now, the regulation mechanism at the level of gene during lipopolysaccharide- (LPS-) induced acute respiratory distress syndrome (ARDS) remains unclear. The discovery of differentially expressed genes (DEGs) between LPS-induced ARDS rats and normal rats by next-generation RNA sequencing analysis is of particular interest for the current study. These DEGs may help clinical diagnosis of ARDS and facilitate the selection of the optimal treatment strategy. Randomly, 20 rats were equally divided into 2 groups, the control group and the LPS group. Three rats from each group were selected at random for RNA sequencing analysis. Sequence reads were obtained from Illumina HiSeq4000 and mapped onto the rat reference genome RN6 using Hisat2. We identified 5244 DEGs (Fold_Change > 1.5, and P < 0.05) in the lung tissues from LPS-treated rats compared with normal rats, including 1413 upregulated and 3831 downregulated expressed genes. Lots of chemokine family members were among the most upregulated genes in LPS group. Gene ontology (GO) analysis revealed that almost all of the most enriched and meaningful biological process terms were mainly involved in the functions like immune-inflammation response and the pathways like cytokine-cytokine receptor interaction. We also found that, as for GO molecular function terms, the enriched terms were mainly related to chemokines and cytokines. DEGs with fold change over 100 were verified by quantitative real-time polymerase chain reaction and reanalyzed by gene-gene coexpression network, and the results elucidated central roles of chemokines in LPS-induced ARDS. Our results revealed some new biomarkers for uncovering mechanisms and processes of ARDS.
Collapse
Affiliation(s)
- Qi-Quan Wan
- Department of Transplant Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Di Wu
- Department of Transplant Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Qi-Fa Ye
- Department of Transplant Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, China
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, China
| |
Collapse
|
33
|
Wohlrab P, Kraft F, Tretter V, Ullrich R, Markstaller K, Klein KU. Recent advances in understanding acute respiratory distress syndrome. F1000Res 2018; 7. [PMID: 29568488 PMCID: PMC5840611 DOI: 10.12688/f1000research.11148.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2018] [Indexed: 12/17/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by acute diffuse lung injury, which results in increased pulmonary vascular permeability and loss of aerated lung tissue. This causes bilateral opacity consistent with pulmonary edema, hypoxemia, increased venous admixture, and decreased lung compliance such that patients with ARDS need supportive care in the intensive care unit to maintain oxygenation and prevent adverse outcomes. Recently, advances in understanding the underlying pathophysiology of ARDS led to new approaches in managing these patients. In this review, we want to focus on recent scientific evidence in the field of ARDS research and discuss promising new developments in the treatment of this disease.
Collapse
Affiliation(s)
- Peter Wohlrab
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Felix Kraft
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Verena Tretter
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Roman Ullrich
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Klaus Markstaller
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Klaus Ulrich Klein
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
34
|
Kreth S, Hübner M, Hinske LC. MicroRNAs as Clinical Biomarkers and Therapeutic Tools in Perioperative Medicine. Anesth Analg 2018; 126:670-681. [DOI: 10.1213/ane.0000000000002444] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
35
|
Chamnanchanunt S, Fucharoen S, Umemura T. Circulating microRNAs in malaria infection: bench to bedside. Malar J 2017; 16:334. [PMID: 28807026 PMCID: PMC5557074 DOI: 10.1186/s12936-017-1990-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/10/2017] [Indexed: 02/06/2023] Open
Abstract
Severe malaria has a poor prognosis with a morbidity rate of 80% in tropical areas. The early parasite detection is one of the effective means to prevent severe malaria of which specific treatment strategies are limited. Many clinical characteristics and laboratory testings have been used for the early diagnosis and prediction of severe disease. However, a few of these factors could be applied to clinical practice. MicroRNAs (miRNAs) were demonstrated as useful biomarkers in many diseases such as malignant diseases and cardiovascular diseases. Recently it was found that plasma miR-451 and miR-16 were downregulated in malaria infection at parasitic stages or with multi-organ failure involvement. MiR-125b, -27a, -23a, -150, 17-92 and -24 are deregulated in malaria patients with multiple organ failures. Here, the current findings of miRNAs were reviewed in relation to clinical severity of malaria infection and emphasized that miRNAs are potential biomarkers for severe malaria infection.
Collapse
Affiliation(s)
- Supat Chamnanchanunt
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand
| | - Tsukuru Umemura
- Department of Medical Technology and Sciences, International University of Health and Welfare, Ohkawa, Japan.,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
36
|
Li Q, Ge YL, Li M, Fang XZ, Yuan YP, Liang L, Huang SQ. miR-127 contributes to ventilator-induced lung injury. Mol Med Rep 2017; 16:4119-4126. [PMID: 28765901 DOI: 10.3892/mmr.2017.7109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/27/2017] [Indexed: 11/06/2022] Open
Abstract
Although it is essential in critical care medicine, mechanical ventilation often results in ventilator‑induced lung injury (VILI). Treating mice with lipopolysaccharide has been reported to upregulate the expression of miR‑127, which has been implicated in the modulation of immune responses. However, the putative roles of miR‑127 during the development of VILI have yet to be elucidated. The present study demonstrated that challenging mice with mechanical ventilation for 6 h significantly upregulated the expression of miR‑127 in bronchoalveolar lavage fluid, serum and lung tissue samples. Conversely, following the downregulation of miR‑127 expression in vivo using an adenovirus delivery system, VILI‑associated pathologies, including alterations in the pulmonary wet/dry ratio, pulmonary permeability, lung neutrophil infiltration and levels of pro‑inflammatory cytokines, were significantly attenuated. In addition, miR‑127 knockdown inhibited the ventilation‑induced activation of nuclear factor (NF)‑κB and p38 mitogen‑activated protein kinase (MAPK). These findings suggested that the upregulation of miR‑127 expression may contribute to the development of VILI, through the modulation of pulmonary permeability, the induction of histopathological alterations, and the potentiation of inflammatory responses involving NF‑κB and p38 MAPK‑associated signaling pathways.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Ya-Li Ge
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Xiang-Zhi Fang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Yan-Ping Yuan
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
37
|
Agustinho DP, de Oliveira MA, Tavares AH, Derengowski L, Stolz V, Guilhelmelli F, Mortari MR, Kuchler K, Silva-Pereira I. Dectin-1 is required for miR155 upregulation in murine macrophages in response to Candida albicans. Virulence 2016; 8:41-52. [PMID: 27294852 DOI: 10.1080/21505594.2016.1200215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The commensal fungal pathogen Candida albicans is a leading cause of lethal systemic infections in immunocompromised patients. One of the main mechanisms of host immune evasion and virulence by this pathogen is the switch from yeast form to hyphal growth morphologies. Micro RNAs (miRNAs), a small regulatory non-coding RNA, has been identified as an important part of the immune response to a wide variety of pathogens. In general, miRNAs act by modulating the intensity of inflammatory responses. miRNAs act by base-paring binding to specific sequences of target mRNAs, generally causing their silencing through mRNA degradation or translational repression. To study the impact of C. albicans cell morphology upon host miRNA expression, we investigated the differential modulation of 9 different immune response-related miRNAs in primary murine bone marrow-derived macrophages (BMDMs) exposed to either yeasts or hyphal forms of Candida albicans. Here, we show that the different growth morphologies induce distinct miRNA expression patterns in BMDMs. Interestingly, our data suggest that the C-Type lectin receptor Dectin-1 is a major PRR that orchestrates miR155 upregulation in a Syk-dependent manner. Our results suggest that PRR-mediating signaling events are key drivers of miRNA-mediated gene regulation during fungal pathogenesis.
Collapse
Affiliation(s)
- Daniel Paiva Agustinho
- a Departamento de Biologia Celular , Laboratório de Biologia Molecular, Universidade de Brasília , Brasília , DF , Brasil
| | - Marco Antônio de Oliveira
- a Departamento de Biologia Celular , Laboratório de Biologia Molecular, Universidade de Brasília , Brasília , DF , Brasil
| | - Aldo Henrique Tavares
- b Departamento de Biologia Celular , Laboratório de Imunologia Aplicada, Instituto de Biologia, Universidade de Brasília , Brasília , DF , Brasil
| | - Lorena Derengowski
- a Departamento de Biologia Celular , Laboratório de Biologia Molecular, Universidade de Brasília , Brasília , DF , Brasil
| | - Valentina Stolz
- c Department of Molecular Genetics , Max F. Perutz Laboratories, Medical University of Vienna , Vienna , Austria
| | - Fernanda Guilhelmelli
- a Departamento de Biologia Celular , Laboratório de Biologia Molecular, Universidade de Brasília , Brasília , DF , Brasil
| | - Márcia Renata Mortari
- d Departamento de Ciências Fisiológicas , Laboratório de Neurofarmacologia, Universidade de Brasília , Brasília , DF , Brasil
| | - Karl Kuchler
- c Department of Molecular Genetics , Max F. Perutz Laboratories, Medical University of Vienna , Vienna , Austria
| | - Ildinete Silva-Pereira
- a Departamento de Biologia Celular , Laboratório de Biologia Molecular, Universidade de Brasília , Brasília , DF , Brasil
| |
Collapse
|
38
|
Rajasekaran S, Pattarayan D, Rajaguru P, Sudhakar Gandhi PS, Thimmulappa RK. MicroRNA Regulation of Acute Lung Injury and Acute Respiratory Distress Syndrome. J Cell Physiol 2016; 231:2097-106. [PMID: 26790856 DOI: 10.1002/jcp.25316] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 01/20/2016] [Indexed: 12/13/2022]
Abstract
The acute respiratory distress syndrome (ARDS), a severe form of acute lung injury (ALI), is a very common condition associated with critically ill patients, which causes substantial morbidity and mortality worldwide. Despite decades of research, effective therapeutic strategies for clinical ALI/ARDS are not available. In recent years, microRNAs (miRNAs), small non-coding molecules have emerged as a major area of biomedical research as they post-transcriptionally regulate gene expression in diverse biological and pathological processes, including ALI/ARDS. In this context, this present review summarizes a large body of evidence implicating miRNAs and their target molecules in ALI/ARDS originating largely from studies using animal and cell culture model systems of ALI/ARDS. We have also focused on the involvement of miRNAs in macrophage polarization, which play a critical role in regulating the pathogenesis of ALI/ARDS. Finally, the possible future directions that might lead to novel therapeutic strategies for the treatment of ALI/ARDS are also reviewed. J. Cell. Physiol. 231: 2097-2106, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Subbiah Rajasekaran
- Department of Biotechnology, Anna University, BIT-Campus, Tiruchirappalli, Tamil Nadu, India
| | - Dhamotharan Pattarayan
- Department of Biotechnology, Anna University, BIT-Campus, Tiruchirappalli, Tamil Nadu, India
| | - P Rajaguru
- Department of Biotechnology, Anna University, BIT-Campus, Tiruchirappalli, Tamil Nadu, India
| | - P S Sudhakar Gandhi
- Department of Biotechnology, Anna University, BIT-Campus, Tiruchirappalli, Tamil Nadu, India
| | - Rajesh K Thimmulappa
- Department of Pulmonary Medicine, JSS Hospital, JSS University, Sri Shivarathreeshwara Nagara, Mysore, Karnataka, India
| |
Collapse
|
39
|
Bedreag OH, Rogobete AF, Dumache R, Sarandan M, Cradigati AC, Papurica M, Craciunescu MC, Popa DM, Luca L, Nartita R, Sandesc D. Use of circulating microRNAs as biomarkers in critically ill polytrauma patients. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.bgm.2015.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Osei ET, Florez-Sampedro L, Timens W, Postma DS, Heijink IH, Brandsma CA. Unravelling the complexity of COPD by microRNAs: it's a small world after all. Eur Respir J 2015; 46:807-18. [PMID: 26250493 DOI: 10.1183/13993003.02139-2014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 06/15/2015] [Indexed: 12/11/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease and is currently the fourth leading cause of death worldwide. Chronic inflammation and repair processes in the small airways are characteristic of COPD. Despite extensive efforts from researchers and industry, there is still no cure for COPD, hence an urgent need for new therapeutic alternatives. MicroRNAs are such an option; they are small noncoding RNAs involved in gene regulation. Their importance has been shown with respect to maintaining the balance between health and disease. Although previous reviews have discussed the expression of microRNAs related to lung disease, a detailed discussion regarding the function of differential miRNA expression in the pathogenesis of COPD is lacking.In this review we link the expression of microRNAs to different features of COPD and explain their importance in the pathogenesis of this disease. We further discuss their potential to contribute to the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Emmanuel T Osei
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands Both authors contributed equally as first authors
| | - Laura Florez-Sampedro
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands Both authors contributed equally as first authors
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands
| | - Dirkje S Postma
- University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands Both authors contributed equally as last authors
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Groningen, The Netherlands Both authors contributed equally as last authors
| |
Collapse
|