1
|
Zheng Y, Li G, Shi A, Guo J, Xu Y, Cai W. Role of miR-455-3p in the alleviation of LPS-induced acute lung injury by allicin. Heliyon 2024; 10:e39338. [PMID: 39502213 PMCID: PMC11535764 DOI: 10.1016/j.heliyon.2024.e39338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Acute lung injury (ALI) is a type of diffuse lung injury that seriously affects the survival of critically ill patients. MicroRNAs (miRNAs) can serve as promising therapeutic targets or offer insights for the development of potential therapeutic strategies against ALI. In our previous study, we demonstrated the protective effect of allicin in ALI, but the role of miRNAs in the alleviation of ALI by allicin remains unclear. This study aimed to investigate whether miRNAs mediate the effects of allicin on ALI. Cell viability and proliferation were determined using CCK-8 and EdU assays, respectively, while cellular apoptosis was analyzed by flow cytometry. The claudin-4 protein was detected by quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR) and western blotting. The binding of miR-455 with claudin-4 was determined by bioinformatics analysis and validated by dual luciferase reporter assays. The lung wet/dry ratio of lipopolysaccharide (LPS)-treated rats was determined by hematoxylin and eosin (HE) and TUNEL staining of the pulmonary tissues. The levels of myeloperoxidase (MPO), interleukin (IL)-2, IL-6, and tumor necrosis factor (TNF)-α were determined by enzyme-linked immunosorbent assay (ELISA). We observed that allicin alleviated LPS-induced injury in A549 cells, and claudin-4 knockdown reversed the protective effect of allicin in ALI. Claudin-4 is a direct target of miR-455-3p, and miR-455-3p overexpression partially reversed the protective effect of allicin in LPS-treated A549 cells. Subsequent in vivo experiments confirmed that allicin protects against LPS-induced ALI by regulating the miR-455-3p/claudin-4 axis. The study revealed that the protective effect of allicin in ALI is mediated via miR-455-3p, which suppresses the expression of claudin-4.
Collapse
Affiliation(s)
- Yueliang Zheng
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Gaoxiang Li
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Aili Shi
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junping Guo
- Rainbowfish Rehabilitation & Nursing School, Hangzhou Vocational & Technical College, Hangzhou, Zhejiang, China
| | - Yingge Xu
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenwei Cai
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Liu L, Zhang H, Chen S, Dian W, Zheng Z. Cinnamaldehyde Alleviates Alveolar Epithelial Cell Injury in ALI by Inhibiting the CaMKII Pathway. Cell Biochem Biophys 2024:10.1007/s12013-024-01544-x. [PMID: 39316262 DOI: 10.1007/s12013-024-01544-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
Alveolar epithelial cell injury plays a key role in acute lung injury (ALI) and is a vital determinant of its severity. Here, we aimed to assess the protective effects of cinnamaldehyde (CA) on lipopolysaccharide (LPS)-induced A549 cells and elucidate the underlying mechanisms. A549 cells were stimulated with 1 μg/mL LPS for 24 h to establish an alveolar epithelial cell injury model and subsequently treated with CA or Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN93. Flow cytometry, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, and lactate dehydrogenase release assays were used to evaluate apoptosis, cell viability, and lactate dehydrogenase activity, respectively. Levels of inflammatory cytokines (interleukin-6, interleukin-1β, tumor necrosis tactor-α, and interferon-γ) and oxidative stress markers (reactive oxygen species, superoxide dismutase, catalase, and malondialdehyde) were determined using enzyme-linked immunosorbent assay and specific assay kits, respectively. Furthermore, levels of apoptosis-related proteins (cleaved caspase-3, Bcl-2-associated X, and Bcl-2) and CaMKII were assessed via western blotting. CA did not exhibit significant cytotoxicity in A549 cells. It dose-dependently improved the cell viability, suppressed apoptosis, decreased cleaved caspase-3 and Bcl-2-associated X levels, and increased Bcl-2 levels in LPS-treated A549 cells. It also inhibited inflammatory factor release and oxidative stress in LPS-induced A549 cells. Similar results were observed in the KN93- and CA-treated groups. Western blotting assay revealed that CA and KN93 inhibited CaMKII pathway activation, as indicated by the reduced p-CaMKII and p-phospholamban (PLN) levels and p-CaMKII/CaMKII and p-PLN/PLN ratios. Overall, CA alleviated alveolar epithelial cell injury by inhibiting the inflammatory response and oxidative stress and inducing cell apoptosis in LPS-induced A549 cells by regulating the CaMKII pathway, serving as a potential candidate for ALI prevention and treatment.
Collapse
Affiliation(s)
- Lei Liu
- Department of Emergency, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China.
| | - Hao Zhang
- Department of Emergency, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Siming Chen
- Department of Emergency, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Wankang Dian
- Department of Emergency, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Zhou Zheng
- Department of Emergency, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Lv Y, Yu W, Xuan R, Yang Y, Xue X, Ma X. Human Placental Mesenchymal Stem Cells-Exosomes Alleviate Endothelial Barrier Dysfunction via Cytoskeletal Remodeling through hsa-miR-148a-3p/ROCK1 Pathway. Stem Cells Int 2024; 2024:2172632. [PMID: 38681858 PMCID: PMC11055650 DOI: 10.1155/2024/2172632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Background Endothelial barrier disruption of human pulmonary vascular endothelial cells (HPVECs) is an important pathogenic factor for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Mesenchymal stem cells-exosome (MSCs-Exo) represents an ideal carrier for cell-free therapy. The therapeutic implication and underlying mechanism of human placental MSCs-Exo (HPMSCs-Exo) in ALI/ARDS need to be further explored. Materials and Methods HPMSCs-Exo was extracted from HPMSCs and characterized. Then, the therapeutic effects of exosomes were evaluated in ALI mice and HPVECs. RNA-sequencing was applied to reveal the miRNA profile of HPMSCs-Exo and differentially expressed genes (DEGs) in HPMSCs-Exo-pretreated HPVECs. The targets of miRNAs were predicted by bioinformatics methods and correlated to DEGs. Finally, the role of hsa-miR-148a-3p/ROCK1 pathway in HPVECs has been further discussed. Results The results showed that HPMSCs-Exo could downregulate Rho-associated coiled-coil-containing protein kinase 1 (ROCK1), upregulate the expression of zonula occludens-1 (ZO-1) and F-actin, promote HPVECs migration and tube formation, reduce cytoskeletal disorders and cell permeability, and thus improve ALI/ARDS. RNA-sequencing revealed the DEGs were mainly enriched in cell junction, angiogenesis, inflammation, and energy metabolism. HPMSCs-Exo contains multiple miRNAs which are associated with cytoskeletal function; the expression abundance of hsa-miR-148a-3p is the highest. Bioinformatic analysis identified ROCK1 as a target of hsa-miR-148a-3p. The overexpression of hsa-miR-148a-3p in HPMSCs-Exo promoted the migration and tube formation of HPVECs and reduced ROCK1 expression. However, the overexpression of ROCK1 on HPVECs reduced the therapeutic effect of HPMSCs-Exo. Conclusions HPMSCs-Exo represents a protective regimen against endothelial barrier disruption of HPVECs in ALI/ARDS, and the hsa-miR-148a-3p/ROCK1 pathway plays an important role in this therapeutics implication.
Collapse
Affiliation(s)
- Yuzhen Lv
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Wenqin Yu
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Ruiui Xuan
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Yulu Yang
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaolan Xue
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaowei Ma
- Intensive Care Unit, Cardiocerebral Vascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| |
Collapse
|
4
|
Yao J, Cheng M, Yang F. Calycosin Attenuates Lipopolysaccharide-Induced Acute Lung Injury in Mice through the miR-375-3p/ROCK2 Axis. J INVEST SURG 2023; 36:2211166. [PMID: 37400250 DOI: 10.1080/08941939.2023.2211166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 07/05/2023]
Abstract
Objective: Septic patients are especially vulnerable to acute lung injury (ALI). Calycosin (CAL) has various promising pharmacological activities. This paper aims to expound on the role of CAL in mice with sepsis-induced ALI and the associated mechanisms.Methods: Mouse models of sepsis-induced ALI were established using lipopolysaccharide (LPS). Pulmonary histopathological changes were observed by HE staining. Cell apoptosis was assessed by TUNEL staining. Pulmonary edema was evaluated by measuring wet/dry weight. Bronchoalveolar lavage fluid (BALF) was collected to count inflammatory cells. In vitro LPS models were established using MLE-12 cells. miR-375-3p expression was determined by RT-qPCR. Cell viability and apoptosis were assessed by MTT assay and flow cytometry. Levels of inflammatory cytokines were determined by ELISA. The target relationship between miR-375-3p and ROCK2 was analyzed by the dual-luciferase assay. ROCK2 protein level was determined by Western blot.Results: miR-375-3p was weakly-expressed in mice with sepsis-induced ALI, and CAL treatment elevated miR-375-3p expression. CAL treatment mitigated pulmonary tissue damage and edema, decreased apoptosis and inflammatory cells, downregulated levels of pro-inflammatory cytokines, and upregulated levels of anti-inflammatory cytokines in mice with sepsis-induced ALI. CAL treatment increased MLE-12 cell viability and decreased apoptosis and inflammation in MLE-12 cells. Inhibition of miR-375-3p partially abrogated CAL-mediated protective action on MLE-12 cells. miR-375-3p attenuated LPS-induced MLE-12 cell injury by targeting ROCK2.Conclusion: CAL upregulates miR-375-3p to target ROCK2, thus protecting against sepsis-induced ALI in mice.
Collapse
Affiliation(s)
- Jie Yao
- Department of Intensive Care Unit, The People's Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, China
| | - Mingfeng Cheng
- Department of Intensive Care Unit, The People's Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, China
| | - Fan Yang
- Department of Intensive Care Unit, The People's Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Zhu M, Yan M, Chen J, Li H, Zhang Y. MicroRNA-129-1-3p attenuates autophagy-dependent cell death by targeting MCU in granulosa cells of laying hens under H 2O 2-induced oxidative stress. Poult Sci 2023; 102:103006. [PMID: 37595500 PMCID: PMC10458330 DOI: 10.1016/j.psj.2023.103006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
The present study aimed to investigate the mechanism of microRNA-129-1-3p (miR-129-1-3p) in regulating hydrogen peroxide (H2O2)-induced autophagic death of chicken granulosa cell by targeting mitochondrial calcium uniporter (MCU). The results indicated that the exposure of hens' ovaries to H2O2 resulted in a significant elevation in reactive oxygen species (ROS) levels, as well as the apoptosis of granulosa cells and follicular atresia. This was accompanied by an upregulation of glucose-regulated protein 75 (GRP75), voltage-dependent anion-selective channel 1 (VDAC1), MCU, mitochondria fission factor (MFF), microtubule-associated protein 1 light chain 3 (LC3) I, and LC3II expression, and a downregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and mitofusin-2 (MFN2) expression. In hens' granulosa cells, a luciferase reporter assay confirmed that miR-129-1-3p directly regulates MCU. The induction of oxidative stress through H2O2 resulted in the activation of the permeability transition pore, an overload of calcium, depolarization of the mitochondrial membrane potential, dysfunction of mitochondria-associated endoplasmic reticulum membranes (MAMs), and ultimately, autophagic cell death. The overexpression of miR-129-1-3p effectively mitigated these H2O2-induced changes. Furthermore, miR-129-1-3p overexpression in granulosa cells prevented the alterations induced by H2O2 in the expression of key proteins that play crucial roles in maintaining the integrity of MAMs and regulating autophagy, such as GRP75, VDAC1, MFN2, PTEN-induced kinase 1 (Pink1), and parkin RBR E3 ubiquitin-protein ligase (Parkin). Together, these in vitro- and in vivo-based experiments suggest that miR-129-1-3p protects granulosa cells from oxidative stress-induced autophagic cell death by downregulating the MCU-mediated mitochondrial autophagy. miR-129-1-3p/MCU calcium signaling pathway may act as a new target to alleviate follicular atresia caused by oxidative stress in laying hens.
Collapse
Affiliation(s)
- Mingkun Zhu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ming Yan
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jianfei Chen
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Huaiyu Li
- Qingdao Animal Husbandry Workstation (Qingdao Institute of Animal Science and Veterinary Medicine), Qingdao, Shandong 266100, China
| | - Yeshun Zhang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China.
| |
Collapse
|
6
|
Khan MJ, Singh P, Jha P, Nayek A, Malik MZ, Bagler G, Kumar B, Ponnusamy K, Ali S, Chopra M, Dohare R, Singh IK, Syed MA. Investigating the link between miR-34a-5p and TLR6 signaling in sepsis-induced ARDS. 3 Biotech 2023; 13:282. [PMID: 37496978 PMCID: PMC10366072 DOI: 10.1007/s13205-023-03700-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 07/28/2023] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are lung complications diagnosed by impaired gaseous exchanges leading to mortality. From the diverse etiologies, sepsis is a prominent contributor to ALI/ARDS. In the present study, we retrieved sepsis-induced ARDS mRNA expression profile and identified 883 differentially expressed genes (DEGs). Next, we established an ARDS-specific weighted gene co-expression network (WGCN) and picked the blue module as our hub module based on highly correlated network properties. Later we subjected all hub module DEGs to form an ARDS-specific 3-node feed-forward loop (FFL) whose highest-order subnetwork motif revealed one TF (STAT6), one miRNA (miR-34a-5p), and one mRNA (TLR6). Thereafter, we screened a natural product library and identified three lead molecules that showed promising binding affinity against TLR6. We then performed molecular dynamics simulations to evaluate the stability and binding free energy of the TLR6-lead molecule complexes. Our results suggest these lead molecules may be potential therapeutic candidates for treating sepsis-induced ALI/ARDS. In-silico studies on clinical datasets for sepsis-induced ARDS indicate a possible positive interaction between miR-34a and TLR6 and an antagonizing effect on STAT6 to promote inflammation. Also, the translational study on septic mice lungs by IHC staining reveals a hike in the expression of TLR6. We report here that miR-34a actively augments the effect of sepsis on lung epithelial cell apoptosis. This study suggests that miR-34a promotes TLR6 to heighten inflammation in sepsis-induced ALI/ARDS. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03700-1.
Collapse
Affiliation(s)
- Mohd Junaid Khan
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, 110025 India
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025 India
| | - Prakash Jha
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, 110007 India
| | - Arnab Nayek
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Md. Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, 15462 Kuwait City, Kuwait
| | - Ganesh Bagler
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020 India
| | - Bhupender Kumar
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, 110036 India
| | - Kalaiarasan Ponnusamy
- Biotechnology and Viral Hepatitis Division, National Centre for Disease Control, Sham Nath Marg, New Delhi, 110054 India
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences Jamia Hamdard, New Delhi, 110062 India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, 110007 India
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025 India
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110019 India
- DBC i4 Center, Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110019 India
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, 110025 India
| |
Collapse
|
7
|
Han XX, Tian YG, Liu WJ, Zhao D, Liu XF, Hu YP, Feng SX, Li JS. Metabolomic profiling combined with network analysis of serum pharmacochemistry to reveal the therapeutic mechanism of Ardisiae Japonicae Herba against acute lung injury. Front Pharmacol 2023; 14:1131479. [PMID: 37554987 PMCID: PMC10405081 DOI: 10.3389/fphar.2023.1131479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Introduction: Acute lung injury (ALI) is a common and devastating respiratory disease associated with uncontrolled inflammatory response and transepithelial neutrophil migration. In recent years, a growing number of studies have found that Ardisiae Japonicae Herba (AJH) has a favorable anti-inflammatory effect. However, its serum material basis and molecular mechanism are still unknown in ALI treatment. In this study, metabolomics and network analysis of serum pharmacochemistry were used to explore the therapeutic effect and molecular mechanism of AJH against lipopolysaccharide (LPS)-induced ALI. Methods: A total of 12 rats for serum pharmacochemistry analysis were randomly divided into the LPS group and LPS + AJH-treated group (treated with AJH extract 20 g/kg/d), which were administered LPS (2 mg/kg) by intratracheal instillation and then continuously administered for 7 days. Moreover, 36 rats for metabolomic research were divided into control, LPS, LPS + AJH-treated (5, 10, and 20 g/kg/d), and LPS + dexamethasone (Dex) (2.3 × 10-4 g/kg/d) groups. After 1 h of the seventh administration, the LPS, LPS + AJH-treated, and LPS + Dex groups were administered LPS by intratracheal instillation to induce ALI. The serum pharmacochemistry profiling was performed by UPLC-Orbitrap Fusion MS to identify serum components, which further explore the molecular mechanism of AJH against ALI by network analysis. Meanwhile, metabolomics was used to select the potential biomarkers and related metabolic pathways and to analyze the therapeutic mechanism of AJH against ALI. Results: The results showed that 71 serum components and 18 related metabolites were identified in ALI rat serum. We found that 81 overlapping targets were frequently involved in AGE-RAGE, PI3K-AKT, and JAK-STAT signaling pathways in network analysis. The LPS + AJH-treated groups exerted protective effects against ALI by reducing the infiltration of inflammatory cells and achieved anti-inflammatory efficacy by significantly regulating the interleukin (IL)-6 and IL-10 levels. Metabolomics analysis shows that the therapeutic effect of AJH on ALI involves 43 potential biomarkers and 14 metabolic pathways, especially phenylalanine, tyrosine, and tryptophan biosynthesis and linoleic acid metabolism pathways, to be influenced, which implied the potential mechanism of AJH in ALI treatment. Discussion: Our study initially elucidated the material basis and effective mechanism of AJH against ALI, which provided a solid basis for AJH application.
Collapse
Affiliation(s)
- Xiao-Xiao Han
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yan-Ge Tian
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wen-Jing Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Di Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xue-Fang Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yan-Ping Hu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Su-Xiang Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jian-Sheng Li
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Xu L, Li Y, Ma W, Sun X, Fan R, Jin Y, Chen N, Zhu X, Guo H, Zhao K, Luo J, Li C, Zheng Y, Yu D. Diesel exhaust particles exposure induces liver dysfunction: Exploring predictive potential of human circulating microRNAs signature relevant to liver injury risk. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:132060. [PMID: 37454487 DOI: 10.1016/j.jhazmat.2023.132060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Diesel exhaust particles (DEP) pollution should be taken seriously because it is an extensive environmental and occupational health concern. Exploring early effect biomarkers is crucial for monitoring and managing DEP-associated health risk assessment. Here, we found that serum levels of 67 miRNAs were dysregulated in DEP exposure group. Notably, 20 miRNAs were identified as each having a significant dose-response relationship with the internal exposure level of DEP. Further, we revealed that the DEP exposure could affect the liver function of subjects and that 7 miRNAs (including the well-known liver injury indicator, miR-122-5p) could serve as the novel epigenetic-biomarkers (epi-biomarkers) to reflect the liver-specific response to the DEP exposure. Importantly, an unprecedented prediction model using these 7 miRNAs was established for the assessment of DEP-induced liver injury risk. Finally, bioinformatic analysis indicated that the unique set of miRNA panel in serum might also contribute to the molecular mechanism of DEP exposure-induced liver damage. These results broaden our understanding of the adverse health outcomes of DEP exposure. Noteworthy, we believe this study could shed light on roles and functions of epigenetic biomarkers from environmental exposure to health outcomes by revealing the full chain of exposure-miRNAs-molecular pathways-disease evidence.
Collapse
Affiliation(s)
- Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yanting Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Xueying Sun
- School of Public Health, Qingdao University, Qingdao, China
| | - Rongrong Fan
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Ningning Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Xiaoxiao Zhu
- School of Public Health, Qingdao University, Qingdao, China
| | - Huan Guo
- School of Public Health, Huazhong University of Science and Technology, Wuhan, China
| | - Kunming Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
9
|
Li H, Hu W, Lin Y, Xu T, Zhang X, Wang C. MicroRNA-9-5p Is Involved in Lipopolysaccharide-Induced Acute Lung Injury Via the Regulation of Macrophage Polarization. Int J Toxicol 2023; 42:156-164. [PMID: 36537157 DOI: 10.1177/10915818221146446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
MicroRNA (miR)-9-5 p has been shown to affect lung cancer progression and lung fibrosis, but the efficacy of miR-9-5 p in acute lung injury (ALI) remained indefinite. The study was performed to probe the modulating mechanism of miR-9-5 p in ALI via regulating macrophage polarization. The ALI mouse model was established and blood samples of ALI patients were obtained. MiR-9-5 p levels in ALI mice and ALI patients were detected. Mouse pulmonary macrophages were extracted from bronchoalveolar lavage fluid and polarized into M1 and M2 macrophages. Intervention of miR-9-5 p expression was performed to observe the effects on M1 polarization and M2 polarization in lung macrophages, inflammatory factors in BALF, wet/dry weight ratio (W/D) in lung tissues, myeloperoxidase (MPO) activity in lung tissues, and lung tissue lesion condition. MiR-9-5 p levels were elevated in the lung tissues of ALI mice and ALI patients. MiR-9-5 p silencing could repress lung macrophages in ALI mice polarized toward the M1 phenotype and promoted the polarization toward the M2 phenotype, reduced the lung lesions, the lung water content, and the secretion levels of the pro-inflammatory factors TNF-α, IL-6, and IL-1β in BALF, increased the secretion of the anti-inflammatory factor IL-10, as well as impeded the MPO activity in the lung tissues of ALI mice. MiR-9-5 p deletion ameliorates LPS-induced inflammatory infiltration in lung tissues via inhibiting the polarization of mouse lung macrophages to the M1 phenotype and promoting the polarization to the M2 phenotype.
Collapse
Affiliation(s)
- Hao Li
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Weimi Hu
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Yueyue Lin
- Department of Gastroscope Room, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Tengxiao Xu
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Xianjing Zhang
- Department of Emergency Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Chen Wang
- Department of Respiratory and Critical Care Medicine, 573752The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| |
Collapse
|
10
|
Dobkin J, Wu L, Mangalmurti NS. The ultimate tradeoff: how red cell adaptations to malaria alter the host response during critical illness. Am J Physiol Lung Cell Mol Physiol 2023; 324:L169-L178. [PMID: 36594846 PMCID: PMC9902222 DOI: 10.1152/ajplung.00127.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
The human immune system evolved in response to pathogens. Among these pathogens, malaria has proven to be one of the deadliest and has exerted the most potent selective pressures on its target cell, the red blood cell. Red blood cells have recently gained recognition for their immunomodulatory properties, yet how red cell adaptations contribute to the host response during critical illness remains understudied. This review will discuss how adaptations that may have been advantageous for host survival might influence immune responses in modern critical illness. We will highlight the current evidence for divergent host resilience arising from the adaptations to malaria and summarize how understanding evolutionary red cell adaptations to malaria may provide insight into the heterogeneity of the host response to critical illness, perhaps driving future precision medicine approaches to syndromes affecting the critically ill such as sepsis and acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Jane Dobkin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ling Wu
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nilam S Mangalmurti
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Yang YQ, Ge P, Lv MQ, Yu PF, Liu ZG, Zhang J, Zhao WB, Han SP, Sun RF, Zhou DX. Rno_circRNA_008646 regulates formaldehyde induced lung injury through Rno-miR-224 mediated FOXI1/CFTR axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113999. [PMID: 35998475 DOI: 10.1016/j.ecoenv.2022.113999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Formaldehyde (FA) serves as a prevailing air pollutant, which has seriously threatened public health in recent years. Of all the known health effects, lung injury is one of the most severe risks. However, little is known about the circRNAs related molecular mechanism in the development of lung injury induced by FA. This study was designed to explore the potential roles of dysregulated circRNAs as well as its mechanism in FA-induced lung injury. In the present study, 24 male SD rats were exposed to formaldehyde (control, 0.5, 2.46 and 5 mg/m3) for 8 h per day for 8 weeks to induce lung injury. We used H&E staining to evaluate the histopathological changes of lung injury indifferent groups. The expression of circRNAs in lung tissue was detected by real-time PCR. Meanwhile, circRNA/miRNA/mRNA interaction networks were predicted by bioinformatics analysis. Our study revealed that formaldehyde exposure resulted in abnormal histopathological changes in lung tissues. Moreover, the expression of rno_circRNA_008646 was significantly higher in lung tissues of formaldehyde exposure rats than in control. Bioinformatics analysis showed that one potential target miRNA/mRNA for rno_circRNA_008646 was rno-miR-224/Forkhead Box I1 (FOXI1). Besides, luciferase report gene confirmed that there was targeted binding relationship between rno_circRNA_008646 and rno-miR-224, rno-miR-224 and FOXI1. Further verification experiments indicated that the expression of rno_circRNA_008646 was negatively correlated rno-miR-224, while it was positively correlated with FOXI1. JASPAR database showed transcription factor FOXI1 located in promotor of CF Transmembrane Conductance Regulator (CFTR). Both FOXI1 and CFTR were up-regulated in lung tissues after formaldehyde exposure. In conclusion, our findings suggested that formaldehyde may induce lung injury, and this may be caused by up-regulatedrno_circRNA_008646, which medicated rno-miR-224/FOXI1/CFTR axis.
Collapse
Affiliation(s)
- Yan-Qi Yang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Pan Ge
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Mo-Qi Lv
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Peng-Fei Yu
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Changlexi St. 127#, Xi'an, Shaanxi, PR China
| | - Zhi-Gang Liu
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China
| | - Jian Zhang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Wen-Bao Zhao
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Shui-Ping Han
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China
| | - Rui-Fang Sun
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China.
| | - Dang-Xia Zhou
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi PR China; Institute of Genetics and Developmental Biology, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, PR China.
| |
Collapse
|
12
|
Xu L, Ma W, Jin Y, Sun X, Chen N, Zhu X, Luo J, Li C, Zhao K, Zheng Y, Yu D. N, N-dimethylformamide exposure induced liver abnormal mitophagy by targeting miR-92a-1-5p-BNIP3L pathway in vivo and vitro. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 839:156218. [PMID: 35623527 DOI: 10.1016/j.scitotenv.2022.156218] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 06/15/2023]
Abstract
N, N-dimethylformamide (DMF) is a widely existing harmful environmental pollutant from industrial emission which can threat human health for both occupational and general populations. Epidemiological and experimental studies have indicated liver as the primary target organ of DMF. However, the molecular mechanism under DMF-induced hepatoxicity remains unclear. In the present study, we identified that DMF could induce abnormal autophagy flux in cells. We also showed that DMF-induced mitochondrial dysfunction and lethal mitophagy which further leads to autophagic cell death. Next, miRNA microarray analysis identified miR-92a-1-5p as the most down-regulated miRNA upon DMF exposure. Mechanistically, miR-92a-1-5p regulated mitochondrial function and mitophagy by targeting mitochondrial protein BNIP3L. Exogenous miR-92a-1-5p significantly attenuated DMF-induced mitochondrial dysfunction and mitophagy in vitro and in vivo. Our study highlights the mechanistic link between miRNAs and mitophagy under environmental stress, which provided a new clue for the mitochondrial epigenetics mechanism on environmental toxicant-induced hepatoxicity.
Collapse
Affiliation(s)
- Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Xueying Sun
- School of Public Health, Qingdao University, Qingdao, China
| | - Ningning Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Xiaoxiao Zhu
- School of Public Health, Qingdao University, Qingdao, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Kunming Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
13
|
Yang H, Zhang C, Zhao Z. Lipopolysaccharide-induced lung cell inflammation and apoptosis are enhanced by circ_0003420/miR-424-5p/TLR4 axis via inactivating the NF-κB signaling pathway. Transpl Immunol 2022; 74:101639. [PMID: 35667541 DOI: 10.1016/j.trim.2022.101639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) can regulate disease progression, including sepsis-induced acute lung injury (ALI). This research aimed at investigating the function of circ_0003420 in lipopolysaccharide (LPS)-treated lung cells, as well as the functional mechanism. METHODS Enzyme-linked immunosorbent assay was used for inflammation analysis. Cell viability and proliferation were examined using Cell Counting Kit-8 assay and EdU assay. Cell apoptosis was measured by flow cytometry. Western blot was used for protein detection. Reverse transcription-quantitative polymerase chain reaction assay was performed for quantification of circ_0003420, microRNA-424-5p (miR-424-5p) or toll-like receptor (TLR4). The interaction between miR-424-5p and circ_0003420 or TLR4 was conducted through dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. RESULTS Lung cell inflammation and apoptosis were promoted, but cell viability and proliferation were inhibited by LPS. Silence of circ_0003420 attenuated the LPS-mediated lung cell injury. Circ_0003420 could interact with miR-424-5p. The protective function by knockdown of si-circ_0003420 was relieved by miR-424-5p inhibition in LPS-treated cells. TLR4 served as a downstream target of miR-424-5p. Overexpression of miR-424-5p repressed inflammatory and apoptotic damages in LPS-treated lung cells via downregulating TLR4. Circ_0003420 upregulated the TLR4 level by targeting miR-424-5p and circ_0003420 regulated the NF-κB signaling pathway through the miR-424-5p/TLR4 axis. CONCLUSION These results uncovered that circ_0003420 contributed to the LPS-induced lung cell injury via activating the miR-424-5p/TLR4-related NF-κB signaling pathway. Circ_0003420 might be a therapeutic target in sepsis-induced ALI.
Collapse
Affiliation(s)
- Hailing Yang
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Chunmei Zhang
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhongyan Zhao
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
14
|
Zhan B, Shen J. Mitochondria and their potential role in acute lung injury (Review). Exp Ther Med 2022; 24:479. [PMID: 35761815 PMCID: PMC9214601 DOI: 10.3892/etm.2022.11406] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Biao Zhan
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
15
|
Qiao L, Li RX, Hu SG, Liu Y, Liu HQ, Wu HJ. microRNA-145-5p attenuates acute lung injury via targeting ETS2. Kaohsiung J Med Sci 2022; 38:565-573. [PMID: 35579106 DOI: 10.1002/kjm2.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/21/2022] [Accepted: 02/16/2022] [Indexed: 11/11/2022] Open
Abstract
The protective effect of microRNA (miR)-145-5p in acute lung injury (ALI) has been discovered previously. Thus, in this study, we attempted to further investigate the mechanism of miR-145-5p in ALI through the downstream E26 transformation-specific proto-oncogene 2 (ETS2)/transforming growth factor β1 (TGF-β1)/Smad pathway. A lipopolysaccharide (LPS)-induced ALI rat model was established. The expression of miR-145-5p in ALI rat lung tissues was up-regulated. Afterward, pathological damage in the lung tissue, the wet/dry (W/D) ratio, apoptosis, and serum inflammatory factor contents were observed. miR-145-5p, ETS2, TGF-β1, Smad2/3, and phosphorylated Smad2/3 levels were measured in rats. miR-145-5p expression was down-regulated, ETS2 expression was up-regulated, and the TGF-β1/Smad pathway was activated in LPS-exposed rats. Overexpression of miR-145-5p inactivated the TGF-β1/Smad pathway and attenuated ALI, as reflected by relieved pathological damage, a decreased W/D ratio, reduced apoptosis, and suppressed inflammatory response. In contrast, loss of miR-145-5p or elevated ETS2 levels worsened ALI and activated the TGF-β1/Smad pathway. Moreover, elevation of ETS2 diminished miR-145-5p-mediated protection against ALI. Evidently, miR-145-5p negatively regulates ETS2 expression and inactivates the TGF-β1/Smad pathway to ameliorate ALI in rats.
Collapse
Affiliation(s)
- Liang Qiao
- Department of Emergency, Henan Province Hospital of TCM (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Rong-Xia Li
- Emergency Center, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Shan-Gang Hu
- Department of Emergency, Henan Province Hospital of TCM (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Yu Liu
- Department of Emergency, Henan Province Hospital of TCM (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Hong-Qiang Liu
- Department of Emergency, Henan Province Hospital of TCM (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Hong-Jun Wu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
16
|
Xia F, Jin Z, Dong J, Chen C, Cai Y, Wang Q, Chen H. KGF-2 Protects against Lung Ischemia-Reperfusion Injury by Inhibiting Inflammation-Induced Damage to Endothelial Barrier Function. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3055039. [PMID: 35600952 PMCID: PMC9117025 DOI: 10.1155/2022/3055039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/18/2022]
Abstract
Lung ischemia-reperfusion injury (LIRI), which has a mortality rate of approximately 50%, is a popular topic in critical care research. Keratinocyte growth factor-2 (KGF-2) is secreted by mesenchymal cells, and it is effective in promoting the proliferation, migration, and differentiation of various epithelial cells. To date, however, only a few reports on KGF-2-related regulators in LIRI have been published. In the current study, an LIRI rat model is constructed, and the upregulation of the fibroblast growth factor receptor 2 (FGFR2) is observed in the LIRI rat model. In addition, LIRI induces NLRP1 inflammasome activation in vivo and in vitro, and KGF-2 inhibits LIRI-induced damage to pulmonary microvascular endothelial cells. Mechanistically, KGF-2 inhibits NLRP1 inflammasome and NF-κB activity. KGF-2 inhibition attenuates LIRI injury-induced damage to endothelial integrity. In conclusion, KGF-2 protects against LIRI by inhibiting inflammation-induced endothelial barrier damage.
Collapse
Affiliation(s)
- Fangfang Xia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Zhousheng Jin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Jiaojiao Dong
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Chaoxing Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Yaoyao Cai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Quanguang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Hongfei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| |
Collapse
|
17
|
Cai Q, Jin Y, Jia Z, Liu Z. Paraquat Induces Lung Injury via miR-199-Mediated SET in a Mouse Model. Front Pharmacol 2022; 13:856441. [PMID: 35431948 PMCID: PMC9011139 DOI: 10.3389/fphar.2022.856441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: To explore the molecular mechanism of lung injury caused by paraquat (PQ) poisoning by investigating miR-199-mediated SET.Methods: A paraquat poisoning model was established in C57BL/6 male mice via intraperitoneal injection of paraquat. The mice were transfected with miR-199 siRNA and or mimic. After 14 days of treatment, pathophysiological changes of the lung were observed and lung tissue was analyzed via Hematoxylin-Eosin staining. The levels of miR-199, SETs, surfactant protein SP-A and SP-B, and inflammatory and oxidative factors were analyzed by qPCR, Western Blot, and ELISA kits.Results: A acute lung-injury (ALI) model was established using PQ treatment and confirmed with edema of pulmonary endothelium with low electronic density of endothelial cytoplasm, presence of protein-rich fluid, and numerous erythrocytes in alveolar space, concentric figures of damaged tubular myelin, alveolar destruction, and increase in inflammatory cell numbers. Compared with the control group, miR-199 and SET levels were reduced in the PQ-treated group. miR-199 siRNA increased the SET level, inflammatory and oxidative levels, and reduced the levels of SP-A and SP-B, and miR-199 mimic reduced the SET level, inflammatory and oxidative levels, and increased the levels of SP-A and SP-B. PQ treatment reduced miR-199 level.Conclusion: Paraquat induces ALI by affecting miR-199-mediated SET.
Collapse
|
18
|
Li P, Gu L, Bian Q, Jiao D, Xu Z, Wang L. Long non-coding RNA MALAT1 enhances the protective effect of dexmedetomidine on acute lung injury by sponging miR-135a-5p to downregulate the ratio of X-box binding proteins XBP-1S/XBP-1U. Bioengineered 2021; 12:6377-6389. [PMID: 34516310 PMCID: PMC8806486 DOI: 10.1080/21655979.2021.1967579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acute lung injury (ALI) is the common and clinically severe complication. Dexmedetomidine (DEX) can protect against lipopolysaccharide (LPS)-induced ALI through anti-apoptosis, anti-inflammatory and immune regulatory actions. It is well documented that major causes of LPS-induced ALI are endoplasmic reticulum stress (ERS) and abnormally elevated CHOP. Moreover, XBP-1 can enhance CHOP expression. XBP-1S can aggravate ERS and XBP-1 U can repress ERS. By querying Starbase, miR-135a-5p interacts with XBP-1 and lncRNA MALAT1 sponges miR-135a-5p. It has been reported that MALAT1 interference markedly promoted the apoptosis of pulmonary microvascular endothelial cells in ALI rats by activating TLR4/NF-κB pathway. miR-135a-5p inhibitor remarkably alleviated LPS-induced A549 cell injury through suppressing cell apoptosis. In the present work, LPS was dripped into the nasal cavity of SD rats to establish the rat model of ALI and LPS was also applied to stimulate BEAS-2B cells to imitate ALI in vitro. Then, the pathology, lung function indexes, levels of inflammatory factors, apoptosis of lung tissues in SD rats and apoptotic level of BEAS-2B cells were measured, so as to confirm whether upregulation of lncRNA MALAT1 was able to suppress ERS, thus enhancing the protective effect of DEX against ALI. Herein, overexpression of lncRNA MALAT1 strengthened the remission effects of DEX on LPS-triggered ALI, severe pulmonary edema, inflammatory response and cell apoptosis of lung tissues in SD rats and reinforced the anti-apoptosis effect of DEX on LPS-stimulated BEAS-2B cells. Mechanically, lncRNA MALAT1 enhanced the protective effect of DEX against ALI by downregulating the ratio of XBP-1S/XBP-1U to repress ERS.
Collapse
Affiliation(s)
- Pengyi Li
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lianbing Gu
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingming Bian
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dian Jiao
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zeping Xu
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lijun Wang
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
A novel miRNA-762/NFIX pathway modulates LPS-induced acute lung injury. Int Immunopharmacol 2021; 100:108066. [PMID: 34492536 DOI: 10.1016/j.intimp.2021.108066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
Severe acute lung injury (ALI) cause significant morbidity and mortality worldwide. MicroRNAs (miRNAs) are possible biomarkers and therapeutic targets for ALI. We aimed to explore the role of miR-762, a known oncogenic factor, in the pathogenesis of ALI. Levels of miR-762 in lung tissues of LPS-treated ALI mice and blood cells of patients with lung injury were measured. Injury of human lung epithelial cell line A549 was induced by LPS stimulation. A downstream target of miR-762, NFIX, was predicted using online tools. Their interactions were validated by luciferase reporter assay. Effects of targeted regulation of the miR-762/NFIX axis on cell proliferation, apoptosis, and inflammatory responses were tested in vitro in A549 cells in vivo with an ALI mouse model. We found that upregulation of miR-762 expression and downregulation of NFIX expression were associated with lung injury. Either miR-762 inhibition or NFIX overexpression in A549 lung cells significantly attenuated LPS-mediated impairment of cell proliferation and viability. Notably, increasing expressions of miR-762 inhibitor or NFIX in vivo via airway lentivirus infection alleviated the LPS-induced ALI in mice. Further, targeted downregulation of miR-762 expression or upregulation of NFIX expression in A549 cells markedly down-regulates NF-κB/IRF3 activation, and substantially reduces the production of inflammatory factors, including TNF-α, IL-6, and IL-8. This study reveals a novel role for the miR-762/NFIX pathway in ALI pathogenesis and sheds new light on targeting this pathway for diagnosis, prevention, and therapy.
Collapse
|
20
|
Ji J, Ye W, Sun G. LncRNA OIP5-AS1 knockdown or miR-223 overexpression can alleviate LPS-induced ALI/ARDS by interfering with miR-223/NLRP3-mediated pyroptosis. J Gene Med 2021; 24:e3385. [PMID: 34346534 DOI: 10.1002/jgm.3385] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening diseases and endothelial barrier injury is an important contributor to the pathogenesis of ALI/ARDS. LncRNA has been proved to participate in the progression of ALI/ARDS. Our study aimed to investigate the function of lncRNA OIP5-AS1 in LPS-induced ALI/ARDS. METHODS OIP5-AS1 and miR-223 levels were detected by PCR in the serum of ALI/ARDS patients or healthy donors. MTT assay were performed to detect the proliferation of HPMECs. Flow cytometry were performed to detect the apoptosis of HPMECs. The protein levels of NLRP3, ASC, GSDMD-N, caspase-1 were measured by western blot to detect the pyroptosis of HPMECs. IL-1β, IL-6, IL-18 and IL-10 was detected by ELISA to measure the inflammatory response of HPMECs. And production of ROS, SOD and MDA was measured to determine the oxidative stress of HPMECs. Targets of OIP5-AS1 and miR-223 were predicted by StarBase and confirmed by dual-luciferase reporter assay. RESULTS We found that OIP5-AS1 was upregulated, while miR-223 was downregulated in the serum of ALI/ARDS patients and LPS-treated HPMECs. Functionally, knockdown of OIP5-AS1 induced proliferation and inhibited apoptosis, pyroptosis, inflammatory response and oxidative stress of LPS-treated HPMECs. Interestingly, miR-223 was a target of OIP5-AS1 and miR-223 inhibition abolished the effects of si-OIP5-AS1 on LPS-induced HPMECs. More importantly, miR-223 directly targeted NLRP3, miR-223 overexpression also promoted proliferation and inhibited apoptosis, pyroptosis, inflammatory response and oxidative stress of LPS-treated HPMECs and which was abolished by NLRP3 overexpression. Finally, we found that OIP5-AS1 knockdown and miR-223 overexpression could both alleviate LPS-induced ALI/ARDS in vivo. CONCLUSION Together, we find that LncRNA OIP5-AS1 aggravates LPS-induced ALI/ARDS via miR-223/NLRP3 axis and provides new targets for ALI/ARDS therapy.
Collapse
Affiliation(s)
- Juanjuan Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Ye
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Zaki A, Ali MS, Hadda V, Ali SM, Chopra A, Fatma T. Long non-coding RNA (lncRNA): A potential therapeutic target in acute lung injury. Genes Dis 2021; 9:1258-1268. [PMID: 35873025 PMCID: PMC9293716 DOI: 10.1016/j.gendis.2021.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/26/2022] Open
Abstract
Acute Lung Injury (ALI) and its severe form Acute Respiratory Distress Syndrome (ARDS) are the major cause of ICU death worldwide. ALI/ARDS is characterized by severe hypoxemia and inflammation that leads to poor lung compliance. Despite many advances in understanding and management, ALI/ARDS is still causing significant morbidity and mortality. Long non-coding RNA (lncRNA) is a fast-growing topic in lung inflammation and injury. lncRNA is a class of non-coding RNA having a length of more than 200 nucleotides. It has been a center of research for understanding the pathophysiology of various diseases in the past few years. Multiple studies have shown that lncRNAs are abundant in acute lung injury/injuries in mouse models and cell lines. By targeting these long non-coding RNAs, many investigators have demonstrated the alleviation of ALI in various mouse models. Therefore, lncRNAs show great promise as a therapeutic target in ALI. This review provides the current state of knowledge about the relationship between lncRNAs in various biological processes in acute lung injury and its use as a potential therapeutic target.
Collapse
|
22
|
Jiang ZF, Shao Y, Zhang L, Shen J. Lung-derived exosomes regulate the function of mesenchymal stem cells and alleviate phosgene-induced lung injury via miR-34c-3p. J Biochem Mol Toxicol 2021; 35:e22851. [PMID: 34331784 DOI: 10.1002/jbt.22851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 12/18/2022]
Abstract
Phosgene may induce acute lung injury (ALI) when a person is exposed to it. Mesenchymal stem cells (MSCs) were affirmed to have therapeutic effects on phosgene-induced ALI. In a previous study, ALI exosomes have been confirmed to promote the proliferation and migration of MSCs. However, the mechanism of this phenomenon is still unclear. MicroRNAs (miRNAs) are essential in the physiological process of cells. In this study, lung-derived exosomes were isolated from phosgene-exposed and normal rats, respectively, through ultracentrifugation and cultured MSCs with these exosomes. We found that rno-miR-34c-3p was downregulated in MSCs cocultured with ALI exosomes. MiR-34c-3p inhibitor promoted the proliferation and migration of MSCs. Moreover, the dual-luciferase reporter assay demonstrated that miR-34c-3p regulated Janus kinase 1 (JAK1) expression. The miR-34c-3p inhibitor also significantly activated the JAK1/signal transducer and activator of transcription 3 (STAT3) signaling pathway. In conclusion, ALI exosomes decrease the miR-34c-3p expression levels, influencing MSCs via the JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhi-Feng Jiang
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yiru Shao
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Intensive Care Unit, Medical Research Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Martucci G, Arcadipane A, Tuzzolino F, Occhipinti G, Panarello G, Carcione C, Bertani A, Conaldi PG, Miceli V. Circulating miRNAs as Promising Biomarkers to Evaluate ECMO Treatment Responses in ARDS Patients. MEMBRANES 2021; 11:membranes11080551. [PMID: 34436314 PMCID: PMC8398026 DOI: 10.3390/membranes11080551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
The use of extracorporeal membrane oxygenation (ECMO) for acute respiratory distress syndrome (ARDS) has increased in the last decade. However, mortality remains high, and the complexity of ECMO requires individualized treatment. There are some biomarkers to monitor progression and predict clinical outcomes of ARDS. This project aims to advance the management of ARDS patients treated with ECMO by exploring miRNA expression in whole blood. The analysis was conducted on two groups with different length of ECMO: Group A (longer runs) and group B (shorter runs). We analyzed miRNAs before ECMO cannulation, and at 7 and 14 days of ECMO support. Our results showed that in the group B patients, 11 deregulated miRNAs were identified, and showed an opposite trend of expression compared to the group A patients. In silico analysis revealed that these 11 miRNAs were related to processes involved in the pathogenesis and evolution of ARDS. This scenario could represent homeostatic mechanisms by which, in ECMO responsive patients, pathways activated during ARDS progression are switched-off. Circulating miRNAs could represent promising biomarkers to monitor the evolution of ARDS under ECMO support. Further studies may shed light on this topic to improve a personalized approach in such a complex setting of patients.
Collapse
Affiliation(s)
- Gennaro Martucci
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90127 Palermo, Italy; (G.M.); (A.A.); (G.O.); (G.P.)
| | - Antonio Arcadipane
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90127 Palermo, Italy; (G.M.); (A.A.); (G.O.); (G.P.)
| | - Fabio Tuzzolino
- Research Department, IRCCS-ISMETT, 90127 Palermo, Italy; (F.T.); (P.G.C.)
| | - Giovanna Occhipinti
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90127 Palermo, Italy; (G.M.); (A.A.); (G.O.); (G.P.)
| | - Giovanna Panarello
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90127 Palermo, Italy; (G.M.); (A.A.); (G.O.); (G.P.)
| | | | - Alessandro Bertani
- Division of Thoracic Surgery and Lung Transplantation, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT, 90127 Palermo, Italy;
| | | | - Vitale Miceli
- Research Department, IRCCS-ISMETT, 90127 Palermo, Italy; (F.T.); (P.G.C.)
- Correspondence: ; Tel.: +39-091-219-2430
| |
Collapse
|
24
|
Martucci G, Arcadipane A, Tuzzolino F, Occhipinti G, Panarello G, Carcione C, Bonicolini E, Vitiello C, Lorusso R, Conaldi PG, Miceli V. Identification of a Circulating miRNA Signature to Stratify Acute Respiratory Distress Syndrome Patients. J Pers Med 2020; 11:jpm11010015. [PMID: 33375484 PMCID: PMC7824233 DOI: 10.3390/jpm11010015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/07/2020] [Accepted: 12/25/2020] [Indexed: 02/07/2023] Open
Abstract
There is a need to improve acute respiratory distress syndrome (ARDS) diagnosis and management, particularly with extracorporeal membrane oxygenation (ECMO), and different biomarkers have been tested to implement a precision-focused approach. We included ARDS patients on veno-venous (V-V) ECMO in a prospective observational pilot study. Blood samples were obtained before cannulation, and screened for the expression of 754 circulating microRNA (miRNAs) using high-throughput qPCR and hierarchical cluster analysis. The miRNet database was used to predict target genes of deregulated miRNAs, and the DIANA tool was used to identify significant enrichment pathways. A hierarchical cluster of 229 miRNAs (identified after quality control screening) produced a clear separation of 11 patients into two groups: considering the baseline SAPS II, SOFA, and RESP score cluster A (n = 6) showed higher severity compared to cluster B (n = 5); p values < 0.05. After analysis of differentially expressed miRNAs between the two clusters, 95 deregulated miRNAs were identified, and reduced to 13 by in silico analysis. These miRNAs target genes implicated in tissue remodeling, immune system, and blood coagulation pathways. The blood levels of 13 miRNAs are altered in severe ARDS. Further investigations will have to match miRNA results with inflammatory biomarkers and clinical data.
Collapse
Affiliation(s)
- Gennaro Martucci
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Antonio Arcadipane
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
- Correspondence: ; Tel.: +39-091-2192332
| | - Fabio Tuzzolino
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| | - Giovanna Occhipinti
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Giovanna Panarello
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | | | - Eleonora Bonicolini
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Chiara Vitiello
- Anesthesia and Intensive Care Department, IRCCS-ISMETT, 90133 Palermo, Italy; (G.M.); (G.O.); (G.P.); (E.B.); (C.V.)
| | - Roberto Lorusso
- Cardio-Thoracic Surgery Department Heart and Vascular Centre, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
- Cardiovascular Research Institute Maastricht (CARIM), 6229HX Maastricht, The Netherlands
| | - Pier Giulio Conaldi
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| | - Vitale Miceli
- Research Department, IRCCS-ISMETT, 90133 Palermo, Italy; (F.T.); (P.G.C.); (V.M.)
| |
Collapse
|