1
|
Feng C, Li Q, Lv M, Ji Q, Ye H, Jiang S, Shao M, Shao Q, Cao L. A pan-cancer analysis of small nucleolar RNA host gene 14 (SNHG14) in human tumors. Gene 2025; 951:149410. [PMID: 40074048 DOI: 10.1016/j.gene.2025.149410] [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: 05/18/2024] [Revised: 12/29/2024] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Long non-coding RNAs (lncRNAs) are associated with tumorigenesis and progression. One of these, short nucleolar RNA host gene 14 (SNHG14), has exhibited significant prognostic value due to its aberrant expression across various tumor types. This study investigates the expression patterns, survival outcomes, and tumor stages associated with SNHG14 across various cancers, employing data from the Genotype-Tissue Expression and The Cancer Genome Atlas databases. The Cancer Genome Atlas database showed SNHG14 overexpression in five tumor types and downregulation in 15 tumor types compared tonormal tissues. In particular, patients with increased SNHG14 expression had reduced overall survival with mesothelioma and stomach adenocarcinoma. A comprehensive literature review was conducted to explore SNHG14's upstream regulators and downstream target genes, shedding light on its role in tumorigenesis. The review underscores that SNHG14 is frequently overexpressed in numerous cancers and predominantly functions as an oncogene. SNHG14 exerts its effects by regulating various microRNAs, which subsequently modulate the expression of target genes and influence critical signaling pathways involved in tumor development and progression. Furthermore, biotin-DNA pulldown coupled with mass spectrometry identified several transcription factors, including c-Myc and CEBPB, as key regulators of SNHG14 transcription. These findings highlight the intricate transcriptional regulation of SNHG14 and its potential involvement in cancer-related processes. In this study, we systematically elucidated the upstream transcriptional regulators and downstream signaling pathways of SNHG14, providing novel insights into its critical role in cancer biology. This comprehensive research highlights SNHG14 as an effective biomarker for prognosis and a target for treating cancer.
Collapse
Affiliation(s)
- Chencheng Feng
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Qianru Li
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Mengyao Lv
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Qiang Ji
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Haoming Ye
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Su Jiang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Min Shao
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China.
| | - Qian Shao
- Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Limian Cao
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China.
| |
Collapse
|
2
|
Qi H, Ying G, Ling W, Jia H, Zhou X, Lin X. The role of lncRNAs in sepsis-induced acute lung injury: Molecular mechanisms and therapeutic potential. Arch Biochem Biophys 2025; 768:110407. [PMID: 40180295 DOI: 10.1016/j.abb.2025.110407] [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: 03/02/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Sepsis, a life-threatening syndrome, results from a dysregulated immune and hemostatic response, contributing to acute lung injury (ALI) and its progression into acute respiratory distress syndrome (ARDS). The development of septic ALI is complex, involving excessive inflammatory mediator production that damages endothelial and epithelial cells, leading to vascular leakage, edema, and vasodilation-key factors in ALI pathogenesis. Long noncoding RNAs (lncRNAs), over 200 nucleotides in length, play critical roles in various biological processes, including sepsis regulation. They exhibit both promotive and inhibitory effects, influencing sepsis progression and resolution. Despite their significance, comprehensive reviews detailing lncRNA involvement in sepsis-induced ALI remain limited. This review aims to address this gap by summarizing the diverse functions of lncRNAs in septic ALI, emphasizing their potential in diagnosis and treatment. Furthermore, we will explore the molecular mechanisms underlying lncRNA involvement, particularly their miRNA-dependent regulatory pathways. Understanding these interactions may provide novel insights into therapeutic strategies for sepsis-induced ALI.
Collapse
Affiliation(s)
- Huijuan Qi
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China.
| | - Gu Ying
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Wang Ling
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Honggang Jia
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Xinxiu Zhou
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| | - Xinyu Lin
- Department of Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan City, 250001, Shandong Province, China
| |
Collapse
|
3
|
Liao X, Jiang Z, Fu Z, Liu M, Guo G. LncRNA SNHG14 Drives NLRP3 Inflammasome Activation in Diabetic Foot Ulcers: Mechanistic Insights and Diagnostic Implications. INT J LOW EXTR WOUND 2025:15347346251339713. [PMID: 40329710 DOI: 10.1177/15347346251339713] [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: 05/08/2025]
Abstract
BackgroundDiabetic foot (DF), a severe complication of diabetes mellitus (DM), poses significant challenges in early diagnosis and mechanistic understanding. This study investigates the expression patterns and clinical relevance of long non-coding RNA SNHG14 (lncRNA SNHG14) and the NLRP3 inflammasome in DF pathogenesis.MethodsA total of 176 DM patients (88 DF cases vs 88 Non-DF controls) admitted between September 2022 and February 2024 were enrolled. Serum SNHG14 and NLRP3 levels were quantified via qRT-PCR, while DF severity was categorized using the Wagner grading system. Pearson's correlation assessed SNHG14-NLRP3 interactions, Spearman's rank correlation evaluated their associations with Wagner grades, logistic regression identified independent risk factors, and ROC analysis determined diagnostic efficacy.ResultsDF patients exhibited significantly prolonged diabetes duration, elevated HbA1c%, FPG, and upregulated SNHG14/NLRP3 expression compared to Non-DF controls (P < 0.05). A robust positive correlation was observed between SNHG14 and NLRP3 (r = 0.7006, P < 0.0001). Multivariate logistic regression revealed diabetes duration (OR = 7.423, P < 0.0001), HbA1c (OR = 19.478, P = 0.002), SNHG14 overexpression (OR = 5941.653, P < 0.001), and NLRP3 upregulation (OR = 529.864, P = 0.036) as independent DF risk factors. Both SNHG14 (r = 0.5953) and NLRP3 (r = 0.5554) positively correlated with Wagner grades (P < 0.0001). ROC analysis demonstrated high diagnostic accuracy for SNHG14 (AUC = 0.8688) and NLRP3 (AUC = 0.8074), with combined detection further improving performance (AUC = 0.8773, sensitivity = 77.27%, specificity = 93.18%).ConclusionOverexpression of SNHG14 and NLRP3 is intricately linked to DF progression, metabolic dysregulation, and ulcer severity. Their combined use synergistically enhances diagnostic precision, highlighting transformative potential in DF management.
Collapse
Affiliation(s)
- Xincheng Liao
- Department of Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhengying Jiang
- Department of Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhonghua Fu
- Department of Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Mingzhuo Liu
- Department of Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Guanghua Guo
- Department of Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
4
|
Musavi M, Haftcheshmeh SM, Fazel H, Momtazi-Borojeni AA. Predicting microRNAs and their Target Genes Involved in Sepsis Pathogenesis by using Bioinformatics Methods. Curr Pharm Des 2025; 31:1067-1077. [PMID: 39754769 DOI: 10.2174/0113816128304401241031094647] [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: 05/31/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 01/06/2025]
Abstract
INTRODUCTION Sepsis, like neutropenic sepsis, is a medical condition in which our body overreacts to infectious agents. It is associated with damage to normal tissues and organs by the immune system, which leads to the spread of inflammation throughout our body. Of note, microRNAs (miRNAs) have been found to have a critical role in the sepsis progression. Such miRNAs are registered in the miRNA databases, such as Gene Expression Omnibus (GEO), with a specific identifier and unique characteristics. There is also computational software, such as TargetScan, that are broadly employed for the analysis of miRNAs, including their identification, target prediction, and functional analysis. METHODS The current In-silico study aimed to predict miRNAs involved in sepsis progression. To this end, the GEO database was employed to find the sepsis-related genome profile. Afterward, down-regulated genes were selected for further bioinformatics analysis with the assumption that their decreased expression is associated with an increased sepsis progression. The miRNAs complementary to the selected genes were then predicted using TargetScan software. Based on the current In-silico analysis, seven miRNAs, including hsa-miR-325-3p, hsa-miR-146a-3p, hsa-miR-126-5p, hsa-miR-22-3p, hsa-miR-223-3p, hsa-miR-145-5p, and has-miR-181 family, were predicted to participate in sepsis pathogenesis. Among the predicted miRNAs, hsa-miR-325-3p has not been previously predicted or validated to be involved in septic conditions. RESULTS Our prediction results showed that hsa-miR-325-3p may target genes implicating in both anti-(ETFB gene) and pro-inflammatory (TCEA1 and PTPN1 genes) responses, suggesting it is an immune hemostasis regulator during sepsis inflammation. Although the role of other predicted miRNAs has been already validated in the sepsis pathogenesis, the current study predicted new targets of these miRNAs, which have not been reported by previous in-silico or experimental studies on sepsis and other pathogenic conditions. Notably, other miRNAs, including hsa-miR-146a-3p, hsa-miR-126-5p, hsa-miR-22-3p, hsa-miR-223-3p, and hsa-miR-145-5p were predicted to target genes participating in inflammatory responses, including BLOC1S1, POLR2G, PTPN1, TCEA1, and CCT3. CONCLUSION In conclusion, the results of the present study can provide promising targets as therapeutic and diagnostic tools to treat and manage inflammation sepsis, such as neutropenic sepsis. However, these findings should be further evaluated in experimental studies to find their exact effects and underlying mechanisms.
Collapse
Affiliation(s)
- Maryam Musavi
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | | | - Hadi Fazel
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amir Abbas Momtazi-Borojeni
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
5
|
Xu H, Dou L, Wang Y, Li Y, Liu D, Gao H. Whole transcriptome sequencing identifies key lncRNAs, circRNAs and miRNAs in sepsis-associated acute lung injury. Exp Lung Res 2024; 50:242-258. [PMID: 39587404 DOI: 10.1080/01902148.2024.2429184] [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: 01/23/2024] [Revised: 09/22/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
Purpose: In this study, we identified differentially expressed genes (DEGs) and signaling pathways to gain insight into the pathogenesis of acute lung injury (ALI). Methods: C57BL/6 mice were intravenously injected with lipopolysaccharide (LPS) to establish a sepsis-induced ALI model. Hematoxylin-eosin (H&E) and enzyme-linked immunosorbent assays (ELISAs) were used to evaluate the model. Whole transcriptome sequencing was performed to identify the expression changes in lncRNAs, circRNAs, miRNAs and mRNAs in lung tissues. The crucial RNAs and the biological function of the target genes were confirmed and annotated based on bioinformatics analysis. Real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was employed to verify the expression levels of key lncRNAs, circRNAs, miRNAs and mRNAs in the lung tissues and human bronchoalveolar lavage (BALF). Results: A total of 3304 (1632 upregulated and 1672 downregulated) differentially expressed mRNAs, 794 (397 up and 397 down) differentially expressed lncRNAs, 89 (58 up and 31 down) differentially expressed circRNAs, and 14 (11 up and 3 down) differentially expressed miRNAs were identified between the control and LPS lung tissues. The lncRNA ceRNA subnetwork and circRNA ceRNA subnetwork were constructed based on the observed interaction and co-expression among the differentially expressed RNAs. An analysis of the protein-protein interaction (PPI) network and hub genes revealed crucial mRNAs for circRNA-Tcf20. The lncRNA-Snhg12, Edn1, Stat1, miR-212-3p and miR-223-3p were upregulated in sepsis ARDS patients. CircRNA-Tcf20, Col1a1, Col1a2 and Flt3 were significantly downregulated in sepsis ARDS patients. The biological function analysis indicated that these genes were enriched in the TNF signaling pathway, Necroptosis signaling pathway and the PI3K-Akt signaling pathway. Conclusions: Our findings suggest that circRNA-Tcf20, miR-212-3p, miR-223-3p, Col1a1, Col1a2 and Flt3 may be new regulatory factors that participate in the pathogenesis of sepsis-related acute lung injury. CircRNA-Tcf20, lncRNA-Snhg12 and all the other RNAs may be potential biomarkers for septic ALI/ARDS.
Collapse
Affiliation(s)
- Hua Xu
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin, China
| | - Lin Dou
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin, China
| | - Yongqiang Wang
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin, China
| | - Yin Li
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin, China
| | - Hongmei Gao
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin, China
| |
Collapse
|
6
|
Tian L, Jin J, Lu Q, Zhang H, Tian S, Lai F, Liu C, Liang Y, Lu Y, Zhao Y, Yao S, Ren W. Bidirectional modulation of extracellular vesicle-autophagy axis in acute lung injury: Molecular mechanisms and therapeutic implications. Biomed Pharmacother 2024; 180:117566. [PMID: 39423751 DOI: 10.1016/j.biopha.2024.117566] [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: 07/24/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Acute lung injury (ALI), a multifactorial pathological condition, manifests through heightened inflammatory responses, compromised lung epithelial-endothelial barrier function, and oxidative stress, potentially culminating in respiratory failure and mortality. This study explores the intricate interplay between two crucial cellular mechanisms-extracellular vesicles (EVs) and autophagy-in the context of ALI pathogenesis and potential therapeutic interventions.EVs, bioactive membrane-bound structures secreted by cells, serve as versatile carriers of molecular cargo, facilitating intercellular communication and significantly influencing disease progression. Concurrently, autophagy, an essential intracellular degradation process, maintains cellular homeostasis and has emerged as a promising therapeutic target in ALI and acute respiratory distress syndrome.Our research unveils a fascinating "EV-Autophagy dual-drive pathway," characterized by reciprocal regulation between these two processes. EVs modulate autophagy activation and inhibition, while autophagy influences EV production, creating a dynamic feedback loop. This study posits that precise manipulation of this pathway could revolutionize ALI treatment strategies.By elucidating the mechanisms underlying this cellular crosstalk, we open new avenues for targeted therapies. The potential for engineered EVs to fine-tune autophagy in ALI treatment is explored, alongside innovative concepts such as EV-based vaccines for ALI prevention and management. This research not only deepens our understanding of ALI pathophysiology but also paves the way for novel, more effective therapeutic approaches in critical care medicine.
Collapse
Affiliation(s)
- Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Clinical Medical Center of Tissue Egineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Jin
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Qianying Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Huajing Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Sijia Tian
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Chuanchuan Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yangfan Liang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yujia Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yanmei Zhao
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China.
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Clinical Medical Center of Tissue Egineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
7
|
Qin J, Chen Y, Zhao X, Yu J. circCUL3 drives malignant progression of cervical cancer by activating autophagy through sponge miR-223-3p upregulation of ATG7. Gene 2024; 925:148572. [PMID: 38759738 DOI: 10.1016/j.gene.2024.148572] [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: 01/23/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Circular RNA (circRNA) has emerged as a pivotal regulatory factor in cancer biology, yet its exact role in cervical cancer remains incompletely understood. In this study, we investigated the functional role of circCUL3 in cervical cancer and explored its potential as a therapeutic target. Functional gain and loss experiments were conducted in Hela and Siha cell lines to elucidate the biological functions of circCUL3 in cervical cancer. The results revealed that circCUL3 overexpression significantly enhanced cell viability, migration, and invasion while suppressing apoptosis, while circCUL3 knockout displayed the opposite effects. Mechanistically, we identified hsa-miR-223-3p as a target of circCUL3, with its expression being negatively regulated by circCUL3. Furthermore, we discovered that circCUL3 could sequester miR-223-3p, leading to the upregulation of ATG7 expression, and this was linked to the regulation of autophagy in cervical cancer cells. In vivo validation using a xenograft mouse model further supported our in vitro findings. Notably, we found that chloroquine (CQ), an autophagy inhibitor, restored miR-223-3p expression and counteracted the oncogenic effect of circCUL3 overexpression. In conclusion, circCUL3 potentially contributes to the malignant progression of cervical cancer by acting as a sponge for miR-223-3p, resulting in the upregulation of ATG7 and the activation of autophagy.
Collapse
Affiliation(s)
- Jiahui Qin
- Department of Gynecology, Hangzhou Third People's Hospital, Hangzhou, Zhejiang, China.
| | - Yan Chen
- Department of Gynecology, Hangzhou Third People's Hospital, Hangzhou, Zhejiang, China
| | - Xia Zhao
- Department of Gynecology, Hangzhou Third People's Hospital, Hangzhou, Zhejiang, China
| | - Jingmin Yu
- Department of Gynecology, Hangzhou Third People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Zhu X, Meng L, Xu L, Hua Y, Feng J. Novel Therapeutic Target for ALI/ARDS: Forkhead Box Transcription Factors. Lung 2024; 202:513-522. [PMID: 39259274 DOI: 10.1007/s00408-024-00740-z] [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: 05/29/2024] [Accepted: 08/17/2024] [Indexed: 09/12/2024]
Abstract
ALI/ARDS can be a pulmonary manifestation of a systemic inflammatory response or a result of overexpression of the body's normal inflammatory response involving various effector cells, cytokines, and inflammatory mediators, which regulate the body's immune response through different signalling pathways. Forkhead box transcription factors are evolutionarily conserved transcription factors that play a crucial role in various cellular processes, such as cell cycle progression, proliferation, differentiation, migration, metabolism, and DNA damage response. Transcription factors control protein synthesis by regulating gene transcription levels, resulting in diverse biological outcomes. The Fox family plays a role in activating or inhibiting the expression of various molecules related to ALI/ARDS through phosphorylation, acetylation/deacetylation, and control of multiple signalling pathways. An in-depth analysis of the integrated Fox family's role in ALI/ARDS can aid in the development of potential diagnostic and therapeutic targets for the condition.
Collapse
Affiliation(s)
- Xi Zhu
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Leyuan Meng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Liqin Xu
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Yun Hua
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Jian Feng
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Zuo Y, Dang R, Peng H, Hu P, Yang Y. LL37-mtDNA regulates viability, apoptosis, inflammation, and autophagy in lipopolysaccharide-treated RLE-6TN cells by targeting Hsp90aa1. Open Life Sci 2024; 19:20220943. [PMID: 39220589 PMCID: PMC11365468 DOI: 10.1515/biol-2022-0943] [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: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis-induced acute lung injury is associated with lung epithelial cell injury. This study analyzed the role of the antimicrobial peptide LL37 with mitochondrial DNA (LL37-mtDNA) and its potential mechanism of action in lipopolysaccharide (LPS)-treated rat type II alveolar epithelial cells (RLE-6TN cells). RLE-6TN cells were treated with LPS alone or with LL37-mtDNA, followed by transcriptome sequencing. Differentially expressed and pivotal genes were screened using bioinformatics tools. The effects of LL37-mtDNA on cell viability, inflammation, apoptosis, reactive oxygen species (ROS) production, and autophagy-related hallmark expression were evaluated in LPS-treated RLE-6TN cells. Additionally, the effects of Hsp90aa1 silencing following LL37-mtDNA treatment were investigated in vitro. LL37-mtDNA further suppressed cell viability, augmented apoptosis, promoted the release of inflammatory cytokines, increased ROS production, and elevated LC3B expression in LPS-treated RLE-6TN cells. Using transcriptome sequencing and bioinformatics, ten candidate genes were identified, of which three core genes were verified to be upregulated in the LPS + LL37-mtDNA group. Additionally, Hsp90aa1 downregulation attenuated the effects of LL37-mtDNA on LPS-treated RLE-6TN cells. Hsp90aa1 silencing possibly acted as a crucial target to counteract the effects of LL37-mtDNA on viability, apoptosis, inflammation, and autophagy activation in LPS-treated RLE-6TN cells.
Collapse
Affiliation(s)
- Yunlong Zuo
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Run Dang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Hongyan Peng
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Peidan Hu
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Yiyu Yang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
10
|
Shen Y, He Y, Pan Y, Liu L, Liu Y, Jia J. Role and mechanisms of autophagy, ferroptosis, and pyroptosis in sepsis-induced acute lung injury. Front Pharmacol 2024; 15:1415145. [PMID: 39161900 PMCID: PMC11330786 DOI: 10.3389/fphar.2024.1415145] [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: 04/10/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Sepsis-induced acute lung injury (ALI) is a major cause of death among patients with sepsis in intensive care units. By analyzing a model of sepsis-induced ALI using lipopolysaccharide (LPS) and cecal ligation and puncture (CLP), treatment methods and strategies to protect against ALI were discussed, which could provide an experimental basis for the clinical treatment of sepsis-induced ALI. Recent studies have found that an imbalance in autophagy, ferroptosis, and pyroptosis is a key mechanism that triggers sepsis-induced ALI, and regulating these death mechanisms can improve lung injuries caused by LPS or CLP. This article summarized and reviewed the mechanisms and regulatory networks of autophagy, ferroptosis, and pyroptosis and their important roles in the process of LPS/CLP-induced ALI in sepsis, discusses the possible targeted drugs of the above mechanisms and their effects, describes their dilemma and prospects, and provides new perspectives for the future treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Yao Shen
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yingying He
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Ying Pan
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yulin Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Xi Q, Liu L, Zhao Q, Zhu S. KLF13 Attenuates Lipopolysaccharide-Induced Alveolar Epithelial Cell Damage by Regulating Mitochondrial Quality Control via Binding PGC-1α. J Interferon Cytokine Res 2024. [PMID: 38949897 DOI: 10.1089/jir.2023.0234] [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: 07/03/2024] Open
Abstract
Sepsis is a clinically life-threatening syndrome, and acute lung injury is the earliest and most serious complication. We aimed to assess the role of kruppel-like factor 13 (KLF13) in lipopolysaccharide (LPS)-induced human alveolar type II epithelial cell damage and to reveal the possible mechanism related to peroxisome proliferator-activated receptor-γ co-activator 1-α (PGC-1α). In LPS-treated A549 cells with or without KLF13 overexpression or PGC-1α knockdown, cell viability was measured by a cell counting kit-8 assay. Enzyme-linked immunosorbent assay kits detected the levels of inflammatory factors, and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining measured cell apoptosis. Besides, mitochondrial reactive oxygen species (MitoSOX) and mitochondrial membrane potential were detected using MitoSOX red- and JC-1 staining. Expression of proteins related to mitochondrial quality control (MQC) was evaluated by western blot. Co-immunoprecipitation (Co-IP) assay was used to analyze the interaction between KLF13 and PGC-1α. Results indicated that KLF13 was highly expressed in LPS-treated A549 cells. KLF13 upregulation elevated the viability and reduced the levels of inflammatory factors in A549 cells exposed to LPS. Moreover, KLF13 gain-of-function inhibited LPS-induced apoptosis of A549 cells, accompanied by upregulated BCL2 expression and downregulated Bax and cleaved caspase3 expression. Furthermore, MQC was improved by KLF13 overexpression, as evidenced by decreased MitoSOX, JC-1 monomers and increased JC-1 aggregates, coupled with the changes of proteins related to MQC. In addition, Co-IP assay confirmed the interaction between KLF13 and PGC-1α. PGC-1α deficiency restored the impacts of KLF13 upregulation on the inflammation, apoptosis, and MQC in LPS-treated A549 cells. In conclusion, KLF13 attenuated LPS-induced alveolar epithelial cell inflammation and apoptosis by regulating MQC via binding PGC-1α.
Collapse
Affiliation(s)
- Qiong Xi
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lin Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qin Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zhu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Yang N, Yan N, Bai Z, Du S, Zhang J, Zhang L, Zhang Z. FTO attenuates LPS-induced acute kidney injury by inhibiting autophagy via regulating SNHG14/miR-373-3p/ATG7 axis. Int Immunopharmacol 2024; 128:111483. [PMID: 38215656 DOI: 10.1016/j.intimp.2023.111483] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/15/2023] [Accepted: 12/31/2023] [Indexed: 01/14/2024]
Abstract
N6-methyladenosine (m6A) is a master driver of RNA function and implicates in the pathogenesis of renal injury. LncRNA SNHG14 is highly expressed in sepsis patients with acute kidney injury (AKI) and aggravates kidney cell dysfunction. This study aimed to explore whether demethylase FTO affect m6A methylation of SNHG14 in AKI injury and its underlying mechanism. The expression level of FTO was obviously downregulated in sepsis-associated AKI patients compared with normal controls. Mechanistically, FTO overexpression impeded SNHG14 expression by decreasing the stability of SNHG14 in an m6A-dependent manner in LPS-induced HK-2 cells. Additionally, FTO overexpression inhibited cell autophagy and apoptosis while promoting cell viability of LPS-induced HK-2 cells. Moreover, overexpression of FTO inhibited SNHG14 expression and autophagy in LPS-induced AKI mice. Functionally, SNHG14 acts as a competing endogenous RNA (ceRNA) via directly sponging miR-373-3p in LPS induced HK-2 cells. Additionally, miR-373-3p directly targets ATG7. Inhibition of SNHG14 suppresses NF-κB signaling pathway and production of inflammatory cytokines (TNF-α, IL-6, and IL-1β) via miR-373-3p/ATG7 in LPS-induced HK-2 cells. Furthermore, the SNHG14/miR-373-3p/ATG7 interaction network contributes to the regulatory effect of FTO on LPS-induced HK-2 cell viability, apoptosis and autophagy. These results suggested demethylase FTO suppressed the m6A modification of lncRNA SNHG14 and inhibits autophagy in LPS-induced AKI via regulating miR-373-3p/ATG7, which provided an important novel perspective for understanding sepsis-associated AKI and is conducive for developing new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Ni Yang
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Na Yan
- Cardio-cerebrovascular Hospital, Affiliated Hospital of Yan'an University, Yan'an, China
| | - Zhenghai Bai
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuang Du
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingjing Zhang
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Zhang
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhengliang Zhang
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
13
|
Wang B, Wei Y, Han T, Ji P, Miao H, Wu X, Qian J, Shao P. LncRNA LBX2-AS1 promotes proliferation and migratory capacity of clear cell renal cell carcinoma through mitophagy. Eur J Med Res 2024; 29:103. [PMID: 38326905 PMCID: PMC10848470 DOI: 10.1186/s40001-024-01690-1] [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: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been extensively investigated in the field of cancer, among which, lncRNA ladybird homeobox 2-antisense RNA 1 (LBX2-AS1) has been demonstrated to exert carcinogenic effects on a variety of malignancies. However, the biological functions of LBX2-AS1 in clear cell renal cell carcinoma (ccRCC) have not been explicitly elucidated. METHODS Arraystar lncRNA chip and qRT-PCR verify the expression of LncRNA LBX2-AS1 in ccRCC. CCK-8 assay and cell cloning assay were used to assess the proliferative capacity of ccRCC cells. Migration abilities were quantified by scratch assay and transwell assay. Potential molecular signaling pathways were determined by high-throughput whole transcriptomics analysis. WB analysis was performed to validate the relationship between LBX2-AS1 and key molecules of mitophagy pathway. The effect of LBX2-AS1 on mitophagy was observed by laser confocal microscopy. Rescue experiments further validated the role of downstream gene FOXO3A in the LBX2-AS1 signaling pathway. Finally, the authentic effect of LBX2-AS1 was verified in vivo. RESULTS LncRNA LBX2-AS1 was over expressed in ccRCC tissues and could enhance the proliferation and migration of ccRCC cells. Autophagic pathway was identified as a possible mechanism involved in the oncogenic effect of LBX2-AS1. Mitophagy levels were observed in LBX2-AS1 low-expressing cells through laser confocal microscopy. Knockdown of LBX2-AS1 significantly elevated mitophagy levels as observed using laser confocal microscopy and led to FOXOA3 decreasing in and BNIP3L and LC3 enrichment. Meanwhile, LBX2-AS1 knocking down dampened the proliferation of ccRCC cells in vivo.
Collapse
Affiliation(s)
- Bao Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuang Wei
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tian Han
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Ji
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haoqi Miao
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangzheng Wu
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Qian
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Pengfei Shao
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
14
|
Zhang X, Wang L, Li M, Dong S. Predictive value of miR-7110-5p and miR-223-3p as biomarkers for sepsis secondary to pneumonia. Technol Health Care 2024; 32:2931-2939. [PMID: 38759032 DOI: 10.3233/thc-231137] [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] [Indexed: 05/19/2024]
Abstract
BACKGROUND Investigating the secondary sepsis of pneumonia is of great significance for rapid diagnosis and early treatment of sepsis. OBJECTIVE This study aimed to investigate the predictive value of micro ribonucleic acids (miRNA) 7110-5p and miR-223-3p in sepsis secondary to pneumonia. A miRNA microarray was used to analyze the differences in miRNA expression between patients with pneumonia and those with sepsis secondary to pneumonia. METHODS The study included a total of 50 patients with pneumonia and 42 patients with sepsis secondary to pneumonia. Quantitative polymerase chain reaction analysis was conducted to measure the circulating miRNA expression levels in patients and assess their correlations with clinical characteristics and prognosis. In this study, nine miRNAs - hsa-miR-4689-5p, hsa-miR-4621-5p, hsa-miR-6740-5p, hsa-miR-7110-5p, hsa-miR-765, hsa-miR-940, hsa-miR-213-5p, hsa-miR-223-3p, and hsa-miR-122 - met the screening criteria of having a fold change ⩾ 2 or < 0.5; p< 0.01 indicated significant differences in the results. RESULTS The expression levels of miR-7110-5p and miR-223-3p differed between the two patient groups, being up-regulated in the plasma of patients with sepsis secondary to pneumonia. miR-7110-5p and miR-223-3p showed higher expression levels in both patients with pneumonia and sepsis compared to healthy controls. Moreover, the receiver operating characteristic curve revealed that the areas under the curve for predicting pneumonia using miR-7110-5p were 0.781 while those for predicting sepsis secondary to pneumonia were 0.862. For miR-223-3p, the corresponding values for predicting pneumonia and sepsis secondary to pneumonia were 0.879 and 0.924, respectively. However, there were no significant differences in the levels of miR-7110-5p and miR-223-3p between the plasma of survived and deceased patients with sepsis. CONCLUSIONS MiR-7110-5p and miR-223-3p have the potential to serve as biological indicators for predicting sepsis secondary to pneumonia.
Collapse
Affiliation(s)
- Xinliang Zhang
- Department of Emergency Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lin Wang
- Department of Trauma Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mei Li
- Department of Emergency Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shimin Dong
- Department of Emergency Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
15
|
Zhao Y, Lu C, Zhu R, Hu X. Study on identification of a three-microRNA panel in serum for diagnosing neonatal early onset sepsis. J Matern Fetal Neonatal Med 2023; 36:2280527. [PMID: 37968923 DOI: 10.1080/14767058.2023.2280527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Comparing with other diseases, early onset sepsis (EOS) is a global health concern in neonatal period for its high morbidity and mortality rates. In recent years, many studies have contributed to the figure out the expression patterns of circulating micro-RNAs (miRNAs) in different diseases and progressions, which could function as diagnostic biomarkers for EOS. The purpose of this study was to analyze the expression patterns of selected miRNAs and evaluate their diagnostic value for early detection and treatment. METHODS This was a prospective cross-sectional study conducted from 1 July 2021 to 30 June 2022. We collected surplus peripheral blood and demographic statistics of septic neonates and non-infected neonates during the first 24 h after delivery and obtained 11 candidate miRNAs by literature screening. First, we extracted the candidate miRNAs from the serum of selected neonates and analyzed their expression levels, and then the receiver operating characteristic (ROC) curve was used to select the differentially expressed miRNAs. We analyzed their sensitivity and specificity and obtained the best diagnostic panel. Finally, with the help of differentially expressed miRNAs, we performed gene ontology (GO) enrichment and protein-protein interaction (PPI) analyses by their target genes. RESULTS In patients with EOS, three miRNAs (mir-223-3p, mir-15a-5p, and mir-17-5p) in serum were significantly downregulated, and mir-146a-5p, mir-1-3p, and mir-16-5p were upregulated. The diagnostic value of these miRNAs (miR-15a-5p, AUC = 0.67; miR-223-3p, AUC = 0.72; miR-16-5p, AUC = 0.68; miR-17-5p, AUC = 0.70; miR-1-3p, AUC = 0.69; miR-146a-5p, AUC = 0.72) was moderate, and the diagnostic panel constructed by miR-15a-5p, miR-223-3p, and miR-16-5p possessed a comparatively higher diagnostic value (AUC = 0.85, sensitivity: 74.6%, specificity: 86%), indicating that their combined application may be a promising biomarker for clinical diagnosis of EOS. According to GO enrichment analysis, most proteins encoded by target genes were located in the cytosol as for cellular component (CC), for molecular function (MF), most proteins acted as regulators in protein binding, and for biological process (BP). Most genes function in positive or negative regulation of transcription from RNA polymerase II promoter, and the top 10 hub genes were CDKN1A, YAP1, CCNE1, CCND1, CKK6, ERBB4, CHEK1, DICER1, VEGFA, and APP by rank degree after PPI construction. CONCLUSIONS The three-miRNA panels (miR-15a-5p, miR-223-3p, and miR-16-5p) may be a novel noninvasive biological marker for EOS screening.
Collapse
Affiliation(s)
- Yihong Zhao
- Pediatrics Department, Peking University Shenzhen Hospital, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Chong Lu
- Pediatrics Department, Peking University Shenzhen Hospital, Shenzhen, China
- Anhui Medical University, Hefei, China
| | - Ruqin Zhu
- Pediatrics Department, Peking University Shenzhen Hospital, Shenzhen, China
- Anhui Medical University, Hefei, China
| | - Xiaoyan Hu
- Pediatrics Department, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
16
|
Ren Q, Xu Y, Xu L, Lu Y, Zheng Y. Hypoxic bone marrow mesenchymal stem cell-derived exosomal lncRNA XIST attenuates lipopolysaccharide-induced acute lung injury via the miR-455-3p/Claudin-4 axis. Int Immunopharmacol 2023; 125:111066. [PMID: 37866316 DOI: 10.1016/j.intimp.2023.111066] [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: 08/18/2023] [Revised: 10/03/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Mesenchymal stem cell-derived exosomes and long non-coding RNAs (lncRNAs) have been identified to play a role in acute lung injury (ALI). In this study, we investigated whether exosomal lncRNAs could regulate ALI and the underlying mechanisms. Bone marrow mesenchymal stem cells (BM-MSCs) were pretreated with hypoxia or normoxia, and exosomes were subsequently extracted from normoxic BM-MSCs (Nor-exos) and hypoxic BM-MSCs (Hypo-exos). A rat model of ALI was established via an airway perfusion of lipopolysaccharide (LPS). Exosomes were administered via the tail vein to evaluate the in vivo effect of exosomes in ALI. LPS-exposed RLE-6TN cells were incubated with exosomes to explore their in vitro effect in ALI. A luciferase reporter assay was used to evaluate the interaction between lncRNA XIST and miR-455-3p, as well as miR-455-3p and Claudin-4. We found that the exosomes attenuated LPS-induced ALI and Hypo-Exos exerted a greater therapeutic effect compared with Nor-exos both in vitro and in vivo. Moreover, an abundance of lncRNA XIST was observed in Hypo-exos compared with Nor-exos. Mechanistically, LncRNA XIST functioned as a miR-455-3p sponge and targeted Claudin-4 in ALI. Our results provide novel insight into the role of exosomal lncRNA XIST for the treatment of ALI. Thus, hypoxic pretreatment may represent an effective method for improving the therapeutic effects of exosomes.
Collapse
Affiliation(s)
- Qinghuan Ren
- Alberta College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingge Xu
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liming Xu
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuanqiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yueliang Zheng
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China; The First People's Hospital of Aksu District in Xinjiang, Aksu, Xinjiang, China.
| |
Collapse
|
17
|
Xue J, Liu L, Liu H, Li Z. LncRNA SNHG14 activates autophagy via regulating miR-493-5p/Mef2c axis to alleviate osteoporosis progression. Commun Biol 2023; 6:1120. [PMID: 37925525 PMCID: PMC10625635 DOI: 10.1038/s42003-023-05493-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/19/2023] [Indexed: 11/06/2023] Open
Abstract
Osteoporosis is a progressive bone disease caused by impaired function of endogenous bone marrow-derived mesenchymal stem cells (BMSCs). Herein, we investigated the mechanism of lncRNA SNHG14 in osteoporosis progression. BMSCs were isolated from BALB/c mice. The osteogenic ability of BMSCs was assessed by Alkaline phosphatase (ALP) and Alizarin Red S Staining (ARS) staining. The interaction between miR-493-5p and SNHG14 or myocyte enhancer factor 2 C (Mef2c) was confirmed by dual-luciferase reporter assay. Bone histomorphometry changes were evaluated to analyze SNHG14'roles in osteoporosis in vivo. Our results illustrated SNHG14 and Mef2c levels were increased in a time-dependent manner in BMSCs, and miR-493-5p expression was decreased. SNHG14 knockdown inhibited osteogenic differentiation of BMSCs, and SNHG14 upregulation had the opposite effect. SNHG14 overexpression elevated bone mineral density and bone trabecular number, and alleviated osteoporosis progression in vivo. Mechanically, miR-493-5p was a target of SNHG14, and miR-493-5p targeted the Mef2c gene directly. SNHG14 overexpression reversed the inhibition of miR-493-5p on the osteogenic ability of BMSCs, and miR-493-5p silencing accelerated BMSCs osteogenesis by activating Mef2c-mediated autophagy to accelerate BMSCs osteogenesis. In short, SNHG14 activated autophagy via regulating miR-493-5p/Mef2c axis to alleviate osteoporosis progression, which might provide a new molecular target for osteoporosis treatment.
Collapse
Affiliation(s)
- Jingbo Xue
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China.
| | - Lulu Liu
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Hao Liu
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Zepeng Li
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| |
Collapse
|
18
|
Zhao J, Liang Q, Fu C, Cong D, Wang L, Xu X. Autophagy in sepsis-induced acute lung injury: Friend or foe? Cell Signal 2023; 111:110867. [PMID: 37633477 DOI: 10.1016/j.cellsig.2023.110867] [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: 06/15/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening syndrome with high mortality and morbidity, resulting in a heavy burden on family and society. As a key factor that maintains cellular homeostasis, autophagy is regarded as a self-digesting process by which damaged organelles and useless proteins are recycled for cell metabolism, and it thus plays a crucial role during physiological and pathological processes. Recent studies have indicated that autophagy is involved in the pathophysiological process of sepsis-induced ALI, including cell apoptosis, inflammation, and mitochondrial dysfunction, which indicates that regulating autophagy may be beneficial for this disease. However, the role of autophagy in the etiology and treatment of sepsis-induced ALI is not well characterized. This review summarizes the autophagy-related signaling pathways in sepsis-induced ALI, as well as focuses on the dual role of autophagy and its regulation by non-coding RNAs during disease progression, for the development of potential therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Jiayao Zhao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qun Liang
- Department of Critical Care Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chenfei Fu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Didi Cong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Long Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaoxin Xu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
19
|
Wang D, Wang X, Yang T, Tian H, Su Y, Wang Q. Long Non-Coding RNA Dancr Affects Myocardial Fibrosis in Atrial Fibrillation Mice via the MicroRNA-146b-5p/Smad5 Axis. ACTA CARDIOLOGICA SINICA 2023; 39:841-853. [PMID: 38022420 PMCID: PMC10646592 DOI: 10.6515/acs.202311_39(6).20230619b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/19/2023] [Indexed: 12/01/2023]
Abstract
Objectives Atrial fibrillation (AF) is the most frequent arrhythmia, and myocardial fibrosis (MF) has a close association with atrial remodeling and leads to AF. This study aimed to explore the function of the long non-coding RNA (lncRNA) differentiation antagonizing non-protein coding RNA (Dancr)/microRNA (miR)-146b-5p/Smad5 axis on MF in AF mice. Methods AF mouse models were established. Overexpression Dancr lentivirus was injected into AF mice to increase Dancr expression in myocardial tissues. LncRNA Dancr, miR-146b-5p, and Smad5 expression levels and inflammatory factors (IL-18 and TNF-α) in the myocardial tissues were measured. MF was measured and the expression levels of MF-related genes (COL1A1, α-SMA, and FN1) were detected. In addition, in vitro HL-1 cell rapid pacing models were constructed, and after lncRNA Dancr and miR-146b-5p-related construct transfection, cell viability and cell apoptosis were determined. Results LncRNA Dancr up-regulation ameliorated MF in the AF mice, reduced IL-18 and TNF-α expression levels in myocardial tissues, and decreased COL1A1, α-SMA, and FN1 expression levels. The in vitro HL-1 cell rapid pacing models suggested that miR-146b-5p overexpression reversed the inhibitory effects of lncRNA Dancr overexpression on MF in HL-1 cells, and Smad5 interference reversed the ameliorative effects of miR-146b-5p interference on MF in HL-1 cells. Conclusions LncRNA Dancr can sponge miR-146b-5p to promote Smad5 expression, thereby delaying MF in AF mice.
Collapse
Affiliation(s)
- Dejin Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Xiqian Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Tianxiao Yang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Hongliang Tian
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Yuanzhen Su
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Qilei Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| |
Collapse
|
20
|
Wu Z, Wang Y, Lu S, Yin L, Dai L. SIRT3 alleviates sepsis-induced acute lung injury by inhibiting pyroptosis via regulating the deacetylation of FoxO3a. Pulm Pharmacol Ther 2023; 82:102244. [PMID: 37499855 DOI: 10.1016/j.pupt.2023.102244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
OBJECTIVE This study mainly analyzes the mechanism of SIRT3 alleviating sepsis-induced acute lung injury (ALI) by regulating the deacetylation of FoxO3a and inhibiting pyroptosis. METHODS SIRT3-overexpressing and silenced BEAS-2B cells were used to evaluate the effect of SIRT3 on apoptosis in LPS-treated lung epithelial cells. FoxO3a-silenced BEAS-2B cells were also used to verify the mechanism by which SIRT3 inhibited oxidative stress and pyroptosis in vitro in ALI. 3-TYP was used to inhibit the deacetylation function of SIRT3 in vivo. Pyroptosis was assessed by detecting GSDMD-N and LDH efflux. RESULTS In CLP-induced ALI mice, GSDMD-N and LDH levels were elevated, pyroptosis was induced. Silencing of SIRT3 exacerbated oxidative stress, NLRP3 activation and pyroptosis, and inhibited the deacetylation of FoxO3a. Overexpression of SIRT3 attenuated pyroptosis, induced deacetylation and restored the expression of FoxO3a and MnSOD. Silencing FoxO3a aggravated pyroptosis. Overexpression of SIRT3 restored the reduced FoxO3a expression and suppressed pyroptosis. 3-TYP blocked the promotion of FoxO3a by SIRT3 and the inhibitory effect of SIRT3 on pyroptosis. CONCLUSION The reduction of SIRT3 in sepsis caused hyperacetylation of FoxO3a, which in turn exacerbates oxidative stress and induces pyroptosis of ALI. Increasing the level of SIRT3 promotes FoxO3a through deacetylation, thereby inhibiting pyroptosis and relieving ALI.
Collapse
Affiliation(s)
- Zheqian Wu
- Department of Emergency, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Yong Wang
- Department of Emergency, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Shijie Lu
- Department of Emergency, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Lili Yin
- Department of Emergency, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China
| | - Lihua Dai
- Department of Emergency, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China.
| |
Collapse
|
21
|
Miao RF, Tu J. LncRNA CDKN2B-AS1 interacts with LIN28B to exacerbate sepsis-induced acute lung injury by inducing HIF-1α/NLRP3-mediated pyroptosis. Kaohsiung J Med Sci 2023; 39:883-895. [PMID: 37265187 DOI: 10.1002/kjm2.12697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 06/03/2023] Open
Abstract
Sepsis-associated acute lung injury (ALI) is a life-threatening condition in intensive care units with high mortality. LncRNAs have been confirmed to participate in the underlying pathogenesis of septic ALI. This study investigated the biological functions of lncRNA CDKN2B-AS1 in septic ALI and its potential mechanism.BEAS-2B cells were challenged with lipopolysaccharide (LPS) and mice were subjected to caecal ligation and puncture (CLP) to induce septic ALI in vitro and in vivo. The expression levels of CDKN2B-AS1, LIN28B, HIF-1α, and pyroptosis-related molecules were assessed by qRT-PCR or Western blotting. The production of IL-1β and IL-18 was detected by ELISA. BEAS-2B cell pyroptosis was examined by flow cytometry. The interaction between LIN28B and CDKN2B-AS1/HIF-1α was validated by RIP and RNA pull-down assays. Colocalization of CDKN2B-AS1 and LIN28B was observed by FISH. ALI was determined by HE staining, the lung wet-to-dry (W/D) weight ratio, inflammatory cell numbers, and total protein concentration in bronchoalveolar lavage fluid (BALF). Caspase-1 expression in the lung tissues was examined by immunohistochemical staining.CDKN2B-AS1 was upregulated in BEAS-2B cells after LPS stimulation. CDKN2B-AS1 knockdown inhibited pyroptosis in LPS-exposed BEAS-2B cells in vitro and the lung tissues of septic mice in vivo. Mechanistically, CDKN2B-AS1 interacted with LIN28B to enhance HIF-1α stability. Rescue experiments showed that HIF-1α overexpression counteracted the inhibitory effect of sh-CDKN2B-AS1 on LPS-induced pyroptosis. CDKN2B-AS1 bound to LIN28B to trigger NLRP3-mediated pyroptosis by stabilizing HIF-1α, which promoted sepsis-induced ALI. CDKN2B-AS1 might be a novel therapeutic target for this disease.
Collapse
Affiliation(s)
- Run-Feng Miao
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, People's Republic of China
| | - Jing Tu
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, People's Republic of China
| |
Collapse
|
22
|
Li Y, Xiao W, Lin X. Long noncoding RNA MALAT1 inhibition attenuates sepsis-induced acute lung injury through modulating the miR-129-5p/PAX6/ZEB2 axis. Microbiol Immunol 2023; 67:142-153. [PMID: 36537561 DOI: 10.1111/1348-0421.13045] [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: 09/29/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
This research aimed to investigate the role of the long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)/microRNA-129-5p (miR-129-5p)/paired box gene 6 (PAX6) axis in sepsis-induced acute lung injury (ALI). MLE-12 cells and C57BL/6 mice were induced by LPS to establish lung injury in in vitro and in vivo models. Cell viability and apoptosis were measured by cell counting kit-8 assay and TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, respectively. Levels of inflammatory cytokines in cell supernatants and bronchoalveolar lavage fluid (BALF) were detected by ELISA. Lung injury was evaluated by lung wet weight-to-dry weight ratio and hematoxylin-eosin staining. MALAT1, PAX6, and zinc finger E-box-binding homeobox 2 (ZEB2) expression was elevated and miR-129-5p expression was reduced in the serum of patients with sepsis-induced ALI, LPS-induced MLE-12 cells, and lung tissues of ALI mice. MALAT1 interference delayed the LPS-induced cell proliferation decrease, apoptosis increase, and inflammatory factor increase. miR-129-5p inhibition could reverse the delaying effect of MALAT1 interference on LPS-induced lung cell injury. PAX6 overexpression (oe) reversed the inhibitory effect of miR-129-5p oe on LPS-induced lung cell injury. Downregulating MALAT1 reduced pulmonary edema, inflammatory cytokine levels, lung injury, and apoptosis in ALI mice. Moreover, miR-129-5p suppression or PAX6 oe reversed the delaying effect of MALAT1 interference on sepsis-induced ALI. MALAT1 aggravates sepsis-induced ALI via the miR-129-5p/PAX6/ZEB2 axis.
Collapse
Affiliation(s)
- Ying Li
- Department of Critical Care Medicine, The First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Wenbiao Xiao
- Department of Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Xiao Lin
- Department of Critical Care Medicine, The First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| |
Collapse
|
23
|
Yang K, Zeng L, Ge A, Wang S, Zeng J, Yuan X, Mei Z, Wang G, Ge J. A systematic review of the research progress of non-coding RNA in neuroinflammation and immune regulation in cerebral infarction/ischemia-reperfusion injury. Front Immunol 2022; 13:930171. [PMID: 36275741 PMCID: PMC9585453 DOI: 10.3389/fimmu.2022.930171] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022] Open
Abstract
Cerebral infarction/ischemia-reperfusion injury is currently the disease with the highest mortality and disability rate of cardiovascular disease. Current studies have shown that nerve cells die of ischemia several hours after ischemic stroke, which activates the innate immune response in the brain, promotes the production of neurotoxic substances such as inflammatory cytokines, chemokines, reactive oxygen species and − nitrogen oxide, and mediates the destruction of blood-brain barrier and the occurrence of a series of inflammatory cascade reactions. Meanwhile, the expression of adhesion molecules in cerebral vascular endothelial cells increased, and immune inflammatory cells such as polymorphonuclear neutrophils, lymphocytes and mononuclear macrophages passed through vascular endothelial cells and entered the brain tissue. These cells recognize antigens exposed by the central nervous system in the brain, activate adaptive immune responses, and further mediate secondary neuronal damage, aggravating neurological deficits. In order to reduce the above-mentioned damage, the body induces peripheral immunosuppressive responses through negative feedback, which increases the incidence of post-stroke infection. This process is accompanied by changes in the immune status of the ischemic brain tissue in local and systemic systems. A growing number of studies implicate noncoding RNAs (ncRNAs) as novel epigenetic regulatory elements in the dysfunction of various cell subsets in the neurovascular unit after cerebral infarction/ischemia-reperfusion injury. In particular, recent studies have revealed advances in ncRNA biology that greatly expand the understanding of epigenetic regulation of immune responses and inflammation after cerebral infarction/ischemia-reperfusion injury. Identification of aberrant expression patterns and associated biological effects of ncRNAs in patients revealed their potential as novel biomarkers and therapeutic targets for cerebral infarction/ischemia-reperfusion injury. Therefore, this review systematically presents recent studies on the involvement of ncRNAs in cerebral infarction/ischemia-reperfusion injury and neuroimmune inflammatory cascades, and elucidates the functions and mechanisms of cerebral infarction/ischemia-reperfusion-related ncRNAs, providing new opportunities for the discovery of disease biomarkers and targeted therapy. Furthermore, this review introduces clustered regularly interspaced short palindromic repeats (CRISPR)-Display as a possible transformative tool for studying lncRNAs. In the future, ncRNA is expected to be used as a target for diagnosing cerebral infarction/ischemia-reperfusion injury, judging its prognosis and treatment, thereby significantly improving the prognosis of patients.
Collapse
Affiliation(s)
- Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Yuan
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Hunan Academy of Chinese Medicine, Changsha, China
- *Correspondence: Jinwen Ge,
| |
Collapse
|
24
|
MiR-223-3p-loaded exosomes from bronchoalveolar lavage fluid promote alveolar macrophage autophagy and reduce acute lung injury by inhibiting the expression of STK39. Hum Cell 2022; 35:1736-1751. [PMID: 35932362 DOI: 10.1007/s13577-022-00762-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/24/2022] [Indexed: 11/04/2022]
Abstract
This study investigated the molecular mechanism by which bronchoalveolar lavage fluid exosomes (BALF-exo) alleviated acute lung injury (ALI). BALF-exo was isolated from mice. LPS was used to induce inflammation in rat alveolar macrophages (NR8383). NR8383 cell models were treated with BALF-exo or BALF-exo loaded with miR-223-3p mimics/inhibitors, or STK39 was overexpressed in NR8383 cells before LPS and BALF-exo treatment. These cells were subjected to apoptosis, autophagy, and inflammation assays. RNA immunoprecipitation and dual-luciferase reporter assay were conducted to verify the binding between miR-223-3p and STK39. LPS-induced ALI mouse models were treated with BALF-exo loaded with miR-223-3p mimics. miR-223-3p was lowly expressed in BALF-exo from LPS-treated mice. BALF-exo loaded with miR-223-3p mimics increased viability and autophagy and decreased apoptosis and inflammation in NR8383 cell models. Inhibition of miR-223-3p in BALF-exo or overexpression of STK39 in NR8383 cells repressed the therapeutic effect of BALF-exo in LPS-treated NR8383 cells. STK39 was a target gene of miR-223-3p. miR-223-3p shuttled by BALF-exo negatively regulated the expression of STK39 in NR8383 cells. BALF-exo loaded with miR-223-3p mimics also reduced lung injuries in ALI mice. In conclusion, miR-223-3p loaded in BALF-exo alleviates ALI by targeting STK39 in alveolar macrophages.
Collapse
|
25
|
Zhang G, Guo J, Zeng J, Zhang X, Chen R, Wang G, Liang W. LncRNA SNHG14 is beneficial to oxygen glucose deprivation/reoxygenation-induced neuro-2a cell injury via mir-98-5p sequestration-caused BCL2L13 upregulation. Metab Brain Dis 2022; 37:2005-2016. [PMID: 35678981 DOI: 10.1007/s11011-022-01009-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The deregulation of long non-coding RNA (lncRNA) is associated with diverse human disorders, including cerebral ischemia/reperfusion injury (CI/RI). LncRNA SNHG14 was reported to function in CI/RI. Whereas, molecular mechanisms regulated by SNHG14 are not fully unveiled. METHODS Mice subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) were used as CI/RI animal models. Neuro-2a (N2A) cells subjected to oxygen glucose deprivation/reoxygenation (OGD/R) were used as CI/RI cell models. The expression of SNHG14, miR-98-5p and BCL2 like 13 (BCL2L13) was examined using quantitative real-time PCR (qPCR) or western blot. Apoptosis was monitored by flow cytometry assay. Apoptosis-related markers and endoplasmic reticulum (ER) stress-related markers were quantified by western blot. Inflammatory factors and oxidative stress were detected using matched commercial kits. The predicted relationship between miR-98-5p and SNHG14 or BCL2L13 was validated by dual-luciferase reporter assay, RIP assay and pull-down assay. RESULTS The high expression of SNHG14 was monitored in MCAO/R-treated mice and OGD/R-treated N2A cells. OGD/R-induced N2A cell apoptosis, ER stress, inflammation and oxidative stress were attenuated by SNHG14 knockdown. SNHG14 targeted miR-98-5p to positively regulate BCL2L13 expression. Inhibition of miR-98-5p recovered cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by SNHG14 knockdown. Overexpression of BCL2L13 enhanced cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by miR-98-5p enrichment. CONCLUSIONS SNHG14 knockdown alleviated OGD/induced N2A cell apoptosis, ER stress, inflammation and oxidative stress by depleting BCL2L13 via increasing miR-98-5p.
Collapse
Affiliation(s)
- Gaolian Zhang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Jianhui Guo
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Jing Zeng
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Xiaoning Zhang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Rui Chen
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Gang Wang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Weibin Liang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China.
| |
Collapse
|
26
|
TNF- α Induces Neutrophil Apoptosis Delay and Promotes Intestinal Ischemia-Reperfusion-Induced Lung Injury through Activating JNK/FoxO3a Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:8302831. [PMID: 35003520 PMCID: PMC8731283 DOI: 10.1155/2021/8302831] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
Background Intestinal ischemia is a common clinical critical illness. Intestinal ischemia-reperfusion (IIR) leads to acute lung injury (ALI), but the causative factors of ALI are unknown. The aim of this study was to reveal the causative factors and mechanisms of IIR-induced lung injury. Methods A mouse model of IIR was developed using C57BL/6 mice, followed by detection of lung injury status and plasma levels of inflammatory factors in sham-operated mice and model mice. Some model mice were treated with a tumor necrosis factor-α (TNF-α) inhibitor lenalidomide (10 mg/kg), followed by observation of lung injury status through hematoxylin and eosin staining and detection of neutrophil infiltration levels through naphthol esterase and Ly6G immunohistochemical staining. Additionally, peripheral blood polymorphonuclear neutrophils (PMNs) were cultured in vitro and then stimulated by TNF-α to mimic in vivo inflammatory stimuli; this TNF-α stimulation was also performed on PMNs after knockdown of FoxO3a or treatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125. PMN apoptosis after stimulation was detected using flow cytometry. Finally, the role of PMN apoptosis in IIR-induced lung injury was evaluated in vivo by detecting the ALI status in the model mice administered with ABT-199, a Bcl-2 inhibitor. Results IIR led to pulmonary histopathological injury and increased lung water content, which were accompanied by increased plasma levels of inflammatory factors, with the TNF-α plasma level showing the most pronounced increase. Inhibition of TNF-α led to effective reduction of lung tissue injury, especially that of the damaging infiltration of PMNs in the lung. In vitro knockdown of FoxO3a or inhibition of JNK activity could inhibit TNF-α-induced PMN apoptosis. Further in vivo experiments revealed that ABT-199 effectively alleviated lung injury and decreased inflammation levels by promoting PMN apoptosis during IIR-induced lung injury. Conclusion TNF-α activates the JNK/FoxO3a pathway to induce a delay in PMN apoptosis, which promotes IIR-induced lung injury.
Collapse
|
27
|
Li X. LncRNA MALAT1 promotes osteogenic differentiation of BMSCs and inhibits osteoclastic differentiation of Mø in osteoporosis via the miR-124-3p/IGF2BP1/Wnt/β-catenin axis. J Tissue Eng Regen Med 2021; 16:311-329. [PMID: 34962086 DOI: 10.1002/term.3279] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 11/12/2022]
Abstract
Osteoporosis is defined as a skeletal disorder characterized by impairment in bone strength. The potential application of lncRNAs as therapeutic targets for osteoporosis has been unveiled. This study investigated the regulatory mechanism of lncRNA MALAT1 in the differentiation of bone marrow stem cells (BMSCs) and macrophages (Mø) in osteoporosis. MALAT1 expression in peripheral blood of elderly osteoporosis patients and healthy volunteers was detected. BMSCs and mononuclear Mø were isolated and cultured. Osteogenic differentiation of BMSCs and osteoclastic differentiation of Mø were induced. BMSCs and Mø were transfected with si-MALAT1, miR-124-3p mimics, miR-124-3p inhibitor, or pcDNA IGF2BP1, followed by detection of cell differentiation. The target microRNAs (miRs) and downstream genes and signaling pathways of MALAT1 were examined. The ovariectomy-induced mouse model of osteoporosis was established, and the mice were injected with pcDNA-MALAT1. MALAT1 was downregulated in osteoporosis patients, increased in BMSCs after osteogenic differentiation, and diminished in Mø after osteoclastic differentiation. Downregulation of MALAT1 repressed osteogenic differentiation of BMSCs and facilitated osteoclastic differentiation of Mø. MALAT1 upregulated IGF2BP1 expression by competitively binding to miR-124-3p. miR-124-3p silencing reversed the effect of si-MALAT1 on BMSCs and Mø differentiation, and IGF2BP1 upregulation averted the effect of overexpressed-miR-124-3p by activating the Wnt/β-catenin pathway. Upregulation of MALAT1 activated the Wnt/β-catenin pathway and attenuated bone injury in mice. In conclusion, lncRNA MALAT1 promoted the osteogenic differentiation of BMSCs and inhibited osteoclastic differentiation of Mø in osteoporosis via the miR-124-3p/IGF2BP1/Wnt/β-catenin axis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xiangxin Li
- Department of Pain, Changchun University of Traditional Chinese Medicine Affiliated Hospital, Changchun, 130021, Jilin, China
| |
Collapse
|