101
|
Wang M, Wei J, Shang F, Zang K, Ji T. Long non‑coding RNA CASC2 ameliorates sepsis‑induced acute kidney injury by regulating the miR‑155 and NF‑κB pathway. Int J Mol Med 2020; 45:1554-1562. [PMID: 32323747 DOI: 10.3892/ijmm.2020.4518] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/19/2019] [Indexed: 11/05/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome that can cause multiple‑organ damage, including acute kidney injury (AKI). Studies have shown that the long non‑coding RNA cancer susceptibility candidate 2 (CASC2) is involved in the occurrence and development of multiple human diseases, although its expression and role in AKI has not yet been reported. The present study demonstrated that the expression of CASC2 was significantly decreased in the serum of patients with sepsis compared with healthy subjects. In addition, the CASC2 level was negatively associated with the severity of AKI. Further experiments revealed that CASC2 promoted cell viability and inhibited inflammatory factor secretion, apoptosis and oxidative stress in lipopolysaccharide‑stimulated human renal tubular epithelial HK‑2 cells. Importantly, the current study observed that CASC2 was negatively associated with a pro‑inflammatory microRNA (miR)‑155. In addition, the upregulation of CASC2 significantly suppressed the nuclear factor κB (NF‑κB) signaling pathway. In conclusion, the results of the present study suggested that CASC2 may serve as a potential target for treating sepsis‑induced AKI by inhibiting the miR‑155 and NF‑κB pathway‑mediated inflammation.
Collapse
Affiliation(s)
- Min Wang
- Department of Intensive Care Unit, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Jilou Wei
- Department of Intensive Care Unit, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Futai Shang
- Department of Intensive Care Unit, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Kui Zang
- Department of Intensive Care Unit, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Ting Ji
- Department of Intensive Care Unit, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
102
|
Guo L, Liu J, Zhang Y, Fu S, Qiu Y, Ye C, Liu Y, Wu Z, Hou Y, Hu CAA. The Effect of Baicalin on the Expression Profiles of Long Non-Coding RNAs and mRNAs in Porcine Aortic Vascular Endothelial Cells Infected with Haemophilus parasuis. DNA Cell Biol 2020; 39:801-815. [PMID: 32096672 DOI: 10.1089/dna.2019.5340] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Haemophilus parasuis can elicit serious inflammatory responses, which contribute to huge economic losses to the swine industry. However, the pathogenic mechanisms underlying inflammation-related damage induced by H. parasuis remain unclear. Accumulating evidence indicates that long non-coding RNAs (lncRNAs) have important functions in the regulation of autoimmune disorders. Baicalin has been shown to have anti-inflammatory, anti-microbial, and anti-oxidant activities. In this study, we investigated whether lncRNAs were involved in the vascular injury or inflammation triggered by H. parasuis and whether baicalin regulated the lncRNA profiles of porcine aortic vascular endothelial cells (PAVECs) infected with H. parasuis. The results showed that the lncRNA and mRNA expression profiles of PAVECs were changed by H. parasuis. Important functions of lncRNAs and mRNAs were predicted. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrated that the targets of differentially expressed lncRNAs of H. parasuis infected PAVECs were mainly involved in the tumor necrosis factor (TNF) signaling pathway, apoptosis, and N-glycan biosynthesis; whereas nicotinate and nicotinamide metabolism, the cytosolic DNA-sensing pathway, the TNF signaling pathway, and the nuclear factor (NF)-kappa B signaling pathway were enriched in PAVECs pretreated with baicalin. In addition, top hub genes and lncRNAs were identified and validated by quantitative polymerase chain reaction. CCL5, GBP1, and SAMHD1 were significantly upregulated after H. parasuis infection, whereas they were significantly downregulated with baicalin pretreatment. LncRNA ALDBSSCT0000001677, ALDBSSCT0000001353, MSTRG.10724.2, and ALDBSSCT0000010434 had the same expression pattern. Collectively, these data suggested that baicalin could modify changes to the lncRNAs profiles or regulate lncRNAs that participate in inflammation-related signaling pathways, thereby alleviating tissue damage or inflammatory responses induced by H. parasuis. To our best knowledge, this is the first article of H. parasuis stimulating changes to the lncRNA profiles of PAVECs and the capability of baicalin to regulate lncRNA changes in PAVECs infected with H. parasuis, which might provide a novel therapeutic target for the control of H. parasuis infection.
Collapse
Affiliation(s)
- Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Jun Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Yunfei Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, P.R. China
| | - Chien-An Andy Hu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, P.R. China.,Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
103
|
Wu H, Wang J, Ma Z. Long noncoding RNA HOXA-AS2 mediates microRNA-106b-5p to repress sepsis-engendered acute kidney injury. J Biochem Mol Toxicol 2020; 34:e22453. [PMID: 32048402 DOI: 10.1002/jbt.22453] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/24/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
HOXA cluster antisense RNA 2 (HOXA-AS2) is a long noncoding RNA associated with the development of numerous cancers. But, whether HOXA-AS2 exhibits a certain function in sepsis-engendered acute kidney injury (AKI) remains uninvestigated. We strived to unveil the role of HOXA-AS2 in sepsis-engendered AKI. The expression of HOXA-AS2 in sepsis patients, animal models and lipopolysaccharide (LPS)-impaired HK-2 cells was primarily assessed via a real-time quantitative polymerase chain reaction. The effects of HOXA-AS2 on cell survival of HK-2 cells under LPS irritation were evaluated after overexpression of HOXA-AS2. The correlation between HOXA-AS2 and microRNA (miR)-106b-5p was forecasted via bioinformatics software and verified by using a luciferase report system. Subsequently, the functions of miR-106b-5p in LPS-damaged HK-2 cells were reassessed. Western blot was used for the determination of Wnt/β-catenin and nuclear factor-κB (NF-κB) pathways. HOXA-AS2 expression was decreased in sepsis patients, animal operation group and LPS-irritated HK-2 cells. Overexpressed HOXA-AS2 mollified LPS-triggered impairment in HK-2 cells. In addition, a negative mediatory relation between HOXA-AS2 and miR-106b-5p was predicated. Synchronously, overexpressed miR-106b-5p counteracted the protection of HOXA-AS2 in LPS-damaged HK-2 cells. Ultimately, Wnt/β-catenin and NF-κB pathways were hindered by HOXA-AS2 via targeting miR-106b-5p. HOXA-AS2 exhibited protection in sepsis-engendered AKI via targeting miR-106b-5p and hindering the Wnt/β-catenin and NF-κB pathways.
Collapse
Affiliation(s)
- Huifeng Wu
- Department of Emergency, Tongchuan People's Hospital, Tongchuan, Shaanxi, China
| | - Jing Wang
- Department of Critical Care Medicine I, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Zhen Ma
- Department of Critical Care Medicine II, Jining No. 1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
104
|
Li S, Zhao D, Cui J, Wang L, Ma X, Li Y. Correlation of microRNA-125a/b with acute respiratory distress syndrome risk and prognosis in sepsis patients. J Clin Lab Anal 2020; 34:e23098. [PMID: 31967348 PMCID: PMC7083491 DOI: 10.1002/jcla.23098] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022] Open
Abstract
Objective This study was conducted to explore the association of microRNA (miR)‐125a and miR‐125b with acute respiratory distress syndrome (ARDS) risk and to investigate their correlation with clinical characteristics and prognosis in sepsis patients. Methods Totally 150 sepsis patients admitted to our hospital were consecutively enrolled and another 150 healthy subjects were enrolled as healthy controls (HCs). Their blood samples were collected for miR‐125a and miR‐125b detection by real‐time quantitative polymerase chain reaction. Besides, ARDS occurrence and 28‐day mortality were documented in all sepsis patients. Results MiR‐125a and miR‐125b relative expressions were increased in ARDS‐sepsis patients/non‐ARDS‐sepsis patients compared with HCs, while only miR‐125b but not miR‐125a was elevated in ARDS‐sepsis patients compared with non‐ARDS‐sepsis patients. Receiver operating characteristic (ROC) curve presented that miR‐125a (AUC: 0.650, 95%CI: 0.549‐0.750) and miR‐125b (AUC: 0.739, 95%CI: 0.653‐0.823) could differentiate ARDS‐sepsis patients from non‐ARDS‐sepsis patients, and miR‐125b was of increased predictive value compared with miR‐125a numerically. In sepsis patients, miR‐125a relative expression was positively associated with serum creatinine (Scr), chronic health evaluation (APACHE) II score, sequential organ failure assessment (SOFA) score, and miR‐125b was positively associated with Scr, C‐reactive protein (CRP), APACHE II score, SOFA score, and chronic obstructive pulmonary disease. All sepsis patients were categorized into survivors and deaths according to 28‐day mortality, and miR‐125b but not miR‐125a was upregulated in deaths compared with survivors. Conclusion Both of miR‐125a and miR‐125b predict ARDS risk, while only miR‐125b is of value in prognosis prediction in sepsis patients.
Collapse
Affiliation(s)
- Shilei Li
- Emergency Department, Cangzhou Central Hospital, Cangzhou, China
| | - Danna Zhao
- Laboratory Department, Cangzhou People Hospital, Cangzhou, China
| | - Jie Cui
- Emergency Department, Cangzhou Central Hospital, Cangzhou, China
| | - Lizeng Wang
- Emergency Department, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaohua Ma
- Emergency Department, Cangzhou Central Hospital, Cangzhou, China
| | - Yong Li
- Emergency Department, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
105
|
Targeting of IL-6-Relevant Long Noncoding RNA Profiles in Inflammatory and Tumorous Disease. Inflammation 2020; 42:1139-1146. [PMID: 30825076 DOI: 10.1007/s10753-019-00995-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin-6 (IL-6) is a critical cytokine with a diverse repertoire of physiological functions. Dysregulation of IL-6 signaling is associated with inflammatory disorders as well as cancers. However, blockade of IL-6 activity via antibodies directed against the IL-6 signaling pathway may compromise the efficacy of the immune system; therefore, patients may not have a uniformly satisfactory response to treatment. Long noncoding RNAs (lncRNAs) have been discovered to be evolutionary conserved transcripts of noncoding DNA sequences and have emerged as biomarkers with great predictive and prognostic value, further employed as a targeted anticancer therapy. LncRNAs have been recently implicated in the regulation of IL-6-related signaling and function; they are tightly linked to the development of a range of IL-6 dysregulated diseases. Here, we will highlight those lncRNAs involved in IL-6 signaling, with an emphasis on the mechanisms of lncRNAs that interact with IL-6. Targeting of such lncRNAs related to IL-6 regulation could be, in the near future, a promising therapeutic strategy in the treatment of inflammatory- and tumor-related diseases.
Collapse
|
106
|
Chen J, He Y, Zhou L, Deng Y, Si L. Long non‑coding RNA MALAT1 serves as an independent predictive biomarker for the diagnosis, severity and prognosis of patients with sepsis. Mol Med Rep 2020; 21:1365-1373. [PMID: 31922243 DOI: 10.3892/mmr.2020.10923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 10/03/2019] [Indexed: 11/06/2022] Open
Abstract
The present prospective study was conducted to investigate the independent risk and predictive value of plasma long non‑coding RNA metastasis‑associated lung adenocarcinoma transcript 1 (MALAT1) as a biomarker for the diagnosis, severity and prognosis of sepsis. A total of 120 patients with sepsis and 60 healthy controls (HCs) were recruited. The expression levels of plasma MALAT1 were detected by quantitative PCR. The results demonstrated that the plasma levels of MALAT1 were significantly increased in patients with sepsis compared with HCs (P<0.001), in patients with septic shock compared with in patients without septic shock (P<0.001), and in non‑survivors compared with in survivors (P<0.001). MALAT1 plasma levels exhibited weak positive correlation with serum procalcitonin levels (r=0.253; P=0.005), arterial lactate levels (r=0.488; P<0.001), sepsis‑related organ failure assessment scores (r=0.566; P<0.001), and acute physiology and chronic health evaluation II scores (r=0.517; P<0.001) in patients with sepsis. Multivariate logistic regression analysis revealed that high MALAT1 expression was an independent risk factor for sepsis (P<0.001), septic shock (P=0.030) and poor prognosis (P=0.015). In addition, the receiver operating characteristic curve exhibited a significant predictive value for MALAT1 in distinguishing patients with sepsis from HCs with an area under the curve (AUC) of 0.910, patients with septic shock from patients without shock with an AUC of 0.836, and non‑survivors from survivors with an AUC of 0.886. In conclusion, plasma MALAT1 may serve as a biomarker for the diagnosis, severity and prognosis of sepsis.
Collapse
Affiliation(s)
- Jianjun Chen
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Yingfeng He
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Liangliang Zhou
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Yijun Deng
- Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224000, P.R. China
| | - Linjie Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
107
|
Silencing of long non-coding RNA MALAT1 suppresses inflammation in septic mice: role of microRNA-23a in the down-regulation of MCEMP1 expression. Inflamm Res 2020; 69:179-190. [PMID: 31893303 DOI: 10.1007/s00011-019-01306-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Sepsis is a life-threatening disease without ideal biomarkers. Some long non-coding RNAs (lncRNAs) are found to be implicated in sepsis. Thus, we investigated the effects of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on inflammation in septic mice and the potential mechanisms of the MALAT1/microRNA-23a (miR-23a)/MCEMP1 axis. METHODS The sepsis mice model was generated by cecal ligation and puncture (CLP). Then the expressions of lncRNA MALAT1, mast cell-expressed membrane protein 1 (MCEMP1), and miR-23a in septic mice were determined. The interaction between lncRNA MALAT1, miR-23a and MCEMP1 was confirmed. Loss- and gain-of-function approaches were used to verify the roles of the lncRNA MALAT1, miR-23a, and MCEMP1 in inflammation, cell proliferation and apoptosis in septic mice. RESULTS AND CONCLUSION The myeloperoxidase (MPO) activity and the expression of interleukin 6 (IL-6), IL-1β, IL-10, and tumor necrosis factor-α (TNF-α) were detected. High expression of the lncRNA MALAT1 and MCEMP1, as well as low expression of miR-23a, was observed in septic mice. LncRNA MALAT1 competitively bound to miR-23a, and miR-23a targeted MCEMP1. Moreover, the down-regulation of lncRNA MALAT1 repressed the expression of MPO, IL-6, IL-10, TNF-α, and IL-1β. Silencing of lncRNA MALAT1 or overexpression of miR-23a reduced inflammation, inhibited cell proliferation, and promoted cell apoptosis in septic mice. Taken together, MALAT1 promotes the inflammation in septic mice by binding to miR-23a to up-regulate MCEMP1. Therefore, silencing of lncRNA MALAT1 might provide a novel therapeutic target for sepsis.
Collapse
|
108
|
Naeli P, Pourhanifeh MH, Karimzadeh MR, Shabaninejad Z, Movahedpour A, Tarrahimofrad H, Mirzaei HR, Bafrani HH, Savardashtaki A, Mirzaei H, Hamblin MR. Circular RNAs and gastrointestinal cancers: Epigenetic regulators with a prognostic and therapeutic role. Crit Rev Oncol Hematol 2020; 145:102854. [PMID: 31877535 PMCID: PMC6982584 DOI: 10.1016/j.critrevonc.2019.102854] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Both environmental and genetic factors are involved in the initiation and development of gastrointestinal cancer. Covalent closed circular RNAs (circRNAs) are produced by a mechanism called "back-splicing" from mRNAs. They are highly stable and show cell and tissue specific expression patterns. Although some functions such as "microRNA sponge" and "RNA binding protein sponge" have been reported for a small number of circRNAs, the function of thousands of other circRNAs is still unknown. Dysregulation of circRNAs has been reported in many GI cancers and are involved in metastasis and invasion. CircRNAs have been reported to be useful as prognostic markers and targets for developing new treatments. We first describe the properties and biogenesis of circRNAs. We then summarize recent reports about circRNA functions, expression status, and their potential to be used as biomarkers in GI cancers including, gastric cancer, colorectal cancer, esophageal cancer, hepatocellular carcinoma, gallbladder cancer and pancreatic cancer.
Collapse
Affiliation(s)
- Parisa Naeli
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran.
| | | | - Mohammad Reza Karimzadeh
- Department of Medical Genetics, School of Medicine, Bam University of Medical Sciences, Bam, Iran.
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, School of Basic Sciences, TarbiatModares University, Tehran, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hassan Hassani Bafrani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
109
|
Wang J, Song J, Li Y, Shao J, Xie Z, Sun K. Down-regulation of LncRNA CRNDE aggravates kidney injury via increasing MiR-181a-5p in sepsis. Int Immunopharmacol 2019; 79:105933. [PMID: 31877497 DOI: 10.1016/j.intimp.2019.105933] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/01/2019] [Accepted: 09/23/2019] [Indexed: 01/10/2023]
Abstract
Long non-coding RNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) is reported to be linked to inflammation and cell apoptosis. However its role in sepsis induced kidney injury remains unclear. This study aims to explore the possible mechanism of CRNDE in kidney injury induced by sepsis. In vivo urine-derived sepsis (US) rat model and in vitro LPS-induced HK-2 and HEK293 cells were established. Kidney function was measured in rats from different groups. Relative levels of tumor necrosis factor-α (TNF-α) and interleukin-1β(IL-1β) in kidney tissue were detected via Enzyme-linked immune sorbent assay (ELISA). Then we up- or down-regulated CRNDE and miRNA-181a-5p expression in the cells. The biological influence of CRNDE and miR-181a-5p on cells was studied using CCK-8 assay and Annexin V assay. Interaction between CRNDE and miR-181a-5p was determined by bioinformatics analysis, RT-PCR, and dual luciferase reporter assay. Peroxisome proliferator-activated receptor-α (PPARα) and cell apoptosis related molecules were detected by western blot. We demonstrated that CRNDE was markedly down-regulated while miR-181a-5p was significantly up-regulated in sepsis models. CRNDE interacted with miR-181a-5p, and negatively regulated its expression level. CRNDE knockdown in rats increased the urea nitrogen and serum creatinine in plasma. Knockdown of CRNDE or transfection of miR-181a-5p significantly inhibited proliferation and promoted apoptosis of HK-2 and HEK293 cells, while overexpression of CRNDE and transfection of miR-181a-5p inhibitors had opposite effects. For mechanism, miR-181a-5p directly targeted the 3' untranslated region of PPARα, and depressed its protein level, and PPARα was regulated indirectly by CRNDE. We concluded that CRNDE protected renal cell from sepsis-induced injury via miR-181a-5p/PPARα pathway.
Collapse
Affiliation(s)
- Jiqin Wang
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Jianfeng Song
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Yanyan Li
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Jinyan Shao
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Zichen Xie
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Keyu Sun
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201199, China.
| |
Collapse
|
110
|
Yan Y, Song D, Song X, Song C. The role of lncRNA MALAT1 in cardiovascular disease. IUBMB Life 2019; 72:334-342. [PMID: 31856403 DOI: 10.1002/iub.2210] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/24/2019] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is the first leading cause of death worldwide. Understanding the molecular mechanism of signaling pathways involved in pathology of CVD is benefit for targeted therapeutics. Recently, long non-coding RNAs (lncRNAs) are found and involved in regulation of pathology of CVD at different levels. Among them, MALAT1 attracted more attention as it was profoundly expressed in endothelial cells or cardiomyocytes in response to the risk factors of CVD, such as hypoxia, high glucose, cytokine, and oxidative stress. In this review, we summarize recent progresses in research on the molecular mechanism of MALAT1 on regulating the pathophysiological processes of CVD as well as its potential therapeutic applications.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Xianjing Song
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chunli Song
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
111
|
MALAT1: a therapeutic candidate for a broad spectrum of vascular and cardiorenal complications. Hypertens Res 2019; 43:372-379. [PMID: 31853043 DOI: 10.1038/s41440-019-0378-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 01/26/2023]
Abstract
Cardiovascular and renal complications cover a wide array of diseases. The most commonly known overlapping complications include cardiac and renal fibrosis, cardiomyopathy, cardiac hypertrophy, hypertension, and cardiorenal failure. The known or reported causes for the abovementioned complications include injury, ischemia, infection, and metabolic stress. To date, various targets have been reported and investigated in detail that are considered to be the cause of these complications. In the past 5 years, the role of noncoding RNAs has emerged in the area of cardiovascular and renal research, especially in relation to metabolic stress. The long noncoding RNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) has shown immense promise among the long noncoding RNA targets for treating cardiorenal complications. In this review, we shed light on the role of MALAT1 as a primary and novel target in treating cardiovascular and renal diseases as a whole.
Collapse
|
112
|
Xin M, Liang H, Wang H, Wen D, Wang L, Zhao L, Sun M, Wang J. Mirt2 functions in synergy with miR-377 to participate in inflammatory pathophysiology of Sjögren's syndrome. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2019; 47:2473-2480. [PMID: 31198060 DOI: 10.1080/21691401.2019.1626413] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 01/08/2023]
Abstract
Background: The interaction of long non-coding RNAs (lncRNAs)-microRNAs (miRs) exerts crucial functions in mediating inflammatory reaction. It is still unclear whether myocardial infarction associated transcript 2 (Mirt2)-miR-377 mediates the inflammatory pathogenesis in Sjögren's syndrome (SS). Methods: The inflammatory lesion model was established by stimulating salivary gland epithelial cells (SGECs) by interferon gamma (IFN-γ). Mirt2- and/or miR-377-transfected SGECs, as well as their negative controls, were applied to investigate the biological functions in inflammation. Cell viability and apoptosis were examined using commercial kits. Western blot was applied to quantify protein level, and enzyme-linked immuno sorbent assay (ELISA) was used to value the secretion of cytokines. Results: The up-regulation of Mirt2 was observed in IFN-γ-treated SGECs. Mirt2 overexpression restored the expression of miR-377 which was repressed by IFN-γ. However, miR-377 silence abolished the protective effect on cell viability, inhibitory effect on apoptosis and prohibitive role in pro-inflammatory factors. Mirt2 diminished the phosphorylated expression of crucial regulators while miR-377 silence restored the phosphorylation in IFN-γ-treated SGECs. Conclusion: Mirt2 was elevated in IFN-γ-treated SGECs and then up-regulated miR-377 in response to inflammatory lesions. Mechanically, in synergy with miR-377 Mirt2 blocked IFN-γ-evoked activation of NF-κB and JAK/STAT signalling pathway.
Collapse
Affiliation(s)
- Miaomiao Xin
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Hongda Liang
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Hongyue Wang
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Dawei Wen
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Liqin Wang
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Lei Zhao
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Mingshu Sun
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| | - Jibo Wang
- a Department of Rheumatology and Immunology, The Affiliated Hospital of Qingdao University , Shandong , China
| |
Collapse
|
113
|
Wang X, Gao S, Dai L, Wang Z, Wu H. Identification of key microRNAs in the carotid arteries of ApoE -/- mice exposed to disturbed flow. Hereditas 2019; 156:35. [PMID: 31719822 PMCID: PMC6833270 DOI: 10.1186/s41065-019-0112-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/23/2019] [Indexed: 11/24/2022] Open
Abstract
Background Atherosclerosis (AS) is one of the main causes of cardiovascular disease. AS plaques often occur in blood vessels with oscillatory blood flow and their formation can be regulated by microRNAs (miRNAs). The aim of this study is to identify the key miRNAs and molecular pathways involved in this pathological process. Methods In this study, gene chip data obtained from the GEO database was analyzed using the LIMMA package to find differentially expressed miRNAs (DE miRNAs) in the carotid arteries of ApoE−/− mice exposed to different blood flow rates. Predicted targets of the DE miRNAs were identified using the TargetScan, miRDB, and DIANA databases respectively, and the potential target genes (PTGs) were found by analyzing the common results of three databases. The DAVID database was used to enrich the PTGs based on gene ontology (GO) and pathway (Kyoto Encyclopedia of Genes and Genomes, KEGG), and the STRING database was used to uncover any protein-protein interactions (PPI) of the PTGs. Results The networks of the DE miRNAs-PTGs, Pathway-PTGs-DE miRNAs, and PTGs PPI, were constructed using Cytoscape, and 11 up-regulated and 13 down-regulated DE miRNAs and 1479 PTGs were found. GO results showed that PTGs were significantly enriched in functions such as transcriptional regulation and DNA binding. KEGG results showed that PTGs were significantly enriched in inflammation-related mitogen-activated protein kinase (MAPK) and AS-related FOXO pathways. The PPI network revealed some key target genes in the PTGs. Conclusions The analysis of key miRNAs and molecular pathways that regulate the formation of AS plaques induced by oscillatory blood flow will provide new ideas for AS treatment.
Collapse
Affiliation(s)
- Xinzhou Wang
- 1Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002 Henan China
| | - Shuibo Gao
- 1Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002 Henan China
| | - Liping Dai
- 2School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046 China
| | - Zhentao Wang
- 3Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, 450002 China
| | - Hong Wu
- 1Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002 Henan China.,3Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, 450002 China
| |
Collapse
|
114
|
Zong D, Liu X, Li J, Ouyang R, Chen P. The role of cigarette smoke-induced epigenetic alterations in inflammation. Epigenetics Chromatin 2019; 12:65. [PMID: 31711545 PMCID: PMC6844059 DOI: 10.1186/s13072-019-0311-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Background Exposure to cigarette smoke (CS) is a major threat to human health worldwide. It is well established that smoking increases the risk of respiratory diseases, cardiovascular diseases and different forms of cancer, including lung, liver, and colon. CS-triggered inflammation is considered to play a central role in various pathologies by a mechanism that stimulates the release of pro-inflammatory cytokines. During this process, epigenetic alterations are known to play important roles in the specificity and duration of gene transcription. Main text Epigenetic alterations include three major modifications: DNA modifications via methylation; various posttranslational modifications of histones, namely, methylation, acetylation, phosphorylation, and ubiquitination; and non-coding RNA sequences. These modifications work in concert to regulate gene transcription in a heritable fashion. The enzymes that regulate these epigenetic modifications can be activated by smoking, which further mediates the expression of multiple inflammatory genes. In this review, we summarize the current knowledge on the epigenetic alterations triggered by CS and assess how such alterations may affect smoking-mediated inflammatory responses. Conclusion The recognition of the molecular mechanisms of the epigenetic changes in abnormal inflammation is expected to contribute to the understanding of the pathophysiology of CS-related diseases such that novel epigenetic therapies may be identified in the near future.
Collapse
Affiliation(s)
- Dandan Zong
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Jinhua Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Ruoyun Ouyang
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
115
|
Xiong W, Qu Y, Chen H, Qian J. Insight into long noncoding RNA-miRNA-mRNA axes in myocardial ischemia-reperfusion injury: the implications for mechanism and therapy. Epigenomics 2019; 11:1733-1748. [PMID: 31701757 DOI: 10.2217/epi-2019-0119] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence has demonstrated that regulatory noncoding RNAs (ncRNAs), such as long noncoding RNAs (lncRNAs) and miRNAs, play crucial roles in the initiation and progress of myocardial ischemia-reperfusion injury (MIRI), which is associated with autophagy, apoptosis and necrosis of cardiomyocytes, as well as oxidative stress, inflammation and mitochondrial dysfunction. LncRNAs serve as a precursor or host of miRNAs and directly/indirectly affecting miRNAs via competitive binding or sponge effects. Simultaneously, miRNAs post-transcriptionally regulate the expression of genes by targeting various mRNA sequences due to their imperfect pairing with mRNAs. This review summarizes the potential regulatory role of lncRNA-miRNA-mRNA axes in MIRI and related molecular mechanisms of cardiac disorders, also provides insight into the potential therapies for MIRI-induced diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| | - Yan Qu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China.,Department of Anesthesiology, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan province 650021, PR China
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Angel Women's & Children's Hospital, Kunming, Yunnan province 650108, PR China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| |
Collapse
|
116
|
Zhu X. MiR-125b but not miR-125a is upregulated and exhibits a trend to correlate with enhanced disease severity, inflammation, and increased mortality in sepsis patients. J Clin Lab Anal 2019; 34:e23094. [PMID: 31696556 PMCID: PMC7083454 DOI: 10.1002/jcla.23094] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022] Open
Abstract
Objective This study aimed to investigate the correlation of miR‐125a/b expressions with disease risk, progression, and prognosis of sepsis. Methods MiR‐125a/b expressions and inflammatory cytokines were detected by RT‐qPCR and ELISA assays in plasma samples from 120 sepsis patients. Besides, blood biochemical indexes, disease severity scores, and in‐hospital mortality of sepsis patients were recorded. Meanwhile, miR‐125a/b expressions in plasma from 120 health controls (HCs) were also detected by RT‐qPCR. Results MiR‐125b was elevated in sepsis patients compared with HCs, and ROC curve revealed that miR‐125b could well distinguish sepsis patients from HCs with AUC 0.658. MiR‐125b positively correlated with APACHE II score, SOFA score, Scr, CRP, PCT, TNF‐α, and IL‐6 levels. Most interestingly, miR‐125b was greatly decreased in survivors compared with non‐survivors, and multivariate analysis revealed that miR‐125b independently predicted higher mortality risk in sepsis patients. Besides, miR‐125a showed no significant correlation with sepsis risk, disease severity, or prognosis. Conclusion MiR‐125b but not miR‐125a is upregulated and exhibits a trend to correlate with enhanced disease severity, inflammation, and increased mortality in sepsis patients.
Collapse
Affiliation(s)
- Xiaoping Zhu
- Departments of Anesthesia, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
117
|
Yong H, Wu G, Chen J, Liu X, Bai Y, Tang N, Liu L, Wei J. lncRNA MALAT1 Accelerates Skeletal Muscle Cell Apoptosis and Inflammatory Response in Sepsis by Decreasing BRCA1 Expression by Recruiting EZH2. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:97-108. [PMID: 31830649 PMCID: PMC6926165 DOI: 10.1016/j.omtn.2019.10.028] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022]
Abstract
Sepsis is a serious and elusive syndrome caused by infection, which is accompanied by a high mortality worldwide. Recent evidence has documented the regulatory role of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) during the inflammatory process, the effects of which in the development of sepsis have become the focus of the current study. An in vivo mouse model and in vitro cell model of sepsis induced by lipopolysaccharide (LPS) were developed. High expression of lncRNA MALAT1 along with low expression of breast cancer susceptibility gene 1 (BRCA1) were identified in septic mice and human skeletal muscle cells of sepsis. Then, lncRNA MALAT1 expression was altered in vivo and in vitro to examine serum levels of inflammatory factors, as well as skeletal muscle cell apoptosis. lncRNA MALAT1 was noted to regulate the expression and export from the nucleus of BRCA1 by recruiting zeste homolog 2 (EZH2) in skeletal muscle cells of sepsis. Silencing lncRNA MALAT1 resulted in reduced serum levels of interleukin (IL)-6, IL-8, and tumor necrosis factor alpha (TNF-α), neutrophil migration, skeletal muscle cell apoptosis, and AKT-1 phosphorylation. Taken together, lncRNA MALAT1 interacting with EZH2 stimulated AKT-1 phosphorylation and decreased BRCA1 expression, consequently aggravating the progression of sepsis, highlighting a promising therapeutic option for sepsis.
Collapse
Affiliation(s)
- Hui Yong
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Gangming Wu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jingyuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xueru Liu
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Yiping Bai
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Ni Tang
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| | - Li Liu
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China.
| | - Jicheng Wei
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| |
Collapse
|
118
|
Chen K, Shi X, Jin Y, Wang F, Shen Q, Xu W. High lncRNA MEG3 expression is associated with high mortality rates in patients with sepsis and increased lipopolysaccharide-induced renal epithelial cell and cardiomyocyte apoptosis. Exp Ther Med 2019; 18:3943-3947. [PMID: 31641380 PMCID: PMC6796421 DOI: 10.3892/etm.2019.8049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/13/2019] [Indexed: 12/29/2022] Open
Abstract
Long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) has been reported to be a key component in cancer biology. Preliminary microarray data from a previous study indicated that lncRNA MEG3 expression was altered in plasma of patients with sepsis. The present study found that the plasma levels of lncRNA MEG3 were significantly higher in sepsis patients compared with healthy controls. Patients were subsequently divided into high and low lncRNA MEG3 expression groups according to the Youden's index. Patients from the high lncRNA MEG3 expression group exhibited a significantly higher mortality rate compared with those from the low lncRNA MEG3 expression group. According to treatment outcomes, patients were divided into survival and mortality groups. It was observed that the pre-therapy expression levels of lncRNA MEG3 were significantly higher in mortality group compared with the survival group. Cell experiments in vitro revealed that lncRNA MEG3 overexpression and silencing using siRNA promoted and inhibited renal epithelial cell and cardiomyocyte apoptosis induced by lipopolysaccharide, respectively. In conclusion, these data suggest that lncRNA MEG3 overexpression may be involved in sepsis, and the downregulation of lncRNA MEG3 may serve as a potential therapeutic target for sepsis.
Collapse
Affiliation(s)
- Kang Chen
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| | - Xiaojun Shi
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| | - Ye Jin
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| | - Feng Wang
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| | - Qing Shen
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| | - Weiming Xu
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201400, P.R. China
| |
Collapse
|
119
|
Mirna M, Paar V, Rezar R, Topf A, Eber M, Hoppe UC, Lichtenauer M, Jung C. MicroRNAs in Inflammatory Heart Diseases and Sepsis-Induced Cardiac Dysfunction: A Potential Scope for the Future? Cells 2019; 8:cells8111352. [PMID: 31671621 PMCID: PMC6912436 DOI: 10.3390/cells8111352] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 02/06/2023] Open
Abstract
Background: MicroRNAs (miRNAs) are small, single-stranded RNA sequences that regulate gene expression on a post-transcriptional level. In the last few decades, various trials have investigated the diagnostic and therapeutic potential of miRNAs in several disease entities. Here, we provide a review of the available evidence on miRNAs in inflammatory heart diseases (myocarditis, endocarditis, and pericarditis) and sepsis-induced cardiac dysfunction. Methods: Systematic database research using the PubMed and Medline databases was conducted between July and September 2019 using predefined search terms. The whole review was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results: In total, 131 studies were screened, 96 abstracts were read, and 69 studies were included in the review. Discussion: In the future, circulating miRNAs could serve as biomarkers for diagnosis and disease monitoring in the context of inflammatory heart diseases and sepsis-induced cardiac dysfunction. Considering the promising results of different animal models, certain miRNAs could also emerge as novel therapeutic approaches in this setting.
Collapse
Affiliation(s)
- Moritz Mirna
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Vera Paar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Richard Rezar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Albert Topf
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Miriam Eber
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Uta C Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
120
|
Zhu W, Men X. Retracted
: Negative feedback of NF‐κB signaling by long noncoding RNA MALAT1 controls lipopolysaccharide‐induced inflammation injury in human lung fibroblasts WI‐38. J Cell Biochem 2019; 121:1945-1952. [DOI: 10.1002/jcb.29429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Wei Zhu
- Department of Respiratory Medicine Jinan No. 4 People's Hospital Jinan Shandong China
| | - Xuelin Men
- Department of Respiratory Medicine Jinan No. 4 People's Hospital Jinan Shandong China
| |
Collapse
|
121
|
Chen M, Guan Y, Li A, Zhao YZ, Zhang L, Zhang L, Gong Y. LncRNA SOX2OT Mediates Mitochondrial Dysfunction in Septic Cardiomyopathy. DNA Cell Biol 2019; 38:1197-1206. [PMID: 31618067 DOI: 10.1089/dna.2019.4839] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Researches establish an indispensable role of mitochondrial dysfunction in septic cardiomyopathy. We aimed to investigate the effects of long noncoding RNA (LncRNA) SOX2 overlapping transcript (SOX2OT) on mitochondrial dysfunction in septic cardiomyopathy. We observed an obvious overexpression of SOX2OT in septic hearts and cardiomyocytes. Knockdown of SOX2OT in mice recovered the reduced cardiac function, and improved the mitochondrial membrane potential impaired by lipopolysaccharide (LPS). SOX2OT overexpressed mice showed the opposite situation. In parallel, knockdown of SOX2OT in cardiomyocytes restored the mitochondrial membrane potential, along with reduced mitochondrial reactive oxygen species production induced by LPS, while overexpression of SOX2OT reversed these effects. Mechanistically, SOX2OT could regulate mitochondrial dysfunction in septic cardiomyopathy via SOX2. In general, SOX2OT contributed to mitochondrial dysfunction progression via inhibiting SOX2 expression in septic cardiomyopathy, which may provide a new insight for treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
- Mengfei Chen
- Department of Emergency, People's Hospital of Ningxia Hui Autonomous Region (The First Affiliated Hospital of Northwest Minzu University), Ningxia, China
| | - Yan Guan
- Department of Emergency, People's Hospital of Ningxia Hui Autonomous Region (The First Affiliated Hospital of Northwest Minzu University), Ningxia, China
| | - Ao Li
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Ying-Zhu Zhao
- People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Ling Zhang
- Department of Emergency, People's Hospital of Ningxia Hui Autonomous Region (The First Affiliated Hospital of Northwest Minzu University), Ningxia, China
| | - Liang Zhang
- Department of Emergency, People's Hospital of Ningxia Hui Autonomous Region (The First Affiliated Hospital of Northwest Minzu University), Ningxia, China
| | - Yanxuan Gong
- Department of Geratology, Lanzhou University, Lanzhou, China.,Department of Geratology, Gansu Second Provincial Hospital, Lanzhou, China.,Xi'An International University, Xi'An, China
| |
Collapse
|
122
|
Wang Y, Xu Z, Yue D, Zeng Z, Yuan W, Xu K. Linkage of lncRNA CRNDE sponging miR-181a-5p with aggravated inflammation underlying sepsis. Innate Immun 2019; 26:152-161. [PMID: 31604377 PMCID: PMC7016407 DOI: 10.1177/1753425919880946] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This investigation was performed to verify whether lncRNA CRNDE sponging miR-181a-5p was involved with sepsis-relevant inflammatory dysfunctions. Aggregately 136 sepsis patients and 151 healthy people were recruited, and their fasting peripheral blood was gathered to detect expressions of CRNDE and miR-181a-5p. In addition, THP-1 cells were transfected with si-CRNDE, miR-181a-5p mimic, pcDNA3.1-TLR4 and si-TLR4, and then sepsis-specific inflammatory cytokines within the cells were quantified. The sponging relationships between CRNDE and miR-181a-5p, as well as between miR-181a-5p and TLR4, were ascertained by means of luciferase reporter gene assay. The experimental results revealed that over-expressed CRNDE and under-expressed miR-181a-5p were associated with shortened lifespan of sepsis patients. Mechanically, si-CRNDE-1 and miR-181a-5p mimic were able to reverse the promoting effects of LPS on production of NF-kB, TNF-α, IL-1β and IL-6 by THP-1 cells. Moreover, the expressional change of miR-181a-5p in THP-1 cells was in part owing to its being sponged by CRNDE. Lastly, TLR4, subjected to targeted modification of miR-181a-5p, was capable of disturbing the contribution of CRNDE and miR-181a-5p to THP-1 cells’ release of NF-kB, TNF-α, IL-1β and IL-6. Collectively, the CRNDE/miR-181a-5p/TLR4 axis seemed to have potential in modifying sepsis-related inflammatory pathogenesis, which offered a direction for sepsis diagnosis and treatment.
Collapse
Affiliation(s)
- Yijun Wang
- Department of Emergency Medicine, Chenzhou NO.1 People's Hospital, Chenzhou, Hunan Province, P. R. China
| | - Ziqiang Xu
- Department of Emergency Medicine, Chenzhou NO.1 People's Hospital, Chenzhou, Hunan Province, P. R. China
| | - Dongyou Yue
- Department of Emergency Medicine, Chenzhou NO.1 People's Hospital, Chenzhou, Hunan Province, P. R. China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Weijie Yuan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P. R. China
| | - Ke Xu
- Department of Critical Care Medicine, Chenzhou NO.1 People's Hospital, Chenzhou, Hunan Province, P. R. China
| |
Collapse
|
123
|
Hsa-miR-346 plays a role in the development of sepsis by downregulating SMAD3 expression and is negatively regulated by lncRNA MALAT1. Mol Cell Probes 2019; 47:101444. [DOI: 10.1016/j.mcp.2019.101444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/28/2022]
|
124
|
Chen J, Gu X, Zhou L, Wang S, Zhu L, Huang Y, Cao F. Long non-coding RNA-HOTAIR promotes the progression of sepsis by acting as a sponge of miR-211 to induce IL-6R expression. Exp Ther Med 2019; 18:3959-3967. [PMID: 31656541 PMCID: PMC6812472 DOI: 10.3892/etm.2019.8063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/29/2019] [Indexed: 12/18/2022] Open
Abstract
Sepsis remains the primary cause of death in intensive care units and multiple long non-coding RNAs (lncRNAs) have been demonstrated to be dysregulated in samples of patients with sepsis. However, whether lncRNA-HOTAIR is involved in the etiology of sepsis remains unclear. The aim of the present study was to investigate the role of HOTAIR in sepsis and to reveal the associated mechanisms. A bioinformatics analysis and dual-luciferase reporter assay was performed to evaluate the interaction between HOTAIR and miR-211, as well as miR-211 and IL-6R. An animal model of sepsis was established in mice via cecal ligation and puncture. Interferon (IFN)-γ, interleukin (IL)-6, IL-17, tumor necrosis factor (TNF)-α, IL-1β, IL-6 receptor (R), microRNA (miR)-211 and HOTAIR expression was measured using reverse transcription-quantitative PCR. Cellular proliferation and apoptosis of monocytes were assessed using cell counting kit-8 assay and flow cytometry, respectively. miR-211 was revealed to be targeted by HOTAIR and IL-6R. The expression of IFN-γ, IL-6, IL-17, TNF-α, IL-1β, IL-6R and HOTAIR was significantly upregulated in the septic mice, whereas miR-211 expression was downregulated. The overexpression of hox transcript antisense RNA (HOTAIR) and knockdown of miR-211 were associated with an increased expression of IFN-γ, IL-6, IL-17, TNF-α, IL-1β and IL-6R in monocytes, while the overexpression of miR-211 exhibited the opposite effect. HOTAIR overexpression and miR-211 knockdown significantly inhibited cellular proliferation and promoted monocyte apoptosis, whereas the overexpression of miR-211 exhibited the opposite effects in monocytes. Therefore, HOTAIR may promote the progression of sepsis by indirectly regulating the expression of IL-6R via miR-211.
Collapse
Affiliation(s)
- Jianan Chen
- Department of Emergency Intensive Care Unit, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Xingsheng Gu
- Department of Emergency, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Li Zhou
- Department of Emergency Intensive Care Unit, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shuguang Wang
- Department of Emergency Intensive Care Unit, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Limei Zhu
- Department of Trauma Orthopedics, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Yangneng Huang
- Department of Emergency, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Feng Cao
- Department of Emergency, Ningbo 6th Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
125
|
Geng F, Liu W, Yu L. Potential role of circulating long noncoding RNA MALAT1 in predicting disease risk, severity, and patients' survival in sepsis. J Clin Lab Anal 2019; 33:e22968. [PMID: 31301104 PMCID: PMC6805255 DOI: 10.1002/jcla.22968] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/29/2022] Open
Abstract
Background This study aimed to investigate the plasma long noncoding RNA metastasis‐associated lung adenocarcinoma transcript 1 (lncRNA MALAT1) expression with risk, severity, inflammation level, and prognosis in sepsis. Methods One hundred and ninety sepsis patients and 190 health controls (HCs) were consecutively recruited. Blood samples within 24 hours after admission of sepsis patients and those on enrollment of HCs were collected, and then, plasma was separated for lncRNA MALAT1 and miR‐125b expressions detections by RT‐qPCR. In sepsis patients, clinical data and 28‐day mortality were recorded, and plasma inflammatory cytokines expressions were detected by ELISA. Results Plasma lncRNA MALAT1 expression was elevated in sepsis patients than HCs (P < 0.001), and ROC curve disclosed that it had a good value in predicting sepsis risk with an area under curve (AUC) of 0.823 (95% CI: 0.783‐0.864). Additionally, lncRNA MALAT1 expression was positively correlated with Scr (P = 0.005), WBC (P = 0.017), CRP (P < 0.001), PCT (P < 0.001), TNF‐α (P < 0.001), IL‐8 (P < 0.001), IL‐17 (P = 0.001), APACHE II score (P < 0.001), and SOFA score (P < 0.001). LncRNA MALA1 expression was elevated in deaths compared with survivors (P < 0.001) and could predict the risk of 28‐day mortality with an AUC of 0.755 (95% CI: 0.682‐0.828). Accumulating survival was worse in patients with lncRNA MALAT1 high expression compared with patients who had lncRNA MALAT1 low expression (P < 0.001). Moreover, lncRNA MALAT1 expression was negatively correlated with miR‐125b level in both sepsis patients (P < 0.001) and HCs (P < 0.001). Conclusion LncRNA MALAT1 could be developed as a potential biomarker for facilitating diagnosis and management in sepsis patients.
Collapse
Affiliation(s)
- Feng Geng
- Department of ICU, The Central Hospital Of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liu
- Department of ICU, The Central Hospital Of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yu
- Department of ICU, The Central Hospital Of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
126
|
Feng LL, Xin WN, Tian XL. MALAT1 modulates miR-146's protection of microvascular endothelial cells against LPS-induced NF-κB activation and inflammatory injury. Innate Immun 2019; 25:433-443. [PMID: 31291804 PMCID: PMC6900645 DOI: 10.1177/1753425919861427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
To investigate the role of miR-146 and its possible relationship with MALAT1 in
LPS-induced inflammation in human microvascular endothelial cells (HMECs),
HMEC-1 cells were treated with LPS to construct an inflammatory injury cell
model, and the cell viability, TNF-α and IL-6 secretion and the expression
levels of VCAM-1, SELE and ICAM-1 were analysed as markers of inflammatory
injury. The regulation mechanisms of miR-146 interacted with MALAT1 and the
downstream NF-κB signalling were also verified by dual-luciferase assay and
knockdown technology. LPS significantly decreased the cell viability, increased
levels of VCAM-1, SELE and ICAM-1 and also up-regulated miR-146a/b, TNF-α and
IL-6 in a dose-dependent manner. Over-expression of miR-146a resulted in
down-regulation of TNF-α and IL-6, as well as VCAM-1, SELE and ICAM-1, while
inhibition of miR-146a led to opposite results. The dual-luciferase reporter
assay showed both miR-146a and miR-146b directly targeted and negatively
regulated the expression of MALAT1. Silencing of MALAT1 suppressed LPS-induced
NF-κB activation and TNF-α and IL-6 secretion, reducing the cell inflammatory
injury, but these changes were reversed after combined treatment with miR-146a
inhibitor. Taken together, we demonstrate that miR-146 protects HMECs against
inflammatory injury by inhibiting NF-κB activation. This process is modulated by
MALAT1.
Collapse
Affiliation(s)
- Lin-Lin Feng
- Department of Respiration, Liaocheng People's Hospital, PR China
| | - Wei-Na Xin
- Department of Respiration, Liaocheng People's Hospital, PR China
| | - Xiu-Li Tian
- Department of Respiration, Liaocheng People's Hospital, PR China
| |
Collapse
|
127
|
Wei L, Li J, Han Z, Chen Z, Zhang Q. Silencing of lncRNA MALAT1 Prevents Inflammatory Injury after Lung Transplant Ischemia-Reperfusion by Downregulation of IL-8 via p300. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:285-297. [PMID: 31604167 PMCID: PMC6796730 DOI: 10.1016/j.omtn.2019.05.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion injury is a common early complication after lung transplantation. It was reported that long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is involved in ischemia-reperfusion injury and regulates inflammation. This study aimed to explore the role of MALAT1 in inflammatory injury following lung transplant ischemia-reperfusion (LTIR). A LTIR rat model was successfully established, with the expression of MALAT1 and interleukin-8 (IL-8) in lung tissues detected. Then, in vitro loss- and gain-of-function investigations were conducted to evaluate the effect of MALAT1 on pulmonary epithelial cell apoptosis and IL-8 expression. The relationship among MALAT1, p300, and IL-8 was tested. Moreover, a sh-MALAT1-mediated model of LTIR was established in vivo to examine inflammatory injury and chemotaxis infiltration. Both IL-8 and MALAT1 were highly expressed in LTIR. MALAT1 interacted with p300 to regulate the IL-8 expression by recruiting p300. Importantly, silencing of MALAT1 inhibited the chemotaxis of neutrophils by downregulating IL-8 expression via binding to p300. Besides, MALAT1 silencing alleviated the inflammatory injury after LTIR by downregulating IL-8 and inhibiting infiltration and activation of neutrophils. Collectively, these results demonstrated that silencing of MALAT1 ameliorated the inflammatory injury after LTIR by inhibiting chemotaxis of neutrophils through p300-mediated downregulation of IL-8, providing clinical insight for LTIR injury.
Collapse
Affiliation(s)
- Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Jiwei Li
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China.
| | - Zhijun Han
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Zhong Chen
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Quan Zhang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| |
Collapse
|
128
|
Gao G, Cheng X, Wei Q, Chen W, Huang W. Long noncoding RNA MALAT‐1 inhibits apoptosis and matrix metabolism disorder in interleukin‐1β‐induced inflammation in articular chondrocytes via the JNK signaling pathway. J Cell Biochem 2019; 120:17167-17179. [PMID: 31111559 DOI: 10.1002/jcb.28977] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Gui‐Cheng Gao
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Xi‐Gao Cheng
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Qiang‐Qiang Wei
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Wei‐Cai Chen
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Wen‐Zhou Huang
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| |
Collapse
|
129
|
Zhang L, Wang L, Guo E, Qi Y. RETRACTED: Silence of lncRNA CHRF protects H9c2 cells against lipopolysaccharide-induced injury via up-regulating microRNA-221. Exp Mol Pathol 2019; 107:43-50. [PMID: 30695715 DOI: 10.1016/j.yexmp.2019.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 01/06/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Given the comments of Dr Elisabeth Bik regarding this article “… the Western blot bands in all 400+ papers are all very regularly spaced and have a smooth appearance in the shape of a dumbbell or tadpole, without any of the usual smudges or stains. All bands are placed on similar looking backgrounds, suggesting they were copy/pasted from other sources, or computer generated”, the journal requested the authors to provide the raw data. However, the authors were not able to fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pediatrics, Jining No.1 People's Hospital, Jining 272011, China
| | - Li Wang
- Department of Pediatrics, Jining No.1 People's Hospital, Jining 272011, China
| | - Enyu Guo
- Department of Pediatrics, Jining No.1 People's Hospital, Jining 272011, China
| | - Yuefeng Qi
- Department of Pediatrics, Jining No.1 People's Hospital, Jining 272011, China.
| |
Collapse
|
130
|
Chen JX, Xu X, Zhang S. Silence of long noncoding RNA NEAT1 exerts suppressive effects on immunity during sepsis by promoting microRNA-125-dependent MCEMP1 downregulation. IUBMB Life 2019; 71:956-968. [PMID: 30883005 DOI: 10.1002/iub.2033] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/01/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
Accumulating studies have recognized microRNAs (miRs) and long noncoding RNAs (lncRNAs) as important molecules involved in the mediation of various biological processes, including innate immunity. In this study, we investigated a novel noncoding RNA regulatory circuitry in the immunity during sepsis. A cecal ligation and puncture-induced sepsis mouse model was established to determine the expression of mast cell expression membrane protein 1 (MCEMP1). The RNA crosstalk among lncRNA nuclear enriched abundant transcript 1 (NEAT1), miR-125, and MCEMP1 was validated. Subsequently, the levels of lncRNA NEAT1, miR-125, and MCEMP1 in T lymphocytes isolated from sepsis mice were up- or downregulated by exogenous transfection in an attempt to investigate their effects on the release of inflammatory factors, the expression of immunoglobulins, the activity of T cell subsets and natural killer (NK) cells, as well as T lymphocyte apoptosis. In sepsis mice, MCEMP1 was highly expressed and verified to be a target gene of miR-125. RNA crosstalk experiment revealed that lncRNA NEAT1 directly inhibited miR-125 to upregulate MCEMP1. We also observed that elevation of miR-125, depletion of MCEMP1, or downregulation of lncRNA NEAT1 resulted in promoted T lymphocyte activity, immunoglobulin expression, and NK cell activity, and inhibited release of inflammatory factors and T lymphocyte apoptosis. Taken together, these findings provided evidence that the downregulation of lncRNA NEAT1 could promote miR-125 to exert an inhibitory effect on the immunity in septic mice by suppressing MCEMP1, highlighting a potential target for the treatment of sepsis. © 2019 IUBMB Life, 2019.
Collapse
Affiliation(s)
- Jian-Xin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China.,First Department of Gastroenterological Surgery, The Affiliated Hospital of Putian University, Putian, People's Republic of China
| | - Xiong Xu
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Sen Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
131
|
Cardiovascular inflammation: RNA takes the lead. J Mol Cell Cardiol 2019; 129:247-256. [PMID: 30880251 DOI: 10.1016/j.yjmcc.2019.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
Abstract
Inflammation has recently gained tremendous attention as a key contributor in several chronic diseases. While physiological inflammation is essential to counter a wide variety of damaging stimuli and to improve wound healing, dysregulated inflammation such as in the myocardium and vasculature can promote cardiovascular diseases. Given the high severity, prevalence, and economic burden of these diseases, understanding the factors involved in the regulation of physiological inflammation is essential. Like other complex biological phenomena, RNA-based processes are emerging as major regulators of inflammatory responses. Among such processes are cis-regulatory elements in the mRNA of inflammatory genes, noncoding RNAs directing the production or localization of inflammatory cytokines/chemokines, or pathogenic RNA driving inflammatory responses. In this review, we describe several specific RNA-based molecular mechanisms by which physiological inflammation pertaining to cardiovascular diseases is regulated. These include the role of AU-rich element-containing mRNAs, long non-coding RNAs, microRNAs, and viral RNAs.
Collapse
|
132
|
LncRNAs expression in adriamycin-induced rats reveals the potential role of LncRNAs contributing to chronic glomerulonephritis pathogenesis. Gene 2019; 687:90-98. [DOI: 10.1016/j.gene.2018.11.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/28/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022]
|
133
|
Yang F, Meng L, Han P, Chen D, Wang M, Jiang Y, Wu Y, Wu Y, Xing N. New therapy with XLQ
®
to suppress chronic prostatitis through its anti‐inflammatory and antioxidative activities. J Cell Physiol 2019; 234:17570-17577. [PMID: 30790289 DOI: 10.1002/jcp.28380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Feiya Yang
- Department of Urology National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing P. R. China
| | - Lingquan Meng
- Department of Urology Qianfoshan Hospital Shandong University Jinan P. R. China
- Department of Urology Beijing Chaoyang Hospital Capital Medical University Beijing P. R. China
| | - Panpan Han
- Department of Hematology Qilu Hospital Shandong University Jinan P. R. China
| | - Dexi Chen
- Department of Hepatology, Beijing You'an Hospital Capital Medical University Beijing P. R. China
- Department of Hepatology, Beijing Institute of Hepatology Beijing P. R. China
| | - Mingshuai Wang
- Department of Urology Beijing Chaoyang Hospital Capital Medical University Beijing P. R. China
| | - Yongguang Jiang
- Department of Urology Beijing Anzhen Hospital Capital Medical University Beijing P. R. China
| | - Yanqiao Wu
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Shijiazhuang Yiling Pharmaceutical Co., Ltd Shijiazhuang P. R. China
| | - Yiling Wu
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Shijiazhuang Yiling Pharmaceutical Co., Ltd Shijiazhuang P. R. China
| | - Nianzeng Xing
- Department of Urology National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing P. R. China
- Department of Urology Beijing Chaoyang Hospital Capital Medical University Beijing P. R. China
| |
Collapse
|
134
|
Shen J, Liu L, Zhang F, Gu J, Pan G. LncRNA TapSAKI promotes inflammation injury in HK-2 cells and urine derived sepsis-induced kidney injury. J Pharm Pharmacol 2019; 71:839-848. [PMID: 30666657 DOI: 10.1111/jphp.13049] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/10/2018] [Indexed: 12/14/2022]
Abstract
Abstract
Objective
To explore the possible mechanism of lncRNA TapSAKI in urine derived sepsis-induced kidney injury.
Materials and methods
In vivo urine-derived sepsis (US) rat model and in vitro LPS-induced HK-2 cells were established, and TapSAKI, miR-22, PTEN, TLR4 and p-p65 expressions were detected by qRT-PCR and western blot. RNA precipitation and RNA pull-down were performed to confirm the interaction between TapSAKI and miR-22.
Results
TapSAKI was up-regulated, miR-22 was down-regulated, PTEN, TLR4 and p-p65 expressions, and inflammatory factors TNF-α and IL-6 levels were up-regulated in kidney tissue of US rats and LPS-induced HK-2 cells. In addition, TapSAKI interacted with miR-22, and negatively modulate miR-22 expression. We also observed TapSAKI promoted PTEN expression, TLR4/NF-κB pathway related proteins TLR4 and p-p65, and apoptosis protein cleaved-caspase-3 through negatively regulating miR-22. Further experiments proved TapSAKI/miR-22/TLR4/NF-κB pathway could promote HK-2 cell apoptosis. Finally, in vivo experiments showed TapSAKI knockdown negatively regulated miR-22 and positively regulate PTEN, decreased renal function indicators blood urea nitrogen and serum creatinine, and reduced TNF-α and IL-6.
Conclusion
TapSAKI was elevated in urine derived sepsis-induced kidney injury, and promoted HK-2 cell apoptosis and inflammatory response through miR-22/PTEN/TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Jun Shen
- Department of Organ Transplantation, The Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Li Liu
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Facai Zhang
- Department of Urology, The Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jiang Gu
- Department of Urology, The Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Guanghui Pan
- Department of Organ Transplantation, The Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
135
|
Yuan D, Wang Q, Ding N, Du P, Peng L, Duan Z, Pan S. Retracted Article: LncRNA MALAT1 aggravates MPP-induced neuronal injury by regulating miR-212 in SH-SY5Y cells. RSC Adv 2019; 9:690-698. [PMID: 35517595 PMCID: PMC9059515 DOI: 10.1039/c8ra09260e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/13/2018] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative disease and its incidence is rising. Long noncoding RNAs (lncRNAs) have been reported to have essential roles in development of PD. LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is dysregulated in PD, while the role of MALAT1 and its mechanism in PD remain poorly understood. In this study, SH-SY5Y cells were exposed to 1-methyl-4-phenylpyridinium (MPP+) to induce a PD model in vitro. Then we explored the effect of MALAT1 on cell viability, apoptosis and inflammatory response as well as its interaction with miR-212 in MPP+-treated SH-SY5Y cells. The results showed that MALAT1 was up-regulated in MPP+-treated SH-SY5Y cells compared with that in the normal group. Overexpression of MALAT1 exacerbated MPP+-induced neuronal injury, uncovered by inhibition of cell viability and increase of cell apoptosis as well as inflammatory cytokine expressions in SH-SY5Y cells. However, knockdown of MALAT1 exerted the opposite effect in MPP+-treated SH-SY5Y cells. Moreover, MALAT1 was bound to miR-212 and negatively regulated the miR-212 level. Furthermore, addition of miR-212 ablated the regulatory effect of MALAT1 on MPP+-induced neuronal injury, as indicated by restoration of cell viability and lower apoptotic rate along with inflammatory cytokine levels in SH-SY5Y cells. Therefore, we concluded that MALAT1 exacerbated MPP+-induced neuronal injury through regulating cell viability, apoptosis and inflammatory cytokines by sponging miR-212, providing a novel theoretical foundation for application of MALAT1 in PD.
Collapse
Affiliation(s)
- Dahua Yuan
- Department of Neurology, Nanfang Hospital, Southern Medical University 1023-1063, Sha Tai Road, Baiyun District 510515 Guangzhou China +86-020-62787320
- Department of Neurology, The First People's Hospital of Foshan Foshan China
| | - Qun Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University 1023-1063, Sha Tai Road, Baiyun District 510515 Guangzhou China +86-020-62787320
| | - Nan Ding
- Department of Neurology, The First People's Hospital of Foshan Foshan China
| | - Pu Du
- Department of Neurology, The First People's Hospital of Foshan Foshan China
| | - Lingmei Peng
- Department of Neurology, The First People's Hospital of Foshan Foshan China
| | - Zhenpeng Duan
- Department of Neurology, The First People's Hospital of Foshan Foshan China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University 1023-1063, Sha Tai Road, Baiyun District 510515 Guangzhou China +86-020-62787320
| |
Collapse
|
136
|
Jia P, Wu N, Jia D, Sun Y. Downregulation of MALAT1 alleviates saturated fatty acid-induced myocardial inflammatory injury via the miR-26a/HMGB1/TLR4/NF-κB axis. Diabetes Metab Syndr Obes 2019; 12:655-665. [PMID: 31123414 PMCID: PMC6511247 DOI: 10.2147/dmso.s203151] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose: The increased level of saturated fatty acids (SFAs) is found in patients with diabetes, obesity, and other metabolic disorders. SFAs can induce lipotoxic damage to cardiomyocytes, but the mechanism is unclear. The long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) acts as a key regulator in palmitic acid (PA)-induced hepatic steatosis, but its role in PA-induced myocardial lipotoxic injury is still unknown. The aim of this study was to explore the role and underlying mechanism of MALAT1 in PA-induced myocardial lipotoxic injury. Methods: MALAT1 expression in PA-treated human cardiomyocytes (AC16 cells) was detected by RT-qPCR. The effect of MALAT1 on PA-induced myocardial injury was measured by Cell Counting Kit-8, lactate dehydrogenase (LDH), and creatine kinase-MB (CK-MB) assays. Apoptosis was detected by flow cytometry. The activities of cytokines and nuclear factor (NF)-κB were detected by enzyme-linked immunosorbent assay. The interaction between MALAT1 and miR-26a was evaluated by a luciferase reporter assay and RT-qPCR. The regulatory effects of MALAT1 on high mobility group box 1 (HMGB1) expression were evaluated by RT-qPCR and western blotting. Results: MALAT1 was significantly upregulated in cardiomyocytes after PA treatment. Knockdown of MALAT1 increased the viability of PA-treated cardiomyocytes, decreased apoptosis, and reduced the levels of LDH, CK-MB, TNF-α, and IL-1β. Moreover, we found that MALAT1 specifically binds to miR-26a and observed a reciprocal negative regulatory relationship between these factors. We further found that the downregulation of MALAT1 represses HMGB1 expression, thereby inhibiting the activation of the Toll-like receptor 4 (TLR4)/NF-κB-mediated inflammatory response. These repressive effects were rescued by an miR-26a inhibitor. Conclusion: We demonstrate that MALAT1 is induced by SFAs and its downregulation alleviates SFA-induced myocardial inflammatory injury via the miR-26a/HMGB1/TLR4/NF-κB axis. Our findings provide new insight into the mechanism underlying myocardial lipotoxic injury.
Collapse
Affiliation(s)
- Pengyu Jia
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People‘s Republic of China
| | - Nan Wu
- The Central Laboratory of the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People‘s Republic of China
| | - Dalin Jia
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People‘s Republic of China
| | - Yingxian Sun
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People‘s Republic of China
- Correspondence: Yingxian Sun; Dalin JiaDepartment of Cardiology, The First Affiliated Hospital of China Medical University, 155th North of Nanjing Street, Heping District, Shenyang 110001, Liaoning, People’s Republic of ChinaTel +86 248 328 2602; Tel +86 248 328 2602Fax +860 248 328 2602; +860 248 328 2688Email ;
| |
Collapse
|
137
|
Sun W, Shi Y, Wang Z, Zhang J, Cai H, Zhang J, Huang D. Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review). Int J Oncol 2018; 53:2343-2355. [PMID: 30272345 DOI: 10.3892/ijo.2018.4575] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/24/2018] [Indexed: 11/05/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) usually refer to non-coding RNA transcripts >200 nucleotides in length. In terms of the full genomic transcript, the proportion of lncRNAs far exceeds that of coding RNA. Initially, lncRNAs were considered to be the transcriptional noise of genes, but it has since been demonstrated that lncRNAs serve an important role in the regulation of cellular activities through interaction with DNA, RNA and protein. Numerous studies have demonstrated that various intricate signaling pathways are closely related to lncRNAs. Here, we focus on a large number of studies regarding the interaction of lncRNAs with important signaling pathways. It is comprehensively illustrated that lncRNAs regulate key metabolic components and regulatory factors of signaling pathways to affect the biological activities of tumor cells. Evidence suggests that the abnormal expression or mutation of lncRNAs in human tumor cells, and their interaction with signaling pathways, may provide a basis and potential target for the diagnosis and treatment of human cancers.
Collapse
Affiliation(s)
- Wei Sun
- Department of Postgraduates, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Ying Shi
- Department of Obstetrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhifei Wang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Jiye Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Hanhui Cai
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Jungang Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| | - Dongsheng Huang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
138
|
Wang Y, Song X, Li Z, Liu B. Long non-coding RNAs in coronary atherosclerosis. Life Sci 2018; 211:189-197. [PMID: 30195033 DOI: 10.1016/j.lfs.2018.08.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/26/2022]
Abstract
Coronary atherosclerosis (CAS), a leading cause of cardiovascular disease, is a major cause of death worldwide. CAS is a chronic disease in the aorta that can be caused by dyslipidemia, abnormal glucose metabolism, endothelial cell dysfunction, vascular smooth muscle cell (VSMC) or fibrous connective tissue hyperplasia, immune inflammatory reactions, and many other factors. The pathogenesis of CAS is not fully understood, as it is a complex lesion complicated by multiple factors. Damage-response theories have put forward endothelial cell (EC) injury as the initiating factor for CAS; the addition of lipid metabolism disorders may enhance monocyte adhesion, increase the proliferation and migration of fibroblasts and VSMCs, and accelerate the development of CAS. Furthermore, inflammatory and immune responses can create a vicious cycle of endothelial injury, which also plays key roles in the formation of CAS. Therefore, in order to elucidate the mechanisms controlling CAS, it is important to study the etiology of vascular cell dysfunction, abnormal energy and metabolism disorders, and immune and inflammatory reactions. Non-coding RNAs play regulatory roles in the pathogenesis of CAS, especially long non-coding RNAs (lncRNAs); lncRNAs have recently become a major focus for cardiovascular disease mechanisms, as they play numerous roles in the progression of CAS. Therefore, in this review, we discuss the role of lncRNAs in the pathogenesis of coronary CAS, and their role in the prevention and treatment of coronary CAS.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xianjing Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
139
|
Pan L, Liu D, Zhao L, Wang L, Xin M, Li X. Retracted
: Long noncoding RNA MALAT1 alleviates lipopolysaccharide‐induced inflammatory injury by upregulating microRNA‐19b in murine chondrogenic ATDC5 cells. J Cell Biochem 2018; 119:10165-10175. [DOI: 10.1002/jcb.27357] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Lin Pan
- Department of Rheumatology Qilu Hospital of Shandong University Jinan China
- Department of Rheumatology and Clinical Immunology The Affiliated Hospital of Qingdao University Qingdao China
| | - Deheng Liu
- Department of Hand and Foot Surgery Qilu Hospital of Shandong University (Qingdao) Qingdao China
| | - Lei Zhao
- Department of Rheumatology and Clinical Immunology The Affiliated Hospital of Qingdao University Qingdao China
| | - Liqin Wang
- Department of Rheumatology and Clinical Immunology The Affiliated Hospital of Qingdao University Qingdao China
| | - Miaomiao Xin
- Department of Rheumatology and Clinical Immunology The Affiliated Hospital of Qingdao University Qingdao China
| | - Xingfu Li
- Department of Rheumatology Qilu Hospital of Shandong University Jinan China
| |
Collapse
|
140
|
Lei L, Chen J, Huang J, Lu J, Pei S, Ding S, Kang L, Xiao R, Zeng Q. Functions and regulatory mechanisms of metastasis‐associated lung adenocarcinoma transcript 1. J Cell Physiol 2018; 234:134-151. [PMID: 30132842 DOI: 10.1002/jcp.26759] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Li Lei
- Department of Dermatology, Xiangya Hospital Central South University Changsha Hunan China
- Department of Hunan Key Laboratory of Skin Cancer and Psoriasis Xiangya Hospital, Central South University Changsha Hunan China
| | - Jing Chen
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Jinhua Huang
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Jianyun Lu
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Shiyao Pei
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Shu Ding
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Liyang Kang
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| | - Rong Xiao
- Department of Dermatology Second Xiangya Hospital, Central South University Changsha Hunan China
| | - Qinghai Zeng
- Department of Dermatology Third Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
141
|
Wang W, Lou C, Gao J, Zhang X, Du Y. LncRNA SNHG16 reverses the effects of miR-15a/16 on LPS-induced inflammatory pathway. Biomed Pharmacother 2018; 106:1661-1667. [PMID: 30119242 DOI: 10.1016/j.biopha.2018.07.105] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 11/18/2022] Open
Abstract
Nowadays, neonatal sepsis has gradually become a global problem for its high incidence and increasing mortality. Previous studies have reported that miR-15a and miR-16 are two important modulators in neonatal sepsis. However, the upstream molecular mechanism of miR-15a/16 cluster is still mysterious. This study aims to explore a lncRNA can bind with miR-15a/16 in neonatal sepsis. Microarray analysis helped us found top ten lncRNAs which were downregulated in neonatal sepsis serum. Among these ten lncRNAs, SNHG16 was uncovered to significantly downregulated both miR-15a and miR-16. According to the result of subcellular fractionation assay, SNHG16 was mainly located in the cytoplasm of RAW264.7 cell, indicating the potential ceRNA role of SNHG16. Mechanism investigations revealed that SNHG16 could act as a ceRNA to upregulate TLR4 which is the target mRNA of miR-15a/16 cluster. At last, rescue assays demonstrated that SNHG16 reversed the effects of miR-15a/16 on LPS-induced inflammatory pathway. In summary, SNHG16 can act as a ceRNA to modulate miR-15a/16 cluster, thereby affecting LSP-induced inflammatory pathway which was downregulated by miR-15a/16 cluster.
Collapse
Affiliation(s)
- Wenying Wang
- Department of Pediatrics, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China
| | - Chunyan Lou
- Department of Pediatrics, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China
| | - Jie Gao
- Department of Pediatrics, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China
| | - Xiaomin Zhang
- Pingdingshan University, Weilai Road, Xincheng District, Pingdingshan, 467000, China
| | - Yaowu Du
- Laboratory for Nanomedicine, School of Basic Medical Science, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
142
|
Chen J, Wan J, Ye J, Xia L, Lu N. Emerging role of lncRNAs in the normal and diseased intestinal barrier. Inflamm Res 2018; 67:757-764. [PMID: 30008030 DOI: 10.1007/s00011-018-1170-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE A significant effort has been made to understand the intestinal barrier, but the effective means to prevent, reduce, and restore intestinal mucosal damage remains unclear. Recently, a few of studies have explained the mechanism of the intestinal barrier in long noncoding RNAs (lncRNAs). This review aims to summarize recent views on the function of lncRNAs in the intestinal barrier and discuss the emerging role of lncRNAs in intestinal barrier diseases caused by inflammatory diseases. METHODS Observations led us to believe that lncRNAs participate in inflammatory responses, cell proliferation, and control microbial susceptibility. In view of these, lncRNAs have been proved to involve in the intestinal barrier. RESULTS lncRNAs directly or indirectly affect TJ mRNA translation and intestinal epithelial cells (IECs) paracellular permeability, as well as IECs proliferation and susceptibility to apoptosis, to modulate the function of the intestinal barrier. miRNAs play a pivotal role in this process. CONCLUSIONS lncRNAs have been shown to be fundamentally involved in intestinal mucosal regeneration, protection, and epithelial barrier function. It may emerge as new and potential factors to be evaluated in the intestinal barrier diseases caused by acute pancreatitis, inflammatory bowel diseases, and imbalance of intestinal flora.
Collapse
Affiliation(s)
- Jie Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jianhua Wan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jianfang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Liang Xia
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| |
Collapse
|
143
|
He X, Yan Q, Kuang G, Wang Y, Cao P, Ou C. Metastasis-associated lung adenocarcinoma transcript 1 regulates tumor progression: old wine in a new bottle. J Thorac Dis 2018; 10:S1088-S1091. [PMID: 29850189 PMCID: PMC5949406 DOI: 10.21037/jtd.2018.04.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/30/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Xiaoyun He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gaoyan Kuang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yixuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|