51
|
|
52
|
Jozawa H, Inoue-Yamauchi A, Arimura S, Yamanashi Y. Loss of C/EBPδ enhances apoptosis of intestinal epithelial cells and exacerbates experimental colitis in mice. Genes Cells 2019; 24:619-626. [PMID: 31233664 DOI: 10.1111/gtc.12711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/08/2019] [Accepted: 06/19/2019] [Indexed: 01/01/2023]
Abstract
Inflammatory bowel diseases (IBDs) are characterized by chronic inflammation involving intestinal tissue damage, which include ulcerative colitis and Crohn's disease as major entities. Accumulating evidence suggests that excessive apoptosis of intestinal epithelial cells (IECs) contributes to the development of IBD. It was recently reported that the transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) is involved in inflammation; however, its role in colitis remains unclear. Here, we found that C/EBPδ knockout mice showed enhanced susceptibility to dextran sodium sulfate (DSS)-induced colitis, a mouse model of IBD, which was associated with severe colonic inflammation and mucosal damage with increased IEC apoptosis. Additionally, DSS stimulation induced increased expression of pro-apoptotic BH3-only protein Bim in the colon of C/EBPδ knockout mice. Collectively, our findings demonstrate that C/EBPδ plays an essential role in suppressing DSS-induced colitis, likely by attenuating IEC apoptosis.
Collapse
Affiliation(s)
- Hiroki Jozawa
- Division of Genetics, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akane Inoue-Yamauchi
- Division of Genetics, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sumimasa Arimura
- Division of Genetics, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuji Yamanashi
- Division of Genetics, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
53
|
Chen G, Feng Y, Li X, Jiang Z, Bei B, Zhang L, Han Y, Li Y, Li N. Post-transcriptional Gene Regulation in Colitis Associated Cancer. Front Genet 2019; 10:585. [PMID: 31275360 PMCID: PMC6593052 DOI: 10.3389/fgene.2019.00585] [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: 02/06/2019] [Accepted: 06/04/2019] [Indexed: 01/07/2023] Open
Abstract
Colitis-associated cancer (CAC) has been linked to microRNA (miRNA) aberrant expression elicited by inflammation. In this study, we used the AOM/DSS-induced CAC mice model to explore the ectopic expression of miRNAs in the precancerous stage of CAC. As a result, we found that miR-31-5p, miR-223-3p, and let-7f-5p were dysregulated during the development of intestinal dysplasia. Subsequently, we first identified the role of these three miRNAs in CAC. Adenomatous polyposis coli (APC) was revealed as a new target of miR-223-3p, and solute carrier family 9- subfamily A-member 9 (SLC9A9) and APC membrane recruitment protein 3 (AMER3) were suggested as two new targets for let-7f-5p. For miR-31-5p, we proved that it can target LATS2 mRNA so as to modulate Hippo pathway in Caco2 cells. Second, to examine if targeting these three miRNAs would lead to CAC prevention, pedunculoside, a natural triterpene glycoside capable of rescuing the down-regulation of LATS2 and APC caused by either miR-31-5p or miR-223-3p overexpression, respectively, was used in the in vivo AOM/DSS-induced CAC model. The results showed that pedunculoside (25 mg/kg) substantially mitigated the damage to mice intestine caused by DSS/AOM. These results suggested that miRNAs-elicited post-transcriptional regulation is involved in the pathogenesis of CAC, and CAC can be prevented through targeting key miRNAs that are ectopically expressed in CAC.
Collapse
Affiliation(s)
- Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuan Feng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xuezheng Li
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Zhe Jiang
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Bei Bei
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Lin Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yueqing Han
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanwu Li
- Pi-Wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
54
|
Cao L, Jin H, Zheng Y, Mao Y, Fu Z, Li X, Dong L. DANCR-mediated microRNA-665 regulates proliferation and metastasis of cervical cancer through the ERK/SMAD pathway. Cancer Sci 2019; 110:913-925. [PMID: 30582654 PMCID: PMC6398927 DOI: 10.1111/cas.13921] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 12/23/2022] Open
Abstract
Emerging evidence has indicated that microRNAs (miRNAs) play an important role in cervical cancer (CC). However, the role of miRNA (miR)-665 in cervical cancer remains unclear. The aim of the present study was to investigate the potential functions of miR-665 in CC and to identify the underlying mechanisms of action. Herein, we show that miR-665 was downregulated in CC tissues and cell lines, which is negatively correlated with tumor size, distant metastasis, advanced TNM stage and poor prognosis. Functionally, miR-665 inhibited cell proliferation, migration and invasion and resistance of cisplatin for CC cells, as well as tumor growth. We validated that transforming growth factor beta receptor 1 (TGFBR1) was a direct target of miR-665 and mediated the ERK/SMAD pathway. In addition, we identified miR-665 as the competing endogenous RNA for long noncoding (lnc)-DANCR. These observations suggested that lnc-DANCR-mediated miR-665 downregulation regulates the malignant phenotype of CC cells by targeting TGFBR1 through the ERK/SMAD pathway, which may present a pathway for novel therapeutic stratagems for CC therapy.
Collapse
Affiliation(s)
- Liyan Cao
- Department of Radiation OncologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Haihong Jin
- Department of GynecologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Yue Zheng
- Department of GastroenterologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Yu Mao
- Department of Radiation OncologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Zhanzhao Fu
- Department of Radiation OncologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Xin Li
- Department of Radiation OncologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Lixin Dong
- Department of Radiation OncologyThe First Hospital of QinhuangdaoQinhuangdaoChina
| |
Collapse
|
55
|
Chen P, Li Y, Li L, Yu Q, Chao K, Zhou G, Qiu Y, Feng R, Huang S, He Y, Chen B, Chen M, Zeng Z, Zhang S. Circulating microRNA146b-5p is superior to C-reactive protein as a novel biomarker for monitoring inflammatory bowel disease. Aliment Pharmacol Ther 2019; 49:733-743. [PMID: 30734320 DOI: 10.1111/apt.15159] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/06/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Owing to the importance of early treatment, simple and reliable methods for monitoring inflammatory bowel disease (IBD) are needed. AIMS To determine whether circulating microRNAs are reliable biomarkers for IBD monitoring. METHODS Serum levels of 17 candidate microRNAs were measured by quantitative real-time polymerase chain reaction in a discovery cohort (n = 120). Differentially expressed serum microRNAs were further investigated in an independent training cohort (n = 341). Correlations between relative microRNA levels and disease activity were evaluated. A disease control group was included to investigate the specificity of microRNA. Logistical regression was used to construct a microRNA classifier to identify endoscopic activity. Its predictive value was explored in the validation cohort (n = 66) using the area under the receiver operating characteristic curve (AUC). RESULTS Serum microRNA146b-5p (miR-146b-5p) expression was 2.87- and 2.72-fold higher in patients with Crohn's disease and ulcerative colitis, respectively, than in healthy controls. Serum miR-146b-5p was significantly correlated with disease activity and was more specific than C-reactive protein (CRP). A classifier was built for Crohn's disease, ie P [Endoscopically active] = 11+e2.937-0.737(miR-146b-5p)-0.008PLT , with a greater AUC of 0.869 [0.764-0.940] than that for CRP (0.680 [0.554-0.790]) (P = 0.0043). CONCLUSIONS MiR-146b-5p may better reflect mucosal inflammation in IBD than CRP. The Crohn's disease classifier developed in this study may be valuable for identifying endoscopic activity in patients with Crohn's disease.
Collapse
Affiliation(s)
- Peng Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Ying Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Li Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qiao Yu
- Division of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Kang Chao
- Division of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Gaoshi Zhou
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yun Qiu
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Rui Feng
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Shanshan Huang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yao He
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Baili Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Minhu Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zhirong Zeng
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
56
|
Lin B, Feng DG, Xu J. microRNA-665 silencing improves cardiac function in rats with heart failure through activation of the cAMP signaling pathway. J Cell Physiol 2019; 234:13169-13181. [PMID: 30666648 DOI: 10.1002/jcp.27987] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022]
Abstract
Heart failure (HF) is a disease with high mortality and morbidity rate. Previous studies have shown that microRNAs (miRNAs) may be implicated in the pathogenesis of HF, potentially being able to improve the cardiac function in an HF rat model. The present study was designed to define the role of miR-665 in the cardiac function of the HF rats. Following the establishm;ent of the rat models of HF, the functional role miR-665 in HF was determined using an ectopic expression and knockdown experiments. The cardiac function was evaluated with the determination of ventricular mass index and hemodynamic parameters. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining was performed, with the apoptosis of cardiac cells detected in the process. The expression of miR-665, glucagon-like peptide 1 receptor (GLP1R), cyclic adenosine monophosphate (cAMP) signaling pathway-related, and apoptosis-related genes was examined. Enzyme-linked immunosorbent assay was conducted to determine the levels of inflammation-related genes. Initially, the upregulation of miR-665, downregulation of GLP1R, and inactivation of cAMP signaling pathway were observed in HF rats. GLP1R was a target of miR-665. Forced expression of miR-665 promoted cell apoptosis and inhibited GLP1R and the cAMP signaling pathway. In addition, miR-665 overexpression has been known to impair cardiac function, promote inflammatory response while elevating malondialdehyde and superoxide dismutase levels, and decreasing mitochondrial respiratory chain enzyme activities. Furthermore, we also observed that the effects of miR-665 inhibition had been reversed when the cAMP signaling pathway was also inhibited. This study demonstrates that miR-665 inhibition can stabilize the cardiac function of HF rats via the cAMP signaling pathway via upregulation of the GLP1R.
Collapse
Affiliation(s)
- Bin Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - De-Guang Feng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
57
|
Moein S, Vaghari-Tabari M, Qujeq D, Majidinia M, Nabavi SM, Yousefi B. MiRNAs and inflammatory bowel disease: An interesting new story. J Cell Physiol 2018; 234:3277-3293. [PMID: 30417350 DOI: 10.1002/jcp.27173] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 07/17/2018] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD), as a chronic and recurrent inflammatory disorder, is caused by a dysregulated and aberrant immune response to exposed environmental factors in genetically susceptible individuals. Despite huge efforts in determining the molecular pathogenesis of IBD, an increasing worldwide incidence of IBD has been reported. MicroRNAs (miRNAs) are a set of noncoding RNA molecules that are about 22 nucleotides long, and these molecules are involved in the regulation of the gene expression. By clarifying the important role of miRNAs in a number of diseases, their role was also considered in IBD; numerous studies have been performed on this topic. In this review, we attempt to summarize a number of studies and discuss some of the recent developments in the roles of miRNAs in the pathophysiology, diagnosis, and treatment of IBD.
Collapse
Affiliation(s)
- Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mostafa Vaghari-Tabari
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Irantab.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
58
|
Wang X, Cui X, Zhu C, Li M, Zhao J, Shen Z, Shan X, Wang L, Wu H, Shen Y, Ni Y, Zhang D, Zhou G. FKBP11 protects intestinal epithelial cells against inflammation‑induced apoptosis via the JNK‑caspase pathway in Crohn's disease. Mol Med Rep 2018; 18:4428-4438. [PMID: 30221722 PMCID: PMC6172375 DOI: 10.3892/mmr.2018.9485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress in intestinal epithelial cells (IECs) has an important role in the pathogenesis of Crohn's disease (CD). FK506 binding protein 11 (FKBP11), a member of the peptidyl‑prolyl cis‑trans isomerase family, is involved in the unfolded protein response (UPR) and is closely associated with inflammation. Previous bioinformatics analysis revealed a potential association between FKBP11 and human CD. Thus, the present study aimed to investigate the potential significance of FKBP11 in IEC homeostasis and CD. In the present study, increased expression of FKBP11 was detected in the intestinal inflammatory tissues of patients with CD. Furthermore, the results of the present study revealed that overexpression of FKBP11 was accompanied by increased expression levels of the ER stress marker 78 kDa glucose‑regulated protein in the colon tissues of a 2, 4, 6‑trinitrobenzenesulphonic acid‑induced mouse colitis model. Using interferon‑γ (IFN‑γ)/tumor necrosis factor‑α (TNF‑α)‑stimulated IECs as an ER stress and apoptosis cell model, the associated of FKBP11 with ER stress and apoptosis levels was confirmed in IECs. Overexpression of FKBP11 was revealed to significantly attenuate the elevated expression of pro‑apoptotic proteins (Bcl2 associated X apoptosis regulator, caspase‑12 and active caspase‑3), suppress the phosphorylation of c‑Jun N‑terminal kinase (JNK), and decrease apoptosis of IFN‑γ/TNF‑α stimulated IECs. Knockdown of FKBP11 by transfection with small interfering RNA further validated the aforementioned results. In conclusion, these results suggest that the UPR protein FKBP11 may protect IECs against IFN‑γ/TNF‑α induced apoptosis by inhibiting the ER stress‑associated JNK/caspase apoptotic pathway in CD.
Collapse
Affiliation(s)
- Xiaotong Wang
- Department of Hepatology and Gastroenterology, The Fifth's People's Hospital of Suzhou, Suzhou, Jiangsu 215000, P.R. China
| | - Xiaopeng Cui
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chuanwu Zhu
- Department of Hepatology and Gastroenterology, The Fifth's People's Hospital of Suzhou, Suzhou, Jiangsu 215000, P.R. China
| | - Ming Li
- Department of Hepatology and Gastroenterology, The Fifth's People's Hospital of Suzhou, Suzhou, Jiangsu 215000, P.R. China
| | - Juan Zhao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhongyi Shen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaohang Shan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Liang Wang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Han Wu
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yanting Shen
- Clinical Medical Research Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - You Ni
- Clinical Medical Research Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Dongmei Zhang
- Clinical Medical Research Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
59
|
Ma X, Long F, Yun Y, Dang J, Wei S, Zhang Q, Li J, Zhang H, Zhang W, Wang Z, Liu Q, Zou C. ORMDL3 and its implication in inflammatory disorders. Int J Rheum Dis 2018; 21:1154-1162. [PMID: 29879314 DOI: 10.1111/1756-185x.13324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A growing body of evidence has suggested the genetic association of ORMDL3 gene (ORMDL Sphingolipid Biosynthesis Regulator 3) polymorphisms with a diverse set of inflammatory disorders that include bronchial asthma, inflammatory bowel disease, ankylosing spondylitis and atherosclerosis. Gene functional investigations have revealed the particular relevance of ORMDL3 in endoplasmic reticulum stress, lipid metabolism and inflammatory reactions. Additionally, several reports have recently added a new dimension to our understanding of the modulation of ORMDL3 gene expression in inflammation. This mini-review summarizes the pertinent publications regarding the genetic association studies and mechanistic exploration of ORMDL3 in common inflammatory disorders.
Collapse
Affiliation(s)
- Xiaochun Ma
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China.,Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Feng Long
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Yan Yun
- Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Dang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China.,Department of Medical Genetics and Cell Biology, Ningxia Medical University, Yinchuan, China
| | - Shijun Wei
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Qian Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Jinzhang Li
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Haizhou Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Wenlong Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
60
|
Jadideslam G, Ansarin K, Sakhinia E, Alipour S, Pouremamali F, Khabbazi A. The MicroRNA-326: Autoimmune diseases, diagnostic biomarker, and therapeutic target. J Cell Physiol 2018; 233:9209-9222. [PMID: 30078204 DOI: 10.1002/jcp.26949] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are uniquely regulated in healthy, inflamed, activated, cancerous, or other cells and tissues of a pathological state. Many studies confirm that immune dysregulation and autoimmune diseases with inflammation are correlated with various miRNA expression changes in targeted tissues and cells in innate or adaptive immunity. In this review, we will explain the history and classification of epigenetic changes. Next, we will describe the role of miRNAs changes, especially mir-326 in autoimmunity, autoinflammatory, and other pathological conditions. A systematic search of MEDLINE, Embase, and Cochrane Library was presented for all related studies from 1899 to 2017 with restrictions in the English language. In recent years, researchers have concentrated on mostly those roles of miRNA that are correlated with the inflammatory and anti-inflammatory process. Latest studies have proposed a fundamental pathogenic role in cancers and autoinflammatory diseases. Studies have described the role of microRNAs in autoimmunity and autoinflammatory diseases, cancers, and so on. The miRNA-326 expression plays a significant role in autoimmune and other types of diseases.
Collapse
Affiliation(s)
- Golamreza Jadideslam
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Connective Tissue Diseases Research Center, Tabriz University of Medical Science, Iran.,Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ebrahim Sakhinia
- Connective Tissue Diseases Research Center, Tabriz University of Medical Science, Iran.,Department of Medical Genetics, Faculty of Medicine and Tabriz Genetic Analysis Centre (TGAC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahriar Alipour
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Connective Tissue Diseases Research Center, Tabriz University of Medical Science, Iran
| | - Farhad Pouremamali
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Khabbazi
- Connective Tissue Diseases Research Center, Tabriz University of Medical Science, Iran
| |
Collapse
|
61
|
Cellular Stress Responses and Gut Microbiota in Inflammatory Bowel Disease. Gastroenterol Res Pract 2018; 2018:7192646. [PMID: 30026758 PMCID: PMC6031203 DOI: 10.1155/2018/7192646] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Progresses in the past two decades have greatly expanded our understanding of inflammatory bowel disease (IBD), an incurable disease with multifaceted and challenging clinical manifestations. The pathogenesis of IBD involves multiple processes on the cellular level, which include the stress response signaling such as endoplasmic reticulum (ER) stress, oxidative stress, and hypoxia. Under physiological conditions, the stress responses play key roles in cell survival, mucosal barrier integrity, and immunomodulation. However, they can also cause energy depletion, trigger cell death and tissue injury, promote inflammatory response, and drive the progression of clinical disease. In recent years, gut microflora has emerged as an essential pathogenic factor and therapeutic target for IBD. Altered compositional and metabolic profiles of gut microbiota, termed dysbiosis, are associated with IBD. Recent studies, although limited, have shed light on how ER stress, oxidative stress, and hypoxic stress interact with gut microorganisms, a potential source of stress in the microenvironment of gastrointestinal tract. Our knowledge of cellular stress responses in intestinal homeostasis as well as their cross-talks with gut microbiome will further our understanding of the pathogenesis of inflammatory bowel disease and probably open avenues for new therapies.
Collapse
|
62
|
Zhou B, Guo W, Sun C, Zhang B, Zheng F. Linc00462 promotes pancreatic cancer invasiveness through the miR-665/TGFBR1-TGFBR2/SMAD2/3 pathway. Cell Death Dis 2018; 9:706. [PMID: 29899418 PMCID: PMC5999603 DOI: 10.1038/s41419-018-0724-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/16/2018] [Accepted: 05/21/2018] [Indexed: 12/31/2022]
Abstract
Emerging evidence has identified that long non-coding RNAs (lncRNAs) may play an important role in the pathogenesis of many cancers, pancreatic cancer (PC) included. However, the role of linc00462 in PC remains unclear. The aim of our present study was to investigate the potential functions of linc00462 in PC and to identify the underlying mechanisms of action. CCK8 assay, transwell assay, cell cycle assay, cell apoptosis assay, EdU assay, western blot assay, cell adhesion assay, HE staining, IF staining, ELISA assay, vivo growth and metastasis assay, and colony formation assay were performed. We demonstrated that OSM mediated up-regulation of linc00462 promoted cell proliferation by accelerating cell cycle process and inhibiting cell apoptosis and adhesion in vitro, enhanced cell migration and invasion by accelerating EMT process, promoted tumor growth and matastasis in vivo and was associated with large tumor size, poor tumor differentiation, TNM stage and distant metastasis in patients of PC. In addition, we demonstrated that linc00462 was a target of miR-665. Linc00462 overexpression enhanced the expression levels of TGFBR1 and TGFBR2, and thus activated the SMAD2/3 pathway in PC cells. In conclusion, linc00462/miR-665/TGFBR1/2 regulatory network may shed light on tumorigenesis in PC.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatopacreatobiliary, Affiliated Hospital of Qingdao University, 266003, Qingdao, China
| | - Weidong Guo
- Department of Hepatopacreatobiliary, Affiliated Hospital of Qingdao University, 266003, Qingdao, China
| | - Chuandong Sun
- Department of Hepatopacreatobiliary, Affiliated Hospital of Qingdao University, 266003, Qingdao, China
| | - Bingyuan Zhang
- Department of Hepatopacreatobiliary, Affiliated Hospital of Qingdao University, 266003, Qingdao, China
| | - Fang Zheng
- School of Integrative Medicine, Tianjin Traditional Medical University, 300193, Tianjin, China.
| |
Collapse
|
63
|
Junjappa RP, Patil P, Bhattarai KR, Kim HR, Chae HJ. IRE1α Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front Immunol 2018; 9:1289. [PMID: 29928282 PMCID: PMC5997832 DOI: 10.3389/fimmu.2018.01289] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) is the most prominent and evolutionarily conserved endoplasmic reticulum (ER) membrane protein. This transduces the signal of misfolded protein accumulation in the ER, named as ER stress, to the nucleus as “unfolded protein response (UPR).” The ER stress-mediated IRE1α signaling pathway arbitrates the yin and yang of cell life. IRE1α has been implicated in several physiological as well as pathological conditions, including immune disorders. Autoimmune diseases are caused by abnormal immune responses that develop due to genetic mutations and several environmental factors, including infections and chemicals. These factors dysregulate the cell immune reactions, such as cytokine secretion, antigen presentation, and autoantigen generation. However, the mechanisms involved, in which these factors induce the onset of autoimmune diseases, are remaining unknown. Considering that these environmental factors also induce the UPR, which is expected to have significant role in secretory cells and immune cells. The role of the major UPR molecule, IRE1α, in causing immune responses is well identified, but its role in inducing autoimmunity and the pathogenesis of autoimmune diseases has not been clearly elucidated. Hence, a better understanding of the role of IRE1α and its regulatory mechanisms in causing autoimmune diseases could help to identify and develop the appropriate therapeutic strategies. In this review, we mainly center the discussion on the molecular mechanisms of IRE1α in the pathophysiology of autoimmune diseases.
Collapse
Affiliation(s)
- Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Prakash Patil
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Kashi Raj Bhattarai
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Hyung-Ryong Kim
- Graduate School, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
64
|
The Role of Autophagy and Related MicroRNAs in Inflammatory Bowel Disease. Gastroenterol Res Pract 2018; 2018:7565076. [PMID: 30046303 PMCID: PMC6038472 DOI: 10.1155/2018/7565076] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/26/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022] Open
Abstract
Accumulating evidence demonstrates that microRNA- (miR-) mediated posttranscriptional regulation plays an important role in autophagy in inflammatory bowel disease (IBD), a disease that is difficult to manage clinically because of the associated chronic recurrent nonspecific inflammation. Research indicates that microRNAs regulate autophagy via different pathways, playing an important role in the IBD process and providing a new perspective for IBD research. Related studies have shown that miR-142-3p, miR-320, miR-192, and miR-122 target NOD2, an IBD-relevant autophagy gene, to modulate autophagy in IBD. miR-142-3p, miR-93, miR-106B, miR-30C, miR-130a, miR-346, and miR-20a regulate autophagy by targeting ATG16L1 through several different pathways. miR-196 can downregulate IRGM and suppress autophagy by inhibiting the accumulation of LC3II. During the endoplasmic reticulum stress response, miR-665, miR-375, and miR-150 modulate autophagy by regulating the unfolded protein response, which may play an important role in IBD intestinal fibrosis. Regarding autophagy-related pathways, miR-146b, miR-221-5p, miR-132, miR-223, miR-155, and miR-21 regulate NF-κB or mTOR signaling to induce or inhibit autophagy in intestinal cells by releasing anti- or proinflammatory factors, respectively.
Collapse
|
65
|
Fu Q, Yang F, Xiang T, Huai G, Yang X, Wei L, Yang H, Deng S. A novel microRNA signature predicts survival in liver hepatocellular carcinoma after hepatectomy. Sci Rep 2018; 8:7933. [PMID: 29785036 PMCID: PMC5962561 DOI: 10.1038/s41598-018-26374-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is the most common type of primary liver cancer. In the current study, genome-wide miRNA-Seq and mRNA profiles in 318 LIHC patients derived from The Cancer Genome Atlas (TCGA) were analysed to identify miRNA-based signatures for LIHC prognosis with survival analysis and a semi-supervised principal components (SPC) method. A seven-miRNA signature was confirmed for overall survival (OS) prediction by comparing miRNA profiles in paired primary tumour and solid tumour normal tissues. Thereafter, a linear prognostic model that consisted of seven miRNAs was established and used to divide patients into high- and low-risk groups according to prognostic scores. Subsequent Kaplan-Meier analysis revealed that the seven-miRNA signature correlated with a good predictive clinical outcome for 5-year survival in LIHC patients. Additionally, this miRNA-based prognostic model could also be used for OS prognosis of LIHC patients in early stages, which could guide the future therapy of those patients and promote the OS rate. Moreover, the seven-miRNA signature was an independent prognostic factor. In conclusion, this signature may serve as a prognostic biomarker and guide LIHC therapy, and it could even be used as an LIHC therapeutic target in the future.
Collapse
Affiliation(s)
- Qiang Fu
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China
| | - Fan Yang
- Women and Children Health Care Center of Luoyang, Luoyang, 471000, Henan, China
| | - Tengxiao Xiang
- People's Hospital of Changshou Chongqing, Chongqing, 401220, China
| | - Guoli Huai
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China
| | - Xingxing Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China
| | - Liang Wei
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China
| | - Hongji Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China. .,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China.
| | - Shaoping Deng
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China. .,Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Chengdu, 610072, Sichuan, China. .,Human Islet Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, 02114, MA, USA.
| |
Collapse
|
66
|
Tan H, Zhao L, Song R, Liu Y, Wang L. microRNA-665 promotes the proliferation and matrix degradation of nucleus pulposus through targeting GDF5 in intervertebral disc degeneration. J Cell Biochem 2018; 119:7218-7225. [PMID: 29761869 DOI: 10.1002/jcb.26888] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/21/2018] [Indexed: 12/30/2022]
Abstract
Growing evidences suggested that microRNAs (miRNAs) played important roles in the development of intervertebral disc degeneration (IDD). However, the expression level and function of miR-665 in IDD remain unknown. In this study, we showed that the expression level of miR-665 was upregulated in degenerative human NP samples. In addition, miR-665 expression level gradually increased with the exacerbation of disc degeneration grade. Moreover, miR-665 expression level was positively associated with the Pfirrmann grade. Ectopic expression of miR-665 promoted NP cell growth. Furthermore, miR-665 overexpression decreased aggrecan and Col II expression and ectopic expression of miR-665 increased MMP-3 and MMP-13 expression in NP cell. We identified growth differentiation factor 5 (GDF5) was a direct target gene of miR-665 in NP cell and enforced expression of miR-665 decreased GDF5 expression. Elevated expression of miR-665 enhanced NP cell proliferation and decreased aggrecan and Col II expression. In addition, ectopic expression of miR-665 increased MMP-3 and MMP-13 expression through inhibiting GDF5 expression in NP cells. These results suggested that dysregulated miR-665 expression might act an important role in the development of IDD.
Collapse
Affiliation(s)
- Hongyu Tan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruipeng Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yilin Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
67
|
Li Z, Wang G, Feng D, Zu G, Li Y, Shi X, Zhao Y, Jing H, Ning S, Le W, Yao J, Tian X. Targeting the miR-665-3p-ATG4B-autophagy axis relieves inflammation and apoptosis in intestinal ischemia/reperfusion. Cell Death Dis 2018; 9:483. [PMID: 29706629 PMCID: PMC5924757 DOI: 10.1038/s41419-018-0518-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 02/08/2023]
Abstract
Autophagy is an essential cytoprotective response against pathologic stresses that selectively degrades damaged cellular components. Impaired autophagy contributes to organ injury in multiple diseases, including ischemia/reperfusion (I/R), but the exact mechanism by which impaired autophagy is regulated remains unclear. Several researchers have demonstrated that microRNAs (miRNAs) negatively regulate autophagy by targeting autophagy-related genes (ATGs). Therefore, the effect of ATG-related miRNAs on I/R remains a promising research avenue. In our study, we found that autophagy flux is impaired during intestinal I/R. A miRNA microarray analysis showed that miR-665-3p was highly expressed in the I/R group, which was confirmed by qRT-PCR. Then, we predicted and proved that miR-665-3p negatively regulates ATG4B expression in Caco-2 and IEC-6 cells. In ileum biopsy samples from patients with intestinal infarction, there was an inverse correlation between miR-665-3p and ATG4B expression, which supports the in vitro findings. Moreover, based on miR-665-3p regulation of autophagy in response to hypoxia/reoxygenation in vitro, gain-of-function and loss-of-function approaches were used to investigate the therapeutic potential of miR-665-3p. Additionally, we provide evidence that ATG4B is indispensable for protection upon inhibition of miR-665-3p. Finally, we observed that locked nucleic acid-modified inhibition of miR-665-3p in vivo alleviates I/R-induced systemic inflammation and apoptosis via recovery of autophagic flux. Our study highlights miR-665-3p as a novel small molecule that regulates autophagy by targeting ATG4B, suggesting that miR-665-3p inhibition may be a potential therapeutic approach against inflammation and apoptosis for the clinical treatment of intestinal I/R.
Collapse
Affiliation(s)
- Zhenlu Li
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Dongcheng Feng
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Guo Zu
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Yang Li
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Xue Shi
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Huirong Jing
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Shili Ning
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Weidong Le
- Clinical Research Center on Neurological Diseases, the First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China.
| | - Xiaofeng Tian
- Department of General Surgery, The Second Hospital of Dalian Medical University, 116023, Dalian, China.
| |
Collapse
|
68
|
Soroosh A, Koutsioumpa M, Pothoulakis C, Iliopoulos D. Functional role and therapeutic targeting of microRNAs in inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 2018; 314:G256-G262. [PMID: 29146677 PMCID: PMC5866423 DOI: 10.1152/ajpgi.00268.2017] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory gastrointestinal diseases, primarily consisting of ulcerative colitis and Crohn's disease. The complex nature of the disease, as well as the limited therapeutic options characterized by low efficiency and major side effects, highlights the importance of developing novel strategies of therapeutic intervention in IBD. Susceptibility loci related to IBD are present only in a small percentage of IBD patients, implying that epigenetic modifications could influence the pathogenesis of the disease. MicroRNAs (miRNAs) are small noncoding RNAs that regulate multiple molecular pathways involved in IBD pathobiology. MiRNA inhibitors targeting the IBD-activated miRNAs could have therapeutic value for IBD patients. This review provides an overview of the recent advances in miRNA biology related to IBD pathogenesis and the pharmacological development of miRNA-based therapeutics.
Collapse
Affiliation(s)
- Artin Soroosh
- 1Center for Systems Biomedicine, University of California at Los Angeles, Los Angeles, California
| | - Marina Koutsioumpa
- 1Center for Systems Biomedicine, University of California at Los Angeles, Los Angeles, California
| | - Charalabos Pothoulakis
- 2Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Dimitrios Iliopoulos
- 1Center for Systems Biomedicine, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
69
|
A decade of research on the 17q12-21 asthma locus: Piecing together the puzzle. J Allergy Clin Immunol 2018; 142:749-764.e3. [PMID: 29307657 PMCID: PMC6172038 DOI: 10.1016/j.jaci.2017.12.974] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 12/20/2022]
Abstract
Chromosome 17q12–21 remains the most highly replicated and significant asthma locus. Genotypes in the core region defined by the first genome-wide association study correlate with expression of 2 genes, ORM1-like 3 (ORMDL3) and gasdermin B (GSDMB), making these prime candidate asthma genes, although recent studies have implicated gasdermin A (GSDMA) distal to and post-GPI attachment to proteins 3 (PGAP3) proximal to the core region as independent loci. We review 10 years of studies on the 17q12–21 locus and suggest that genotype-specific risks for asthma at the proximal and distal loci are not specific to early-onset asthma and mediated by PGAP3, ORMDL3, and/or GSDMA expression. We propose that the weak and inconsistent associations of 17q single nucleotide polymorphisms with asthma in African Americans is due to the high frequency of some 17q alleles, the breakdown of linkage disequilibrium on African-derived chromosomes, and possibly different early-life asthma endotypes in these children. Finally, the inconsistent association between asthma and gene expression levels in blood or lung cells from older children and adults suggests that genotype effects may mediate asthma risk or protection during critical developmental windows and/or in response to relevant exposures in early life. Thus studies of young children and ethnically diverse populations are required to fully understand the relationship between genotype and asthma phenotype and the gene regulatory architecture at this locus. (J Allergy Clin Immunol 2018;142:749–64.)
Collapse
|
70
|
MicroRNAs in intestinal barrier function, inflammatory bowel disease and related cancers-their effects and therapeutic potentials. Curr Opin Pharmacol 2017; 37:142-150. [PMID: 29154194 DOI: 10.1016/j.coph.2017.10.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/17/2022]
Abstract
The initiation and development or inflammatory bowel disease (IBD) and associated colorectal cancers, have been linked to inflammation. MicroRNAs are non-coding regulators of gene expression that have gained great attention due to their capability to regulate the expression of a number of target transcripts. It is now generally admitted that microRNAs are instrumental in gut pathologies, in particular through their targeting of transcripts encoding proteins of the intestinal barrier (IB) and their regulators. Intense research is conducted to identify microRNAs susceptible to be used as biomarkers and to design new therapeutic approaches based upon using synthetic microRNA mimics and inhibitors as well as finding new drugs capable to restore or modify microRNA expression in the context of gut pathologies.
Collapse
|
71
|
MicroRNA in gastrointestinal cell signalling. Inflammopharmacology 2017; 26:1-14. [PMID: 29110118 DOI: 10.1007/s10787-017-0414-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/28/2017] [Indexed: 12/17/2022]
Abstract
Our gut forms an important organ and its formation, functioning and homeostasis are maintained by several factors including cell signalling pathways and commensal microflora. These factors affect pathological, physiological and immunological parameters to maintain gut health and prevent its inflammation. Among these, different intracellular signalling pathways play an important role in regulating gut homeostasis. These pathways are in turn regulated by various microRNAs that play a key role in maintaining the balance between tolerance and inflammation. This review highlights the importance of various cell signalling pathways in modulating gut homeostasis and the role specific miRNAs play in their regulation.
Collapse
|
72
|
Li K, Pan J, Wang J, Liu F, Wang L. MiR-665 regulates VSMCs proliferation via targeting FGF9 and MEF2D and modulating activities of Wnt/β-catenin signaling. Am J Transl Res 2017; 9:4402-4414. [PMID: 29118903 PMCID: PMC5666050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/01/2017] [Indexed: 06/07/2023]
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) contributes to the development of cardiovascular diseases. Studies have showed the great impact of microRNAs (miRNAs) on the cell proliferation in VSMCs. This study examined the in vitro functional roles of miR-665 in the VSMCs and explored the underlying molecular mechanisms. The mRNA and protein expression levels were determined by qRT-PCR and western blot assays, respectively. CCK-8, transwell invasion and wound healing assays were performed to measure VSMCs proliferation, invasion and migration, respectively. The miR-665 targeted-3'UTR of fibroblast growth factor 9 (FGF9) and myocyte enhancer factor 2D (MEF2D) was confirmed by luciferase reporter assay. Platelet-derived growth factor-bb (PDGF-bb) and 20% serum promoted cell proliferation and suppressed the expression of miR-665 in VSMCs. In vitro functional assays demonstrated that miR-665 inhibited VSMCs proliferation, invasion and migration. Bioinformatics analysis showed that FGF9 and MEF2D were found to be downstream targets of miR-665. Luciferase report assay confirmed that FGF9 and MEF2D 3'UTRs are direct targets of miR-665, and miR-665 overexpression suppressed both the mRNA and protein expression levels of FGF9 and MEF2D. Furthermore, rescue experiments showed that enforced expression of FGF9 or MEF2D attenuated the inhibitory effects of miR-665 on VSMCs proliferation. More importantly, overexpression of miR-665 also suppressed the mRNA and protein expression levels of β-catenin, c-myc and cyclin D1. In summary, miR-665 suppressed the VSMCs proliferation, invasion and migration via targeting FGF9 and MEF2D, and the in vitro effects of miR-665 on VSMCs may be associated with modulation of Wnt/β-catenin signaling activities.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The First Affiliated Hospital of Xi’an Medical UniversityXi’an, Shaanxi Province, China
| | - Jin Pan
- Clinical Medical College, Xi’an Medical UniversityXi’an, Shaanxi Province, China
| | - Jianjun Wang
- Department of Cardiology, The First Affiliated Hospital of Xi’an Medical UniversityXi’an, Shaanxi Province, China
| | - Fengrui Liu
- Department of Cardiology, The First Affiliated Hospital of Xi’an Medical UniversityXi’an, Shaanxi Province, China
| | - Li Wang
- Department of Cardiology, The First Affiliated Hospital of Xi’an Medical UniversityXi’an, Shaanxi Province, China
| |
Collapse
|
73
|
Zhu Y, Gu L, Li Y, Lin X, Shen H, Cui K, Chen L, Zhou F, Zhao Q, Zhang J, Zhong B, Prochownik E, Li Y. miR-148a inhibits colitis and colitis-associated tumorigenesis in mice. Cell Death Differ 2017; 24:2199-2209. [PMID: 28960206 DOI: 10.1038/cdd.2017.151] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022] Open
Abstract
miR-148a has been shown to regulate inflammation, immunity and the growth of certain tumors, but its roles in colitis and colorectal tumorigenesis remain largely undetermined. Here we found miR-148a-deficient mice to be more susceptible to colitis and colitis-associated tumorigenesis. Both were associated with increased nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) signaling. Bone marrow- and non-bone marrow-derived miR-148a contributed to colitis and colitis-associated tumorigenesis. miR-148a loss of heterozygosity exacerbated Apcmin/+ colon and small intestinal spontaneous tumor development. Restoring miR-148a expression prevented both spontaneous and carcinogen-induced colon tumor development. miR-148a was downregulated in human inflammatory bowel disease (IBD) and colorectal cancer patient tissues. This correlated with a high degree of miR-148a promoter methylation mediated by a complex comprised of P65 and DNA methyltransferase 3 alpha (DNMT3A). miR-148a directly targets several well-accepted upstream regulators of NF-κB and STAT3 signaling, including GP130, IKKα, IKKβ, IL1R1 and TNFR2, which leads to decreased NF-κB and STAT3 activation in macrophages and colon tissues. Our findings reveal that miR-148a is an indirect tumor suppressor that modulates colitis and colitis-associated tumorigenesis by suppressing the expression of signaling by NF-κB and STAT3 and their pro-inflammatory consequences.
Collapse
Affiliation(s)
- Yahui Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Li Gu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Yajun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xi Lin
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Hongxing Shen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Kaisa Cui
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Li Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Feng Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan 430071 China.,Hubei Clinical Center and Key Laboratory for Intestinal and Colorectal Diseases, Wuhan 430071, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan 430071 China.,Hubei Clinical Center and Key Laboratory for Intestinal and Colorectal Diseases, Wuhan 430071, China
| | - Jinxiang Zhang
- Department of Surgery, Wuhan Union Hospital, Wuhan 430022, China
| | - Bo Zhong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Edward Prochownik
- Division of Hematology/Oncology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA.,The Department of Microbiology and Molecular Genetics, The University of Pittsburgh Medical Center, Pittsburgh, PA 15224, USA
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|