151
|
Che X, Deng X, Xie K, Wang Q, Yan J, Shao X, Ni Z, Ying L. Long noncoding RNA MEG3 suppresses podocyte injury in diabetic nephropathy by inactivating Wnt/β-catenin signaling. PeerJ 2019; 7:e8016. [PMID: 31799068 PMCID: PMC6885352 DOI: 10.7717/peerj.8016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background Diabetic nephropathy (DN) is one of the principal complications of diabetes and podocyte injury plays an important role in the DN pathogenesis. Wnt/β-catenin signaling overactivation confers podocyte injury and promotes multiple types of renal disease. However, the underlying mechanism of Wnt/β-catenin signaling activation in DN progression has not been fully elucidated. Long noncoding RNA (lncRNA) is a large class of endogenous RNA molecules lacking functional code capacity and which participates in the pathogenesis of human disease, including DN. Method A diabetes model was constructed by intraperitoneal injection of Streptozotocin in rats. The MPC5 cells were used to create the in vitro model. Western blot and Quantitative reverse-transcriptase-PCR were used to examine the expression of protein and mRNA. The migrated capacity was analyzed by Transwell migration assay. The cell viability was detected by CCK8. Results In the present study, we revealed the association of lncRNA Maternally Expressed Gene 3 (MEG3) with aberrant activation of Wnt/β-catenin signaling and the role of MEG3/Wnt axis in podocyte injury. We found that high glucose (HG) treatment suppressed MEG3 expression in cultured podocytes, activated Wnt/β-catenin signaling and caused podocyte injury as indicated by the downregulation of podocyte-specific markers (podocin and synaptopodin) and the upregulation of snail1 and α-smooth muscle actin. Overexpression of MEG3 attenuated HG-induced podocyte injury by reducing Wnt/β-catenin activity, repressing cell migration, reactive oxygen species production and increasing the viability of podocytes. Furthermore, we provided evidences that restoration of Wnt/β-catenin signaling by specific agonist impeded the protective effect of MEG3 on podocyte injury. Current results demonstrated that MEG3/Wnt axis plays an important role in fostering podocyte injury and may serve as a potential therapeutic target for the treatment of DN. Conclusion lncRNA MEG3 ameliorates podocyte injury in DN via inactivating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xiajing Che
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xin Deng
- Department of Nephrology, Changshu NO. 1 People Hospital, Jiangsu, China
| | - Kewei Xie
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Qin Wang
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Jiayi Yan
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xinghua Shao
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Liang Ying
- Department of Urology, RenJi Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
152
|
Bai L, Li J, Li H, Song J, Zhou Y, Lu R, Liu B, Pang Y, Zhang P, Chen J, Liu X, Wu J, Liang C, Zhou J. Renoprotective effects of artemisinin and hydroxychloroquine combination therapy on IgA nephropathy via suppressing NF-κB signaling and NLRP3 inflammasome activation by exosomes in rats. Biochem Pharmacol 2019; 169:113619. [DOI: 10.1016/j.bcp.2019.08.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022]
|
153
|
Majumder S, Hadden MJ, Thieme K, Batchu SN, Niveditha D, Chowdhury S, Yerra VG, Advani SL, Bowskill BB, Liu Y, Vakili H, Alghamdi TA, White KE, Geldenhuys L, Siddiqi FS, Advani A. Dysregulated expression but redundant function of the long non-coding RNA HOTAIR in diabetic kidney disease. Diabetologia 2019; 62:2129-2142. [PMID: 31399844 DOI: 10.1007/s00125-019-4967-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/21/2019] [Indexed: 01/21/2023]
Abstract
AIMS/HYPOTHESIS Long non-coding RNAs (lncRNAs) are garnering increasing attention for their putative roles in the pathogenesis of chronic diseases, including diabetic kidney disease (DKD). However, much about in vivo lncRNA functionality in the adult organism remains unclear. To better understand lncRNA regulation and function in DKD, we explored the effects of the modular scaffold lncRNA HOTAIR (HOX antisense intergenic RNA), which approximates chromatin modifying complexes to their target sites on the genome. METHODS Experiments were performed in human kidney tissue, in mice with streptozotocin-induced diabetes, the db/db mouse model of type 2 diabetes, podocyte-specific Hotair knockout mice and conditionally immortalised mouse podocytes. RESULTS HOTAIR was observed to be expressed by several kidney cell-types, including glomerular podocytes, in both human and mouse kidneys. However, knockout of Hotair from podocytes had almost no effect on kidney structure, function or ultrastructure. Glomerular HOTAIR expression was found to be increased in human DKD, in the kidneys of mice with streptozotocin-induced diabetes and in the kidneys of db/db mice. Likewise, exposure of cultured mouse podocytes to high glucose caused upregulation of Hotair expression, which occurred in a p65-dependent manner. Although HOTAIR expression was upregulated in DKD and in high glucose-exposed podocytes, its knockout did not alter the development of kidney damage in diabetic mice. Rather, in a bioinformatic analysis of human kidney tissue, HOTAIR expression closely paralleled the expression of its genic neighbour, HOXC11, which is important to developmental patterning but which has an uncertain role in the adult kidney. CONCLUSIONS/INTERPRETATION Many lncRNAs have been found to bind to the same chromatin modifying complexes. Thus, there is likely to exist sufficient redundancy in the system that the biological effects of dysregulated lncRNAs in kidney disease may often be inconsequential. The example of the archetypal scaffold lncRNA, HOTAIR, illustrates how lncRNA dysregulation may be a bystander in DKD without necessarily contributing to the pathogenesis of the condition. In the absence of in vivo validation, caution should be taken before ascribing major functional roles to single lncRNAs in the pathogenesis of chronic diseases.
Collapse
Affiliation(s)
- Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India
| | - Mitchell J Hadden
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Karina Thieme
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sri N Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Divya Niveditha
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India
| | - Shibasish Chowdhury
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India
| | - Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Hana Vakili
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tamadher A Alghamdi
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada
| | - Kathryn E White
- Electron Microscopy Research Services, Newcastle University, Newcastle upon Tyne, UK
| | | | - Ferhan S Siddiqi
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, 209 Victoria Street, Toronto, ON, M5C 1T8, Canada.
| |
Collapse
|
154
|
Wang WJ, Jiang X, Gao CC, Chen ZW. Salusin-α mitigates diabetic nephropathy via inhibition of the Akt/mTORC1/p70S6K signaling pathway in diabetic rats. Drug Chem Toxicol 2019; 45:283-290. [PMID: 31665937 DOI: 10.1080/01480545.2019.1683572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Salusin-α is a bioactive peptide that protects against atherosclerosis and hepatosteatosis. Serum salusin-α level is declined in patients suffering with renal insufficiency. However, it is still undefined whether salusin-α plays a role in diabetic nephropathy. The present study was designed to investigate the potential roles of salusin-α in diabetic renal disease. Herein, we demonstrated that the salusin-α levels in both plasma and kidney tissues from diabetic rats were obviously downregulated. Exogenous administration of salusin-α eliminated the typical characteristics of diabetic nephropathy. Salusin-α treatment decreased renal fibrosis, which was related with reduced epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells. Injection of salusin-α suppressed the production of reactive oxygen species (ROS) via attenuation of NADPH oxidase subunits protein expressions and recovery of the antioxidant system. Mechanistically, the activated Akt/mTORC1/p70S6K signaling pathway in diabetic nephropathy was counteracted by salusin-α treatment. Our results demonstrated that salusin-α exerted protective effect against diabetic nephropathy via reduced oxidative stress and fibrosis, dependent on inactivation of the Akt/mTORC1/p70S6K signaling cascade. Salusin-α may be considered as a promising target for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, China
| | - Xia Jiang
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, China
| | - Chang-Chun Gao
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, China
| | - Zhi-Wei Chen
- Department of Nephrology, Center of Blood Purification, The Second People's Hospital of Nantong, Nantong, China
| |
Collapse
|
155
|
Guo FX, Wu Q, Li P, Zheng L, Ye S, Dai XY, Kang CM, Lu JB, Xu BM, Xu YJ, Xiao L, Lu ZF, Bai HL, Hu YW, Wang Q. The role of the LncRNA-FA2H-2-MLKL pathway in atherosclerosis by regulation of autophagy flux and inflammation through mTOR-dependent signaling. Cell Death Differ 2019; 26:1670-1687. [PMID: 30683918 PMCID: PMC6748100 DOI: 10.1038/s41418-018-0235-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 10/28/2018] [Accepted: 10/30/2018] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a progressive, chronic inflammation in arterial walls. Long noncoding RNAs (lncRNAs) participate in inflammation, but the exact mechanism in atherosclerosis is unclear. Our microarray analyses revealed that the levels of lncRNA-FA2H-2 were significantly decreased by oxidized low-density lipoprotein (OX-LDL). Bioinformatics analyses indicated that mixed lineage kinase domain-like protein (MLKL) might be regulated by lncRNA-FA2H-2. In vitro experiments showed that lncRNA-FA2H-2 interacted with the promoter of the MLKL gene, downregulated MLKL expression, and the binding sites between -750 and 471 were necessary for lncRNA-FA2H-2 responsiveness to MLKL. Silencing lncRNA-FA2H-2 and overexpression of MLKL could activate inflammation and inhibited autophagy flux. Both lncRNA-FA2H-2 knockdown and overexpression of MLKL could significantly aggravate inflammatory responses induced by OX-LDL. We found that the 3-methyladenine (3-MA) and Atg7-shRNA enhanced inflammatory responses induced by knockdown of lncRNA-FA2H-2 and overexpression of MLKL. We demonstrated that the effects of MLKL on autophagy might be associated with a mechanistic target of rapamycin (mTOR)-dependent signaling pathways. In vivo experiments with apoE knockout mice fed a western diet demonstrated that LncRNA-FA2H-2 knockdown decreased microtubule-associated expression of microtubule-associated protein 1 light chain 3 II and lysosome-associated membrane protein 1, but increased expression of sequestosome 1 (p62), MLKL, vascular cell adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-6 in atherosclerotic lesions. Our findings indicated that the lncRNA-FA2H-2-MLKL pathway is essential for regulation of autophagy and inflammation, and suggested that lncRNA-FA2H-2 and MLKL could act as potential therapeutic targets to ameliorate atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Feng-Xia Guo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Wu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pan Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Xiao-Yan Dai
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Chun-Min Kang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing-Bo Lu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bang-Ming Xu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan-Jun Xu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Xiao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Feng Lu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huan-Lan Bai
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
156
|
Sun F, Zhuang Y, Zhu H, Wu H, Li D, Zhan L, Yang W, Yuan Y, Xie Y, Yang S, Luo S, Jiang W, Zhang J, Pan Z, Lu Y. LncRNA PCFL promotes cardiac fibrosis via miR-378/GRB2 pathway following myocardial infarction. J Mol Cell Cardiol 2019; 133:188-198. [DOI: 10.1016/j.yjmcc.2019.06.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/30/2019] [Accepted: 06/15/2019] [Indexed: 12/13/2022]
|
157
|
Cheng J, Liu Q, Hu N, Zheng F, Zhang X, Ni Y, Liu J. Downregulation of hsa_circ_0068087 ameliorates TLR4/NF-κB/NLRP3 inflammasome-mediated inflammation and endothelial cell dysfunction in high glucose conditioned by sponging miR-197. Gene 2019; 709:1-7. [DOI: 10.1016/j.gene.2019.05.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 04/03/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
|
158
|
Li Y, Xie M, Men L, Du J. O-GlcNAcylation in immunity and inflammation: An intricate system (Review). Int J Mol Med 2019; 44:363-374. [PMID: 31198979 PMCID: PMC6605495 DOI: 10.3892/ijmm.2019.4238] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic, low‑grade inflammation associated with obesity and diabetes result from the infiltration of adipose and vascular tissue by immune cells and contributes to cardiovascular complications. Despite an incomplete understanding of the mechanistic underpinnings of immune cell differentiation and inflammation, O‑GlcNAcylation, the addition of O‑linked N‑acetylglucosamine (O‑GlcNAc) to cytoplasmic, nuclear and mitochondrial proteins by the two cycling enzymes, the O‑linked N‑acetylglucosamine transferase (OGT) and the O‑GlcNAcase (OGA), may contribute to fine‑tune immunity and inflammation in both physiological and pathological conditions. Early studies have indicated that O‑GlcNAcylation of proteins play a pro‑inflammatory role in diabetes and insulin resistance, whereas subsequent studies have demonstrated that this post‑translational modification could also be protective against acute injuries. These studies suggest that diverse types of insults result in dynamic changes to O‑GlcNAcylation patterns, which fluctuate with cellular metabolism to promote or inhibit inflammation. In this review, the current understanding of O‑GlcNAcylation and its adaptive modulation in immune and inflammatory responses is summarized.
Collapse
Affiliation(s)
- Yu Li
- Department of Endocrinology
| | - Mingzheng Xie
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | | | - Jianling Du
- Department of Endocrinology
- Correspondence to: Dr Jianling Du, Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning 116011, P.R. China, E-mail:
| |
Collapse
|
159
|
Chen W, Peng R, Sun Y, Liu H, Zhang L, Peng H, Zhang Z. The topological key lncRNA H2k2 from the ceRNA network promotes mesangial cell proliferation in diabetic nephropathyviathe miR‐449a/b/Trim11/Mek signaling pathway. FASEB J 2019; 33:11492-11506. [DOI: 10.1096/fj.201900522r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wenyun Chen
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| | - Rui Peng
- Department of BioinformaticsChongqing Medical UniversityChongqingChina
| | - Yan Sun
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| | - Handeng Liu
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| | - Luyu Zhang
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| | - Huimin Peng
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| | - Zheng Zhang
- Molecular Medicine and Cancer Research CenterChongqing Medical UniversityChongqingChina
| |
Collapse
|
160
|
Ge Y, Wang J, Wu D, Zhou Y, Qiu S, Chen J, Zhu X, Xiang X, Li H, Zhang D. lncRNA NR_038323 Suppresses Renal Fibrosis in Diabetic Nephropathy by Targeting the miR-324-3p/DUSP1 Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 17:741-753. [PMID: 31430717 PMCID: PMC6709345 DOI: 10.1016/j.omtn.2019.07.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/21/2019] [Accepted: 07/10/2019] [Indexed: 12/19/2022]
Abstract
Several studies have suggested that long intergenic noncoding RNAs are involved in the progression of diabetic nephropathy (DN). However, the exact role and regulatory mechanism of long noncoding RNA (lncRNA) NR_038323 in diabetic nephropathy (DN) remain largely unclear. In the present study, we found that lncRNA NR_038323 overexpression ameliorated the high glucose (HG)-induced expression levels of collagen I, collagen IV, and fibronectin, whereas lncRNA NR_038323 knockdown exerted the opposite effects. Moreover, the results of bioinformatic prediction, luciferase assay, and fluorescence in situ hybridization (FISH) demonstrated that lncRNA NR_038323 directly interacted with miR-324-3p. Additionally, miR-324-3p mimic aggravated the HG-induced expression levels of collagen I, collagen IV, and fibronectin by dual-specificity protein phosphatase-1 (DUSP1) expression to activate p38 mitogen-activated protein kinase (MAPK) and ERK1/2 pathways. In contrast, overexpression of DUSP1 attenuated the HG-induced expression levels of collagen I, collagen IV, and fibronectin via inactivation of p38 MAPK and ERK1/2 pathways. In addition, lncRNA NR_038323 knockdown increased the expression levels of collagen I, collagen IV, and fibronectin by upregulating DUSP1 expression during HG treatment, which were markedly reversed by miR-324-3p inhibitor. Furthermore, these molecular changes were verified in the human kidney samples of DN patients. Finally, overexpression of lncRNA NR_038323 ameliorated the interstitial fibrosis in STZ-induced diabetic nephrology (DN) rat via miR-324-3p/DUSP1/p38MAPK and ERK1/2 axis. In conclusion, our data indicate that overexpression of lncRNA NR_038323 may suppress HG-induced renal fibrosis via the miR-324-3p/DUSP1/p38MAPK and ERK1/2 axis, which provides new insights into the pathogenesis of DN.
Collapse
Affiliation(s)
- Yanni Ge
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Juan Wang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Dengke Wu
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yu Zhou
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Shuangfa Qiu
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Junxiang Chen
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xuejin Zhu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xudong Xiang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Huiling Li
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Dongshan Zhang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
161
|
Zhang P, Sun Y, Peng R, Chen W, Fu X, Zhang L, Peng H, Zhang Z. Long non-coding RNA Rpph1 promotes inflammation and proliferation of mesangial cells in diabetic nephropathy via an interaction with Gal-3. Cell Death Dis 2019; 10:526. [PMID: 31285427 PMCID: PMC6614467 DOI: 10.1038/s41419-019-1765-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/02/2019] [Accepted: 06/25/2019] [Indexed: 12/28/2022]
Abstract
Diabetic nephropathy (DN) is one of the most significant complications of diabetes and is the primary cause of end-stage kidney disease. Cumulating evidence has shown that renal inflammation plays a role in the development and progression of DN, but the exact cellular mechanisms are unclear. Irregular expression of long non-coding RNAs (lncRNAs) is present in many diseases, including DN. However, the relationship between lncRNAs and inflammation in DN is unclear. In this study, we identified differentially expressed lncRNAs in DN using RNA-sequencing. Among these lncRNAs, we identified seven DN-related lncRNAs in vivo and in vitro using quantitative real-time PCR. One lncRNA in particular, Rpph1 (ribonuclease P RNA component H1), exhibited significantly increased expression. Further, over-expression or knockdown of Rpph1 was found to regulate cell proliferation and the expression of inflammatory cytokines in mesangial cells (MCs). The results revealed that Rpph1 directly interacts with the DN-related factor galectin-3 (Gal-3). Further, over-expression of Rpph1 promoted inflammation and cell proliferation through the Gal-3/Mek/Erk signaling pathway in MCs under low glucose conditions, while knockdown of Rpph1 inhibited inflammation and cell proliferation through the Gal-3/Mek/Erk pathway in MCs under high glucose conditions. These results provide new insight into the association between Rpph1 and the Gal-3/Mek/Erk signaling pathway during DN progression.
Collapse
Affiliation(s)
- Panyang Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China
| | - Yan Sun
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China
| | - Rui Peng
- Department of Bioinformatics, Chongqing Medical University, 400016, Chongqing, China
| | - Wenyun Chen
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China
| | - Xia Fu
- People's Hospital of Fuling District, 408000, Chongqing, China
| | - Luyu Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China
| | - Huimin Peng
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China
| | - Zheng Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
162
|
Long Non-Coding RNAs in Kidney Disease. Int J Mol Sci 2019; 20:ijms20133276. [PMID: 31277300 PMCID: PMC6650856 DOI: 10.3390/ijms20133276] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 02/01/2023] Open
Abstract
Non-coding RNA species contribute more than 90% of all transcripts and have gained increasing attention in the last decade. One of the most recent members of this group are long non-coding RNAs (lncRNAs) which are characterized by a length of more than 200 nucleotides and a lack of coding potential. However, in contrast to this simple definition, lncRNAs are heterogenous regarding their molecular function—including the modulation of small RNA and protein function, guidance of epigenetic modifications and a role as enhancer RNAs. Furthermore, they show a highly tissue-specific expression pattern. These aspects already point towards an important role in cellular biology and imply lncRNAs as players in development, health and disease. This view has been confirmed by numerous publications from different fields in the last years and has raised the question as to whether lncRNAs may be future therapeutic targets in human disease. Here, we provide a concise overview of the current knowledge on lncRNAs in both glomerular and tubulointerstitial kidney disease.
Collapse
|
163
|
Yang Z, Jiang S, Shang J, Jiang Y, Dai Y, Xu B, Yu Y, Liang Z, Yang Y. LncRNA: Shedding light on mechanisms and opportunities in fibrosis and aging. Ageing Res Rev 2019; 52:17-31. [PMID: 30954650 DOI: 10.1016/j.arr.2019.04.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Fibrosis is universally observed in multiple aging-related diseases and progressions and is characterized by excess accumulation of the extracellular matrix. Fibrosis occurs in various human organs and eventually results in organ failure. Noncoding RNAs (ncRNAs) have emerged as essential regulators of cellular signaling and relevant human diseases. In particular, the enigmatic class of long noncoding RNAs (lncRNAs) is a kind of noncoding RNA that is longer than 200 nucleotides and does not possess protein coding ability. LncRNAs have been identified to exert both promotive and inhibitory effects on the multifaceted processes of fibrosis. A growing body of studies has revealed that lncRNAs are involved in fibrosis in various organs, including the liver, heart, lung, and kidney. As lncRNAs have been increasingly identified, they have become promising targets for anti-fibrosis therapies. This review systematically highlights the recent advances regarding the roles of lncRNAs in fibrosis and sheds light on the use of lncRNAs as a potential treatment for fibrosis.
Collapse
|
164
|
Long noncoding RNA: an emerging player in diabetes and diabetic kidney disease. Clin Sci (Lond) 2019; 133:1321-1339. [PMID: 31221822 DOI: 10.1042/cs20190372] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/16/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease (DKD) is among the most common complications of diabetes mellitus (DM), and remains the leading cause of end-stage renal diseases (ESRDs) in developed countries, with no definitive therapy yet available. It is imperative to decipher the exact mechanisms underlying DKD and identify novel therapeutic targets. Burgeoning evidence indicates that long non-coding RNAs (lncRNAs) are essential for diverse biological processes. However, their roles and the mechanisms of action remain to be defined in disease conditions like diabetes and DKD. The pathogenesis of DKD is twofold, so is the principle of treatments. As the underlying disease, diabetes per se is the root cause of DKD and thus a primary focus of therapy. Meanwhile, aberrant molecular signaling in kidney parenchymal cells and inflammatory cells may directly contribute to DKD. Evidence suggests that a number of lncRNAs are centrally involved in development and progression of DKD either via direct pathogenic roles or as indirect mediators of some nephropathic pathways, like TGF-β1, NF-κB, STAT3 and GSK-3β signaling. Some lncRNAs are thus likely to serve as biomarkers for early diagnosis or prognosis of DKD or as therapeutic targets for slowing progression or even inducing regression of established DKD. Here, we elaborated the latest evidence in support of lncRNAs as a key player in DKD. In an attempt to strengthen our understanding of the pathogenesis of DKD, and to envisage novel therapeutic strategies based on targeting lncRNAs, we also delineated the potential mechanisms of action as well as the efficacy of targeting lncRNA in preclinical models of DKD.
Collapse
|
165
|
Liu B, Lu R, Li H, Zhou Y, Zhang P, Bai L, Chen D, Chen J, Li J, Yu P, Wu J, Liang C, Song J, Liu X, Zhou J. Zhen-wu-tang ameliorates membranous nephropathy rats through inhibiting NF-κB pathway and NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152913. [PMID: 30991182 DOI: 10.1016/j.phymed.2019.152913] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/23/2019] [Accepted: 04/01/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Zhen-wu-tang (ZWT), a traditional herbal formula, has been widely used for the treatment of kidney diseases in clinics, but the underlying molecular mechanisms have not been fully understood. PURPOSE Inflammation mediated podocyte injury has been reported to constitute a crucial part in the pathogenesis of membranous nephropathy (MN). The current study was designed to evaluate the effect of ZWT on MN related to nuclear factor-κB (NF-κB) pathway and NLRP3 inflammasome. METHODS The main components of ZWT were identified by 3D-ultra performance liquid chromatography (3D-UPLC) assay. A MN rat model induced by cationic-bovine serum albumin (C-BSA) and podocytes stimulated by TNF-α were used in this study. The 24 h urine protein, serum total cholesterol (TC) and triglyceride (TG), as well as kidney histology were measured to evaluate kidney damage. The expressions of IgG and complement 3 (C3), and the co-localization of NLRP3 and ASC were detected by immunofluorescence. The expressions of podocyte injury related protein desmin, podocin were measured by immunohistochemistry and western blot. Cell vitality of cultured podocytes was detected by MTT assay, as apoptosis assay was measured via flow cytometry. The protein expressions of p-p65, p-IκBα, NLRP3, Caspase-1, IL-1β were detected by western blot. RESULTS Our results showed that ZWT significantly ameliorated kidney damage in MN model rats by decreasing the levels of 24 h urine protein, TC and TG. ZWT also improved renal histology and reduced the expressions of IgG and C3 in glomerulus. In addition, ZWT lessened the expressions of desmin, but increased podocin expression in vivo and vitro. ZWT protected cultured podocytes by maintaining cell vitality and inhibiting apoptosis. Moreover, we found that ZWT suppressed the expressions of NLRP3, Caspase-1, IL-1β and the co-localization of NLRP3 and ASC. Furthermore, the inhibition of NLRP3 inflammasome under ZWT treatment were accompanied by down-regulation of NF-κB pathway, as the p-p65 and p-IκBα protein expression were reduced. CONCLUSIONS Our present study indicates that the inhibition of NF-κB pathway and NLRP3 inflammasome might be the potential mechanisms for the therapeutic effects of ZWT against MN.
Collapse
Affiliation(s)
- Bihao Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Ruirui Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Honglian Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Yuan Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Peichun Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Lixia Bai
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Dandan Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Junqi Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Jicheng Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Pang Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China
| | - Junbiao Wu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Chunling Liang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Jianping Song
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510400, PR China
| | - Xusheng Liu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 232 WaiHuan East Road, Guangzhou University Town, Guangzhou 510006, PR China.
| |
Collapse
|
166
|
Wen L, Zhang Z, Peng R, Zhang L, Liu H, Peng H, Sun Y. Whole transcriptome analysis of diabetic nephropathy in the db/db mouse model of type 2 diabetes. J Cell Biochem 2019; 120:17520-17533. [PMID: 31106482 DOI: 10.1002/jcb.29016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
Abstract
Whole-transcriptome analysis using RNA sequencing (RNA-seq) affords broader insights about gene expression regulatory networks in diabetic nephropathy (DN). To better explore the molecular basis of DN, kidney tissue from db/db DN model mice and control mice were submitted to RNA-seq analysis. Thousands of long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) were found to be significantly differentially expressed in the DN group relative to the control group. To research the regulatory mechanism of these lncRNAs and mRNAs, the integrated co-expression networks were constructed for 322 mRNAs and 27 lncRNAs that revealed significantly correlated expression patterns in DN. The potential roles of these co-expressed mRNAs were classified by Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analyses. The co-expression networks involved 27 lncRNAs interacting with 38 key mRNAs related to metabolic processes, including ND4/4L, Ndufa2/5, Ndufb4/7, Ndufs3, Uqcrc1, Aco2, Alad, Alas1, Alpl, Atp5j2, Coq5, Coq6, Cth, and CytB, all of which are highly related to encoding subunits of the mitochondrial complexes. Thus, mitochondrial dysfunction could result in renal function decline in DN. Seven dysregulated lncRNAs and nine dysregulated mRNAs in the DN model were confirmed by quantitative real-time polymerase chain reaction. The lncRNA-mRNA co-expression network provides novel evidence to support the contention that metabolic changes are associated with metabolic reprogramming in the kidneys, and that these changes play a critical role during the progression of DN.
Collapse
Affiliation(s)
- Li Wen
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China.,Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Zhang
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| | - Rui Peng
- Department of Bioinformatics, Chongqing Medical University, Chongqing, China
| | - Luyu Zhang
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| | - Handeng Liu
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| | - Huimin Peng
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| |
Collapse
|
167
|
Non-Coding RNAs as New Therapeutic Targets in the Context of Renal Fibrosis. Int J Mol Sci 2019; 20:ijms20081977. [PMID: 31018516 PMCID: PMC6515288 DOI: 10.3390/ijms20081977] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/15/2022] Open
Abstract
Fibrosis, or tissue scarring, is defined as the excessive, persistent and destructive accumulation of extracellular matrix components in response to chronic tissue injury. Renal fibrosis represents the final stage of most chronic kidney diseases and contributes to the progressive and irreversible decline in kidney function. Limited therapeutic options are available and the molecular mechanisms governing the renal fibrosis process are complex and remain poorly understood. Recently, the role of non-coding RNAs, and in particular microRNAs (miRNAs), has been described in kidney fibrosis. Seminal studies have highlighted their potential importance as new therapeutic targets and innovative diagnostic and/or prognostic biomarkers. This review will summarize recent scientific advances and will discuss potential clinical applications as well as future research directions.
Collapse
|
168
|
The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun 2019; 10:1495. [PMID: 30940803 PMCID: PMC6445148 DOI: 10.1038/s41467-019-09482-6] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
The inflammasome has an essential function in innate immune, responding to a wide variety of stimuli. Here we show that the lncRNA Neat1 promotes the activation of several inflammasomes. Neat1 associates with the NLRP3, NLRC4, and AIM2 inflammasomes in mouse macrophages to enhance their assembly and subsequent pro-caspase-1 processing. Neat1 also stabilizes the mature caspase-1 to promote interleukin-1β production and pyroptosis. Upon stimulation with inflammasome-activating signals, Neat1, which normally resides in the paraspeckles, disassociates from these nuclear bodies and translocates to the cytoplasm to modulate inflammasome activation using above mechanism. Neat1 is also up-regulated under hypoxic conditions in a HIF-2α-dependent manner, mediating the effect of hypoxia on inflammasomes. Moreover, in the mouse models of peritonitis and pneumonia, Neat1 deficiency significantly reduces inflammatory responses. These results reveal a previously unrecognized role of lncRNAs in innate immunity, and suggest that Neat1 is a common mediator for inflammasome stimuli. The inflammasomes are important mediators of protective immunity by promoting inflammatory cytokine production and cell death. Here the authors show that a lncRNA, Neat1, is mobilized by inflammasome-activating signals to promote the assembly of several inflammasome complexes and cytokine maturation to regulate inflammation.
Collapse
|
169
|
Ma J, Zhao N, Du L, Wang Y. Downregulation of lncRNA NEAT1 inhibits mouse mesangial cell proliferation, fibrosis, and inflammation but promotes apoptosis in diabetic nephropathy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1174-1183. [PMID: 31933932 PMCID: PMC6947069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/18/2019] [Indexed: 06/10/2023]
Abstract
This study aimed to investigate the effect of long non-coding RNA nuclear enriched abundant transcript 1 (lnc-NEAT1) on mouse mesangial cells (MMCs) proliferation, apoptosis, fibrosis as well as inflammation in diabetic nephropathy (DN). MMCs (SV40 MES13 cells) were cultured under 30 mM glucose to construct DN cellular model (high glucose (HG) group); meanwhile, MMCs cultured under 5.6 mM glucose (normal glucose (NG) group) and 5.6 mM glucose plus 24.4 mM 3-O-methyl-D-glucose (osmotic control (OC) group) served as controls, and lnc-NEAT1 expression was determined by qPCR assay. Lnc-NEAT1 interference plasmids and control interference plasmids were transfected into DN cellular model as Sh-NEAT1 group and Sh-NC group. Cell proliferation, apoptosis, fibrosis, and inflammation were detected using Counting Kit-8, Annexin V/propidium iodide, western blot and quantitative polymerase chain reaction assays. Lnc-NEAT1 expression was elevated in HG group compared to NG group and OC group. Cell proliferation was decreased, and proliferative marker protein Cyclin D1 and proliferating cell nuclear antigen expressions also decreased in Sh-NEAT1 group compared to Sh-NC group. For cell apoptosis, apoptosis rate was increased, and apoptotic protein Cleaved Caspase3 expression enhanced but anti-apoptosis protein Bcl-2 expression decreased in Sh-NEAT1 group compared to Sh-NC group. For fibrosis markers (including fibronectin and collagen I) and inflammatory cytokines (including tumor necrosis factor-α, interleukin-1β and interleukin-6), their expressions were reduced in Sh-NEAT1 group compared to Sh-NC group. Lnc-NEAT1 is overexpressed, and its downregulation inhibits cell proliferation, fibrosis, and inflammation but promotes cell apoptosis in HG-induced MMCs DN cellular model.
Collapse
Affiliation(s)
- Jian Ma
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine Harbin, China
| | - Na Zhao
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine Harbin, China
| | - Likun Du
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine Harbin, China
| | - Yang Wang
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine Harbin, China
| |
Collapse
|
170
|
Peng W, Huang S, Shen L, Tang Y, Li H, Shi Y. Long noncoding RNA NONHSAG053901 promotes diabetic nephropathy via stimulating Egr-1/TGF-β-mediated renal inflammation. J Cell Physiol 2019; 234:18492-18503. [PMID: 30927260 DOI: 10.1002/jcp.28485] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 02/01/2023]
Abstract
Diabetic nephropathy (DN) is an important factor leading to end-stage kidney disease that affects diabetes mellitus patients globally. Our previous transcriptome sequencing has identified a large group of differentially expressed long noncoding RNA (lncRNA) in early development of DN. On basis of this, we aimed to investigate the function of lncRNA NONHSAG053901 in DN pathogenesis. In this study, we revealed that the expression of NONHSAG053901 was drastically elevated in both DN mouse model and mesangial cells (MCs). It was found that overexpression of NONHSAG053901 remarkably promoted inflammation, fibrosis and proliferation in MCs. Consistently, further investigations suggested that the stimulation of NONHSAG053901 on proinflammatory cytokines via direct binding to early growth response protein 1 (Egr-1). Interaction between Egr-1 and transforming growth factor β (TGF-β) could augment TGF-β function in DN inflammation. Furthermore, the effects of NONHSAG053901 on stimulation of proinflammatory cytokines were abolished by knockdown of Egr-1. These results together suggested that NONHSAG053901 promoted proinflammatory cytokines via stimulating Egr-1/TGF-β mediated renal inflammation.
Collapse
Affiliation(s)
- Wenfang Peng
- Endocrinology Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China.,Endocrinology Department, Shanghai Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Shan Huang
- Endocrinology Department, Shanghai Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Lisha Shen
- Endocrinology Department, Shanghai Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yubing Tang
- Endocrinology Department, Shanghai Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Huihua Li
- Endocrinology Department, Shanghai Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yongquan Shi
- Endocrinology Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
171
|
Biswas S, Chakrabarti S. Increased Extracellular Matrix Protein Production in Chronic Diabetic Complications: Implications of Non-Coding RNAs. Noncoding RNA 2019; 5:E30. [PMID: 30909482 PMCID: PMC6468528 DOI: 10.3390/ncrna5010030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Management of chronic diabetic complications remains a major medical challenge worldwide. One of the characteristic features of all chronic diabetic complications is augmented production of extracellular matrix (ECM) proteins. Such ECM proteins are deposited in all tissues affected by chronic complications, ultimately causing organ damage and dysfunction. A contributing factor to this pathogenetic process is glucose-induced endothelial damage, which involves phenotypic transformation of endothelial cells (ECs). This phenotypic transition of ECs, from a quiescent state to an activated dysfunctional state, can be mediated through alterations in the synthesis of cellular proteins. In this review, we discussed the roles of non-coding RNAs, specifically microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in such processes. We further outlined other epigenetic mechanisms regulating the biogenesis and/or function of non-coding RNAs. Overall, we believe that better understanding of such molecular processes may lead to the development of novel biomarkers and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Saumik Biswas
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| |
Collapse
|
172
|
Shang J, Wang S, Jiang Y, Duan Y, Cheng G, Liu D, Xiao J, Zhao Z. Identification of key lncRNAs contributing to diabetic nephropathy by gene co-expression network analysis. Sci Rep 2019; 9:3328. [PMID: 30824724 PMCID: PMC6397236 DOI: 10.1038/s41598-019-39298-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/21/2019] [Indexed: 01/13/2023] Open
Abstract
LncRNA is reported to have important role in diabetic nephropathy (DN). Here, we aim to identify key lncRNAs of DN using bioinformatics and systems biological methods. Method: Five microarray data sets from Gene Expression Omnibus (GEO) database were included. Probe sets were re-annotated. In the training set, differential expressed genes (DEGs) were identified. Weighted gene co-expression network analysis (WGCNA) was constructed to screen diabetic-related hub genes and reveal their potential biological function. Two more human data sets and mouse data sets were used as validation sets. Results: A total of 424 DEGs, including 10 lncRNAs, were filtered in the training data set. WGCNA and enrichment analysis of hub genes showed that inflammation and metabolic disorders are prominent in DN. Three key lncRNAs (NR_130134.1, NR_029395.1 and NR_038335.1) were identified. These lncRNAs are also differently expressed in another two human data sets. Functional enrichment of the mouse data sets showed consistent changes with that in human, indicating similar changes in gene expression pattern of DN and confirmed confidence of our analysis. Human podocytes and mesangial cells were culture in vitro. QPCR and fluorescence in situ hybridization were taken out to validate the expression and relationship of key lncRNAs and their related mRNAs. Results were also consistent with our analysis. Conclusions: Inflammation and metabolic disorders are prominent in DN. We identify three lncRNAs that are involved in these processes possibly by interacting with co-expressed mRNAs.
Collapse
Affiliation(s)
- Jin Shang
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Shuai Wang
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Yumin Jiang
- Department of Emergency, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Yiqi Duan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Genyang Cheng
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Dong Liu
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Jing Xiao
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Zhanzheng Zhao
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.
| |
Collapse
|
173
|
Abstract
PURPOSE OF REVIEW Epigenetic variations have been shown to reveal vulnerability to diabetes and its complications. Although it has become clear that metabolic derangements, especially hyperglycemia, can impose a long-term metabolic memory that predisposes to diabetic complications, the underlying mechanisms remain to be understood. It has been suggested that epigenetics (e.g., histone modification, DNA methylation, and non-coding RNAs) help link metabolic disruption to aberrancies related to diabetic kidney disease (DKD). In this review, we discuss the key findings and advances made in the epigenetic risk profile of DKD and provide perspectives on the emerging topics that implicate epigenetics in DKD. RECENT FINDINGS Epigenetic profiles can be profoundly altered in patients with diabetes, in circulating blood cells as well as in renal tissues. These changes provide useful insight into the mechanisms of diabetic kidney injury and progressive kidney dysfunction. Increasing evidence supports the role of epigenetic regulation in DKD. More studies are needed to elucidate the mechanism and importance of epigenetic changes in the initiation and progression of DKD and to further explore their diagnostic and therapeutic potential in the clinical management of patients with diabetes who have a high risk for DKD.
Collapse
Affiliation(s)
- Lixia Xu
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
- Division of Nephrology, Guangdong Academy of Medical Science and Guangdong General Hospital, 106 Zhongshan Er Rd, Guangzhou, 510080, China
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
| |
Collapse
|
174
|
Long noncoding RNAs in cancer cells. Cancer Lett 2019; 419:152-166. [PMID: 29414303 DOI: 10.1016/j.canlet.2018.01.053] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 12/11/2022]
Abstract
Long noncoding RNA (lncRNA) has recently been investigated as key modulators that regulate many biological processes in human cancers via diverse mechanisms. LncRNAs can interact with macromolecules such as DNA, RNA, or protein to exert cellular effects and to act as either tumor promoters or tumor suppressors in various malignancies. Moreover, the aberrant expression of lncRNAs may be detected in multiple cancer phenotypes by employing the rapidly developing modern gene chip technology and bioinformatics analysis. Herein, we highlight the mechanisms of action of lncRNAs, their functional cellular roles and their involvement in cancer progression. Finally, we provide an overview of recent progress in the lncRNA field and future potential for lncRNAs as cancer diagnostic markers and therapeutics.
Collapse
|
175
|
Li Y, Xu K, Xu K, Chen S, Cao Y, Zhan H. Roles of Identified Long Noncoding RNA in Diabetic Nephropathy. J Diabetes Res 2019; 2019:5383010. [PMID: 30891461 PMCID: PMC6390257 DOI: 10.1155/2019/5383010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is the leading chronic disease in the world, and diabetic nephropathy (DN) as one of its complications could increase the mortality. The development of DN is associated to abnormal hemodynamic factors like cytokine networks and the intervention of metabolic risk factors like blood pressure, blood glucose, and blood lipid. However, the pathogenesis of DN is still poorly understood. Although glucose-lowering drugs and insulins have significant effects on blood glucose, the fluctuation of blood glucose or other risk factors could continuously damage the kidney. Recent studies reported that the progression of DN is closely related to the expression of long noncoding RNA (lncRNA), which is important for the early diagnosis and targeted intervention of DN. In this review, we briefly summarize the published studies on the functions and potential mechanism of reported lncRNA in the regulation of DN.
Collapse
Affiliation(s)
- Yan Li
- The First Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075 Sichuan, China
| | - Keyang Xu
- Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang, China
| | - Kechen Xu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000 Zhejiang, China
| | - Sixiang Chen
- Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang, China
| | - Yifang Cao
- The First Hospital of Jiaxing, Jiaxing, 314001 Zhejiang, China
| | - Huakui Zhan
- The First Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075 Sichuan, China
| |
Collapse
|
176
|
Abstract
The main cellular constituents in glomerular mesangium are mesangial cells, which account for approximately 30-40% of the total cells in the glomerulus. Together with the mesangial matrix, mesangial cells form the glomerular basement membrane (GBM) in the glomerulus, whose main function is to perform the filtration. Under the pathologic conditions, mesangial cells are activated, leading to hyperproliferation and excess extracellular matrix (ECM). Moreover, mesangial cells also secrete several kinds of inflammatory cytokines, adhesion molecules, chemokines, and enzymes, all of which participate in the process of renal glomerular fibrosis. During the past years, researchers have revealed the roles of mesangial cells and the associated signal pathways involved in renal fibrosis. In this section, we will discuss how mesangial cells are activated and its contributions to renal fibrosis, as well as the molecular mechanisms and novel anti-fibrotic agents. Full understanding of the contributions of mesangial cells to renal fibrosis will benefit the clinical drug developing.
Collapse
Affiliation(s)
- Jing-Hong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
177
|
Yaribeygi H, Katsiki N, Butler AE, Sahebkar A. Effects of antidiabetic drugs on NLRP3 inflammasome activity, with a focus on diabetic kidneys. Drug Discov Today 2019; 24:256-262. [DOI: 10.1016/j.drudis.2018.08.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022]
|
178
|
Ji E, Kim C, Kim W, Lee EK. Role of long non-coding RNAs in metabolic control. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1863:194348. [PMID: 30594638 DOI: 10.1016/j.bbagrm.2018.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of gene expression by influencing various biological processes including proliferation, apoptosis, differentiation, and senescence. Accumulating evidence implicates lncRNAs in the maintenance of metabolic homeostasis; dysregulation of certain lncRNAs promotes the progression of metabolic disorders such as diabetes, obesity, and cardiovascular diseases. In this review, we discuss our understanding of lncRNAs implicated in metabolic control, focusing on in particular diseases arising from chronic inflammation, insulin resistance, and lipid homeostasis. We have analyzed lncRNAs and their molecular targets involved in the pathogenesis of chronic liver disease, diabetes, and obesity, and have discussed the rising interest in lncRNAs as diagnostic and therapeutic targets improving metabolic homeostasis. This article is part of a Special Issue entitled: ncRNA in control of gene expression edited by Kotb Abdelmohsen.
Collapse
Affiliation(s)
- Eunbyul Ji
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea
| | - Chongtae Kim
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Eun Kyung Lee
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul 06591, South Korea.
| |
Collapse
|
179
|
Downregulation of DAPK1 promotes the stemness of cancer stem cells and EMT process by activating ZEB1 in colorectal cancer. J Mol Med (Berl) 2018; 97:89-102. [DOI: 10.1007/s00109-018-1716-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 08/25/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022]
|
180
|
Abstract
SIGNIFICANCE Diabetes is associated with markedly accelerated rates of micro- and macrovascular complications that increase morbidity and mortality. Understanding the molecular mechanisms can promote much needed therapeutics. Recent Advances: Long noncoding RNAs (lncRNAs) are important regulators of gene regulation and cellular function and are emerging as important players in diabetes and its complications. There are number of examples in which lncRNAs are responsive to hyperglycemia and clearly involved in regulation of genes and pathways associated with the development of diabetic complications. CRITICAL ISSUES As there are likely thousands of lncRNAs that are expressed in any given tissue, understanding how they are regulated and function in the normal healthy state as well as pathological states is a challenge. FUTURE DIRECTIONS Further studies in how lncRNAs are involved in the development and progression of diabetic complications as well as development of methods to target dysregulated lncRNAs or evaluate them as biomarkers of early detection of organ dysfunction will be highly beneficial to treating diabetic patients.
Collapse
Affiliation(s)
- Amy Leung
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope , Duarte, California
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope , Duarte, California
| |
Collapse
|
181
|
Zhu X, Shi J, Li H. Liquiritigenin attenuates high glucose-induced mesangial matrix accumulation, oxidative stress, and inflammation by suppression of the NF-κB and NLRP3 inflammasome pathways. Biomed Pharmacother 2018; 106:976-982. [DOI: 10.1016/j.biopha.2018.07.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/30/2022] Open
|
182
|
Qiao Q, Wang F, Gu Y. LncRNA Gm4419 promotes the development of cardiac diseases in type 2 diabetic patients with diabetic nephropathy. Int J Diabetes Dev Ctries 2018. [DOI: 10.1007/s13410-018-0690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
183
|
Ren G, Zhu J, Li J, Meng X. Noncoding RNAs in acute kidney injury. J Cell Physiol 2018; 234:2266-2276. [PMID: 30146769 DOI: 10.1002/jcp.27203] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/16/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Gui‐Ling Ren
- Department of PharmacyThe 105 Hospital of Chinese People’s Liberation ArmyHefei China
| | - Jie Zhu
- Department of PharmacyThe 105 Hospital of Chinese People’s Liberation ArmyHefei China
| | - Jun Li
- Department of PharmacologySchool of Pharmacy, Anhui Medical UniversityHefei China
- Anhui Institute of Innovative Drugs, Anhui Medical UniversityHefei China
| | - Xiao‐Ming Meng
- Department of PharmacologySchool of Pharmacy, Anhui Medical UniversityHefei China
- Anhui Institute of Innovative Drugs, Anhui Medical UniversityHefei China
| |
Collapse
|
184
|
Sathishkumar C, Prabu P, Mohan V, Balasubramanyam M. Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Hum Genomics 2018; 12:41. [PMID: 30139387 PMCID: PMC6107963 DOI: 10.1186/s40246-018-0173-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022] Open
Abstract
Background Studying epigenetics is expected to provide precious information on how environmental factors contribute to type 2 diabetes mellitus (T2DM) at the genomic level. With the progress of the whole-genome resequencing efforts, it is now known that 75–90% of the human genome was transcribed to generate a series of long non-coding RNAs (lncRNAs). While lncRNAs are gaining widespread attention as potential and robust biomarkers in the genesis as well as progression of several disease states, their clinical relevance and regulatory mechanisms are yet to be explored in the field of metabolic disorders including diabetes. Despite the fact that Asian Indians are highly insulin resistant and more prone to develop T2DM and associated vascular complications, there is virtually lack of data on the role of lncRNAs in the clinical diabetes setting. Therefore, we sought to evaluate a panel of lncRNAs and senescence-inflammation signatures in peripheral blood mononuclear cells (PBMCs) from patients with type 2 diabetes (T2DM; n = 30) compared to individuals with normal glucose tolerance (NGT; n = 32). Results Compared to control subjects, expression levels of lncRNAs in PBMCs from type 2 diabetes patients showed significantly (p < 0.05) increased levels of HOTAIR, MEG3, LET, MALAT1, MIAT, CDKN2BAS1/ANRIL, XIST, PANDA, GAS5, Linc-p21, ENST00000550337.1, PLUTO, and NBR2. In contrast, lncRNA expression patterns of THRIL and SALRNA1 were significantly (p < 0.05) decreased in patients with T2DM compared to control subjects. At the transcriptional level, senescence markers (p53, p21, p16, and β-galactosidase), proinflammatory markers (TNF-α, IL6, MCP1, and IL1-β), and epigenetic signature of histone deacetylase-3 (HDAC3) were significantly (p < 0.05) elevated in patients with type 2 diabetes compared to control subjects. Interestingly, mRNA expression of Sirt1 and telomere length were significantly (p < 0.05) decreased in patients with type 2 diabetes compared to control subjects. Majority of the altered lncRNAs were positively correlated with poor glycemic control, insulin resistance, transcriptional markers of senescence, inflammation, and HDAC3 and negatively correlated with telomere length. Logistic regression analysis revealed a significant association of altered lncRNA signatures with T2DM, but this association was lost after adjusting for insulin resistance (HOMA-IR) and senescence markers. Conclusion Our study provides a clinically relevant evidence for the association of altered lncRNAs with poor glycemic control, insulin resistance, accelerated cellular senescence, and inflammation. Electronic supplementary material The online version of this article (10.1186/s40246-018-0173-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chandrakumar Sathishkumar
- Department of Cell and Molecular Biology and Dr. Rema Mohan High-Throughput Screening (HTS) Lab, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, 600 086, India
| | - Paramasivam Prabu
- Department of Cell and Molecular Biology and Dr. Rema Mohan High-Throughput Screening (HTS) Lab, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, 600 086, India
| | - Viswanathan Mohan
- Department of Cell and Molecular Biology and Dr. Rema Mohan High-Throughput Screening (HTS) Lab, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, 600 086, India
| | - Muthuswamy Balasubramanyam
- Department of Cell and Molecular Biology and Dr. Rema Mohan High-Throughput Screening (HTS) Lab, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, 600 086, India.
| |
Collapse
|
185
|
Bai X, Geng J, Li X, Wan J, Liu J, Zhou Z, Liu X. Long Noncoding RNA LINC01619 Regulates MicroRNA-27a/Forkhead Box Protein O1 and Endoplasmic Reticulum Stress-Mediated Podocyte Injury in Diabetic Nephropathy. Antioxid Redox Signal 2018; 29:355-376. [PMID: 29334763 DOI: 10.1089/ars.2017.7278] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Altered activities of long noncoding RNAs (lncRNAs) have been implicated in the regulation of microRNAs. microRNA-27a (miR-27a) upregulation has been shown to induce endoplasmic reticulum (ER) stress podocyte injury in diabetic nephropathy (DN). Herein, we aim to interrogate the mutually regulated network of miR-27a with long intergenic noncoding RNA 1619 (LINC01619) and the target gene. RESULTS LINC01619 downregulation was found in human DN renal biopsy tissues and contributed to proteinuria and diminished renal function. LINC01619 was expressed in podocyte cytoplasm and involved in ER stress signaling pathway. LINC01619 exerted biological function by serving as a "sponge" for miR-27a, which negatively targeted forkhead box protein O1 (FOXO1) and activated ER stress. In diabetic rats and high-glucose cultured podocytes, LINC01619 triggered oxidative stress and podocyte injuries as demonstrated by increased apoptosis, diffuse podocyte foot process effacement, and decreased renal function. Innovation and Conclusion: This study demonstrates that LINC01619 functions as a competing endogenous RNA and regulates miR-27a/FOXO1-mediated ER stress and podocyte injury in DN. Antioxid. Redox Signal. 29, 355-376.
Collapse
Affiliation(s)
- Xiaoyan Bai
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Jian Geng
- 2 Department of Pathology, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Xiao Li
- 3 Department of Emergency, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Jiao Wan
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Jixing Liu
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Zhanmei Zhou
- 1 Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University , Guangzhou, People's Republic of China
| | - Xiaoting Liu
- 4 Department of Pathology, King Medical Diagnostics Center , Guangzhou, People's Republic of China
| |
Collapse
|
186
|
Xu B, Wang Y, Li X, Mao Y, Deng X. RNA‑sequencing analysis of aberrantly expressed long non‑coding RNAs and mRNAs in a mouse model of ventilator‑induced lung injury. Mol Med Rep 2018; 18:882-892. [PMID: 29845294 PMCID: PMC6059720 DOI: 10.3892/mmr.2018.9034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/22/2018] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are closely associated with the regulation of various biological processes and are involved in the pathogenesis of numerous diseases. However, to the best of our knowledge, the role of lncRNAs in ventilator‑induced lung injury (VILI) has yet to be evaluated. In the present study, high‑throughput sequencing was applied to investigate differentially expressed lncRNAs and mRNAs (fold change >2; false discovery rate <0.05). Bioinformatics analysis was employed to predict the functions of differentially expressed lncRNAs. A total of 104 lncRNAs (74 upregulated and 30 downregulated) and 809 mRNAs (521 upregulated and 288 downregulated) were differentially expressed in lung tissues from the VILI group. Gene ontology analysis demonstrated that the differentially expressed lncRNAs and mRNAs were mainly associated with biological functions, including apoptosis, angiogenesis, neutrophil chemotaxis and skeletal muscle cell differentiation. The top four enriched pathways were the tumor necrosis factor (TNF) signaling pathway, P53 signaling pathway, neuroactive ligand‑receptor interaction and the forkhead box O signaling pathway. Several lncRNAs were predicted to serve a vital role in VILI. Subsequently, three lncRNAs [mitogen‑activated protein kinase kinase 3, opposite strand (Map2k3os), dynamin 3, opposite strand and abhydrolase domain containing 11, opposite strand] and three mRNAs (growth arrest and DNA damage‑inducible α, claudin 4 and thromboxane A2 receptor) were measured by reverse transcription‑quantitative polymerase chain reaction, in order to confirm the veracity of RNA‑sequencing analysis. In addition, Map2k3os small interfering RNA transfection inhibited the expression of stretch‑induced cytokines [TNF‑α, interleukin (IL)‑1β and IL‑6] in MLE12 cells. In conclusion, the results of the present study provided a profile of differentially expressed lncRNAs in VILI. Several important lncRNAs may be involved in the pathological process of VILI, which may be useful to guide further investigation into the pathogenesis for this disease.
Collapse
Affiliation(s)
- Bo Xu
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yizhou Wang
- Department of Hepatic Surgery IV, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Xiujuan Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yanfei Mao
- Department of Anesthesiology and SICU, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P.R. China
| | - Xiaoming Deng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
187
|
Goyal N, Kesharwani D, Datta M. Lnc-ing non-coding RNAs with metabolism and diabetes: roles of lncRNAs. Cell Mol Life Sci 2018; 75:1827-1837. [PMID: 29387902 PMCID: PMC11105777 DOI: 10.1007/s00018-018-2760-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/29/2017] [Accepted: 01/24/2018] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes is a complex metabolic disorder characterized by insulin resistance and pancreatic β-cell dysfunction. Deregulated glucose and lipid metabolism are the primary underlying manifestations associated with this disease and its complications. Long non-coding RNAs (lncRNAs) are a novel class of functional RNAs that regulate a variety of biological processes by a diverse interplay of mechanisms including recruitment of epigenetic modifiers, transcriptional and post-transcriptional regulation, control of mRNA decay, and sequestration of transcription factors. Although the underlying causes that define the diabetic phenotype are extremely intricate, most of the studies in the last decades were mostly centered on protein-coding genes. However, current opinion in the recent past has authenticated the contributions of diverse lncRNAs as critical regulatory players during the manifestation of diabetes. The current review will highlight the importance of lncRNAs in regulating cellular processes that govern metabolic homeostasis in key metabolic tissues. A more in-depth understanding of lncRNAs may enable their exploitation as biomarkers or for therapeutic applications during diabetes and its associated complications.
Collapse
Affiliation(s)
- Neha Goyal
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research, Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, India
| | - Devesh Kesharwani
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India
- Academy of Scientific and Innovative Research, Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, India
| | - Malabika Datta
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.
- Academy of Scientific and Innovative Research, Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, India.
| |
Collapse
|
188
|
Liu YW, Hao YC, Chen YJ, Yin SY, Zhang MY, Kong L, Wang TY. Protective effects of sarsasapogenin against early stage of diabetic nephropathy in rats. Phytother Res 2018; 32:1574-1582. [DOI: 10.1002/ptr.6088] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/11/2018] [Accepted: 03/14/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Yao-Wu Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
- Department of Pharmacology, School of Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Yun-Chao Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Ya-Jing Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Shen-Yuan Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Meng-Ya Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Li Kong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical University; Xuzhou 221004 Jiangsu China
| | - Tao-Yun Wang
- College of Chemistry, Biology and Material Engineering; Suzhou University of Science and Technology; Suzhou 215009 China
| |
Collapse
|
189
|
Sharma A, Tate M, Mathew G, Vince JE, Ritchie RH, de Haan JB. Oxidative Stress and NLRP3-Inflammasome Activity as Significant Drivers of Diabetic Cardiovascular Complications: Therapeutic Implications. Front Physiol 2018. [PMID: 29515457 PMCID: PMC5826188 DOI: 10.3389/fphys.2018.00114] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It is now increasingly appreciated that inflammation is not limited to the control of pathogens by the host, but rather that sterile inflammation which occurs in the absence of viral or bacterial pathogens, accompanies numerous disease states, none more so than the complications that arise as a result of hyperglycaemia. Individuals with type 1 or type 2 diabetes mellitus (T1D, T2D) are at increased risk of developing cardiac and vascular complications. Glucose and blood pressure lowering therapies have not stopped the advance of these morbidities that often lead to fatal heart attacks and/or stroke. A unifying mechanism of hyperglycemia-induced cellular damage was initially proposed to link elevated blood glucose levels with oxidative stress and the dysregulation of metabolic pathways. Pre-clinical evidence has, in most cases, supported this notion. However, therapeutic strategies to lessen oxidative stress in clinical trials has not proved efficacious, most likely due to indiscriminate targeting by antioxidants such as vitamins. Recent evidence now suggests that oxidative stress is a major driver of inflammation and vice versa, with the latest findings suggesting not only a key role for inflammatory pathways underpinning metabolic and haemodynamic dysfunction in diabetes, but furthermore that these perturbations are driven by activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. This review will address these latest findings with an aim of highlighting the interconnectivity between oxidative stress, NLRP3 activation and inflammation as it pertains to cardiac and vascular injury sustained by diabetes. Current therapeutic strategies to lessen both oxidative stress and inflammation will be emphasized. This will be placed in the context of improving the burden of these diabetic complications.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mitchel Tate
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geetha Mathew
- Cellular Therapies Laboratory, Westmead Hospital, Sydney, NSW, Australia
| | - James E Vince
- Inflammation Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Judy B de Haan
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
190
|
Moghaddas Sani H, Hejazian M, Hosseinian Khatibi SM, Ardalan M, Zununi Vahed S. Long non-coding RNAs: An essential emerging field in kidney pathogenesis. Biomed Pharmacother 2018; 99:755-765. [PMID: 29710473 DOI: 10.1016/j.biopha.2018.01.122] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 12/21/2022] Open
Abstract
Human Genome Project has made it clear that a majority of the genome is transcribed into the non-coding RNAs including microRNAs as well as long non-coding RNAs (lncRNAs) which both can affect different features of cells. LncRNAs are long heterogenous RNAs that regulate gene expression and a variety of signaling pathways involved in cellular homeostasis and development. Studies over the past decade have shown that lncRNAs have a major role in the kidney pathogenesis. The effective roles of lncRNAs have been recognized in renal ischemia, injury, inflammation, fibrosis, glomerular diseases, renal transplantation, and renal cell carcinoma. The present review outlines the role and function of lncRNAs in kidney pathogenesis as novel essential regulators. Molecular mechanism insights into the functions of lncRNAs in kidney pathophysiological processes may contribute to effective future therapeutics.
Collapse
Affiliation(s)
| | - Mina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|
191
|
Leung A, Amaram V, Natarajan R. Linking diabetic vascular complications with LncRNAs. Vascul Pharmacol 2018; 114:139-144. [PMID: 29398367 DOI: 10.1016/j.vph.2018.01.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022]
Abstract
Diabetes leads to markedly accelerated rates of many associated macrovascular complications like hypertension and atherosclerosis, and microvascular complications like nephropathy and retinopathy. High glucose, the hallmark of diabetes, drives changes in vascular and inflammatory cells that promote the development of these complications. Understanding the molecular processes involved in the development of diabetes and its debilitating complications can lead to much needed newer clinical therapies. Recently, long-noncoding RNAs (lncRNAs) have been shown to be important in the biology of vascular cells and there is growing evidence that lncRNAs are also involved in the cell biology relevant to diabetic vascular complications. In this review, we provide an overview of lncRNAs that function in vascular cells, and those that have been linked to diabetic complications.
Collapse
Affiliation(s)
- Amy Leung
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States
| | - Vishnu Amaram
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, United States
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, United States.
| |
Collapse
|
192
|
Raut SK, Khullar M. The Big Entity of New RNA World: Long Non-Coding RNAs in Microvascular Complications of Diabetes. Front Endocrinol (Lausanne) 2018; 9:300. [PMID: 29915562 PMCID: PMC5994400 DOI: 10.3389/fendo.2018.00300] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 05/18/2018] [Indexed: 12/14/2022] Open
Abstract
A major part of the genome is known to be transcribed into non-protein coding RNAs (ncRNAs), such as microRNA and long non-coding RNA (lncRNA). The importance of ncRNAs is being increasingly recognized in physiological and pathological processes. lncRNAs are a novel class of ncRNAs that do not code for proteins and are important regulators of gene expression. In the past, these molecules were thought to be transcriptional "noise" with low levels of evolutionary conservation. However, recent studies provide strong evidence indicating that lncRNAs are (i) regulated during various cellular processes, (ii) exhibit cell type-specific expression, (iii) localize to specific organelles, and (iv) associated with human diseases. Emerging evidence indicates an aberrant expression of lncRNAs in diabetes and diabetes-related microvascular complications. In the present review, we discuss the current state of knowledge of lncRNAs, their genesis from genome, and the mechanism of action of individual lncRNAs in the pathogenesis of microvascular complications of diabetes and therapeutic approaches.
Collapse
|
193
|
Zhang YL, Wang RB, Li WY, Xia FZ, Liu L. Pioglitazone ameliorates retinal ischemia/reperfusion injury via suppressing NLRP3 inflammasome activities. Int J Ophthalmol 2017; 10:1812-1818. [PMID: 29259897 DOI: 10.18240/ijo.2017.12.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/25/2017] [Indexed: 01/16/2023] Open
Abstract
AIM To explore the role of Pioglitazone (Pio) on a mouse model of retinal ischemia/reperfusion (I/R) injury and to elucidate the potential mechanism. METHODS Retinal ischemia was induced in mice by increasing the intraocular pressure, and Pio was administered 4h though periocular injection before I/R. The number of cells in the ganglion cell layer (GCL) was counted 7d after retinal I/R injury. Glial fibrillary acidic protein (GFAP), nuclear factor-kappa B (NF-κB), p38, phosphorylated-p38, PPAR-γ, interleukin-1β (IL-1β), Toll-like receptor 4 (TLR4), NLRP3, cleaved caspase-1, caspase-1 were determined by real-time polymerase chain reaction and Western blotting. RESULTS Pio promoted the survival of retinal cells in GCL following retinal I/R injury (P<0.05). Besides, retinal I/R injury stimulated the expression of GFAP and TLR4, which were partially reversed by Pio treatment (P<0.05). Retinal I/R injury-upregulated expression of NLRP3, cleaved caspase-1, IL-1β was attenuated after Pio treatment (P<0.05). Moreover, I/R injury induced activation of NF-κB and p38 were inhibited by Pio treatment (P<0.05). CONCLUSION Pio promotes retinal ganglion cells survival by suppressing I/R-induced activation of TLR4/NLRP3 inflammasomes via inhibiting NF-κB and p38 phosphorylation.
Collapse
Affiliation(s)
- Yue-Lu Zhang
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ruo-Bing Wang
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei-Yi Li
- Department of Ophthalmology, Shandong University Qilu Hospital (Qingdao), Qingdao 266035, Shandong Province, China
| | - Fang-Zhou Xia
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lin Liu
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
194
|
Chen D, Xiong XQ, Zang YH, Tong Y, Zhou B, Chen Q, Li YH, Gao XY, Kang YM, Zhu GQ. BCL6 attenuates renal inflammation via negative regulation of NLRP3 transcription. Cell Death Dis 2017; 8:e3156. [PMID: 29072703 PMCID: PMC5680929 DOI: 10.1038/cddis.2017.567] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022]
Abstract
Renal inflammation contributes to the pathogeneses of hypertension. This study was designed to determine whether B-cell lymphoma 6 (BCL6) attenuates renal NLRP3 inflammasome activation and inflammation and its underlying mechanism. Male spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were used in the present study. Angiotensin (Ang) II or lipopolysaccharides (LPS) was used to induce inflammation in HK-2 cells, a human renal tubular epithelial (RTE) cell line. NLRP3 inflammasome was activated and BCL6 was downregulated in the kidneys of SHR. Either Ang II or LPS suppressed BCL6 expression in HK-2 cells. BCL6 overexpression in HK-2 cells attenuated Ang II-induced NLRP3 upregulation, inflammation and cell injury. The inhibitory effects of BCL6 overexpression on NLRP3 expression and inflammation were also observed in LPS-treated HK-2 cells. BCL6 inhibited the NLRP3 transcription via binding to the NLRP3 promoter. BCL6 knockdown with shRNA increased NLRP3 and mature IL-1β expression levels in both PBS- or Ang II-treated HK-2 cells but had no significant effects on ASC, pro-caspase-1 and pro-IL-1β expression levels. BCL6 overexpression caused by recombinant lentivirus expressing BCL6 reduced blood pressure in SHR. BCL6 overexpression prevented the upregulation of NLRP3 and mature IL-1β expression levels in the renal cortex of SHR. The results indicate that BCL6 attenuates Ang II- or LPS-induced inflammation in HK-2 cells via negative regulation of NLRP3 transcription. BCL6 overexpression in SHR reduced blood pressure, NLRP3 expression and inflammation in the renal cortex of SHR.
Collapse
Affiliation(s)
- Dan Chen
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Qing Xiong
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying-Hao Zang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Tong
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bing Zhou
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xing-Ya Gao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
195
|
Sun HJ, Ren XS, Xiong XQ, Chen YZ, Zhao MX, Wang JJ, Zhou YB, Han Y, Chen Q, Li YH, Kang YM, Zhu GQ. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis 2017; 8:e3074. [PMID: 28981106 PMCID: PMC5680591 DOI: 10.1038/cddis.2017.470] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 01/12/2023]
Abstract
Inflammation is involved in pathogenesis of hypertension. NLRP3 inflammasome activation is a powerful mediator of inflammatory response via caspase-1 activation. The present study was designed to determine the roles and mechanisms of NLRP3 inflammasome in phenotypic modulation and proliferation of vascular smooth muscle cells (VSMCs) in hypertension. Experiments were conducted in spontaneously hypertensive rats (SHR) and primary aortic VSMCs. NLRP3 inflammasome activation was observed in the media of aorta in SHR and in the VSMCs from SHR. Knockdown of NLRP3 inhibited inflammasome activation, VSMC phenotypic transformation and proliferation in SHR-derived VSMCs. Increased NFκB activation, histone acetylation and histone acetyltransferase expression were observed in SHR-derived VSMCs and in media of aorta in SHR. Chromatin immunoprecipitation analysis revealed the increased histone acetylation, p65-NFκB and Pol II occupancy at the NLRP3 promoter in vivo and in vitro. Inhibition of NFκB with BAY11-7082 or inhibition of histone acetyltransferase with curcumin prevented the NLRP3 inflammasome activation, VSMC phenotype switching and proliferation in VSMCs from SHR. Moreover, curcumin repressed NFκB activation. Silencing of NLRP3 gene ameliorated hypertension, vascular remodeling, NLRP3 inflammasome activation and phenotype switching in the aorta of SHR. These results indicate that NLRP3 inflammasome activation response to histone acetylation and NFκB activation contributes to VSMC phenotype switching and proliferation and vascular remodeling in hypertension.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xing-Sheng Ren
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Qing Xiong
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun-Zhi Chen
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Xia Zhao
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jue-Jin Wang
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ye-Bo Zhou
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ying Han
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710061, China
| | - Guo-Qing Zhu
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
196
|
He X, Ou C, Xiao Y, Han Q, Li H, Zhou S. LncRNAs: key players and novel insights into diabetes mellitus. Oncotarget 2017; 8:71325-71341. [PMID: 29050364 PMCID: PMC5642639 DOI: 10.18632/oncotarget.19921] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/19/2017] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (LncRNAs) are a class of endogenous RNA molecules, which have a transcribing length of over 200 nt, lack a complete functional open reading frame (ORF), and rarely encode a functional short peptide. Recent studies have revealed that disruption of LncRNAs levels correlates with several human diseases, including diabetes mellitus (DM), a complex multifactorial metabolic disorder affecting more than 400 million people worldwide. LncRNAs are emerging as pivotal regulators in various biological processes, in the progression of DM and its associated complications, involving pancreatic β-cell disorder, insulin resistance, and epigenetic regulation, etc. Further investigation into the mechanisms of action of LncRNAs in DM will be of great value in the thorough understanding of pathogenesis. However, prior to successful application of LncRNAs, further search for molecular biomarkers and drug targets to provide a new strategy for DM prevention, early diagnosis, and therapy is warranted.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunlin Ou
- Cancer Research Institute, Central South University, Changsha 410078, China
| | - Yanhua Xiao
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Qing Han
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Hao Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Suxian Zhou
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| |
Collapse
|
197
|
Yu Y, Cao F, Ran Q, Wang F. Long non-coding RNA Gm4419 promotes trauma-induced astrocyte apoptosis by targeting tumor necrosis factor α. Biochem Biophys Res Commun 2017; 491:478-485. [DOI: 10.1016/j.bbrc.2017.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/04/2017] [Indexed: 12/20/2022]
|
198
|
Sun J, Zhang S, Shi B, Zheng D, Shi J. Transcriptome Identified lncRNAs Associated with Renal Fibrosis in UUO Rat Model. Front Physiol 2017; 8:658. [PMID: 28912732 PMCID: PMC5583212 DOI: 10.3389/fphys.2017.00658] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
Renal fibrosis represents a final common outcome of many renal diseases and has attracted a great deal of attention. To better understand whether lncRNAs could be a player in this process or be a biomarker for renal fibrosis diagnosis, we compared transcriptome sequencing data on renal tissues and urine respectively between UUO (unilateral ureteral obstruction) and shamed (Sham) rat model. Numerous genes including lncRNAs with significant changes in their expression were identified. 24 lncRNAs were up-regulated and 79 lncRNAs were down-regulated in the renal tissues of the UUO rats. 625 lncRNAs were up-regulated and 177 lncRNAs were down-regulated in urines of the UUO rats. Among the lncRNAs upregulated in renal tissue of UUO rats, 19 lncRNAs were predicted containing several conserved Smad3 binding motifs in the promoter. Among them, lncRNAs with putative promoter containing more than 4 conserved Smad3 binding motifs were demonstrated to be induced by TGF-β significantly in normal rat renal tubular epithelial NRK-52E cells. We further confirmed that lncRNA TCONS_00088786 and TCONS_01496394 were regulated by TGF-β stimulation and also can influence the expression of some fibrosis-related genes through a feedback loop. Based on transcriptome sequencing data, bioinformatics analysis and qRT-PCR detection, we also demonstrated lncRNA in urine are detectable and might be a novel biomarker of renal fibrosis. These results provide new information for the involvement of lncRNAs in renal fibrosis, indicating that they may serve as candidate biomarkers or therapeutic targets in the future.
Collapse
Affiliation(s)
- Jiazeng Sun
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Shang Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Bianhua Shi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Dexian Zheng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Juan Shi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
199
|
Leti F, DiStefano JK. Long Noncoding RNAs as Diagnostic and Therapeutic Targets in Type 2 Diabetes and Related Complications. Genes (Basel) 2017; 8:genes8080207. [PMID: 28829354 PMCID: PMC5575670 DOI: 10.3390/genes8080207] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 01/20/2023] Open
Abstract
Protein-coding genes represent only a small fraction of the human genome. In the past, the majority of the genomic sequence has been considered transcriptionally silent, but recent large-scale studies have uncovered an array of functionally significant elements, including non-protein-coding transcripts, within these noncoding regions of the human genome. Long noncoding RNAs (lncRNAs), a class of noncoding transcripts with lengths >200 nucleotides, are pervasively transcribed in the genome and function as signals, decoys, guides, or scaffolds to regulate gene expression. More than 200 diseases have been associated with dysregulated or dysfunctional lncRNAs, and new associations continue to accumulate in the literature. The role of lncRNAs in the pathogenesis of type 2 diabetes mellitus and related complications has only recently been recognized, but there is already evidence for their involvement in many of the pathophysiological mechanisms underlying the disease. In this review, we summarize the current knowledge of the functions and underlying mechanisms of lncRNA activity with a focus on type 2 diabetes mellitus and related renal and retinal complications of the disease. We also discuss the potential of lncRNAs to serve as therapeutic targets for drug development and diagnostic markers for clinical applications in the management of diabetes.
Collapse
Affiliation(s)
- Fatjon Leti
- Department of Biomedical Research, National Jewish Health, Denver, CO 80210, USA.
| | - Johanna K DiStefano
- Department of Biomedical Research, National Jewish Health, Denver, CO 80210, USA.
| |
Collapse
|
200
|
Jiang X, Zhang F. Long noncoding RNA: a new contributor and potential therapeutic target in fibrosis. Epigenomics 2017; 9:1233-1241. [PMID: 28809130 DOI: 10.2217/epi-2017-0020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is the excess deposition of extracellular matrix components which occur in multiple organs and ultimately leads to organ failure. Long noncoding RNAs (lncRNAs) are a kind of noncoding RNAs longer than approximately 200 nucleotides with no protein-encoding capacity. A growing body of evidence suggests that lncRNAs are also involved in tissues fibrosis in several organs, such as lungs fibrosis, liver fibrosis, renal fibrosis and cardiac fibrosis. In this review, we summarized the current studies of lncRNAs in the process of fibrosis and hopefully aid in better understanding the molecular mechanism of fibrosis and provide a basis to explore new therapeutic targets of fibrosis.
Collapse
Affiliation(s)
- Xiaoying Jiang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Fujun Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| |
Collapse
|