1
|
Li Y, Wang W, Wang Y, Ai H. FTO-mediated m6A demethylation of KLF4 promotes the proliferation and collagen deposition of keloid fibroblasts. Toxicol Res (Camb) 2025; 14:tfaf058. [PMID: 40291597 PMCID: PMC12031721 DOI: 10.1093/toxres/tfaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/22/2024] [Accepted: 02/04/2025] [Indexed: 04/30/2025] Open
Abstract
This study aims to elucidate the molecular mechanism mechanism by which FTO affects fibroblast proliferation and collagen deposition in keloids. Human keloid fibroblasts (KFs) and normal fibroblasts were cultured in vitro. FTO expression was silenced in KFs, and cell viability and proliferation were evaluated via CCK-8 and clone formation assays. FTO, KLF4, and MC1R expressions were quantified via qRT-PCR, while the protein levels of FTO, KLF4, MC1R, Collagen I, and Collagen III were determined by Western blot. The m6A RNA methylation status of total RNA was evaluated using the EpiQuik m6A RNA Methylation Quantification Kit. Post-actinomycin D treatment, the stability of KLF4 mRNA and its m6A modification level were measured. ChIP and dual-luciferase reporter assays confirmed the binding between KLF4 and MC1R promoter. KFs presented with significantly enhanced proliferation and collagen deposition, correlating with elevated FTO expression. Silence of FTO repressed the proliferation and collagen deposition of KFs, and elevated the m6A levels of total RNA and KLF4 mRNA in KFs, resulting in enhanced KLF4 mRNA stability and expression. KLF4 bound to the MC1R promoter and promoted MC1R expression. In conclusion, FTO represses KLF4 expression by removing m6A modification and further diminishes MC1R expression, thereby facilitating KF proliferation and collagen deposition.
Collapse
Affiliation(s)
- Yanqi Li
- Department of Plastic Surgery, Emergency General Hospital/National Research Center for Emergency Medicine, No. 29, Xi Ba He Nan Li, Chaoyang District, Beijing 100020, China
| | - Wanchao Wang
- Department of Plastic Surgery, Emergency General Hospital/National Research Center for Emergency Medicine, No. 29, Xi Ba He Nan Li, Chaoyang District, Beijing 100020, China
| | - Yuge Wang
- Department of Plastic Surgery, Emergency General Hospital/National Research Center for Emergency Medicine, No. 29, Xi Ba He Nan Li, Chaoyang District, Beijing 100020, China
| | - Hongmei Ai
- Department of Plastic Surgery, Emergency General Hospital/National Research Center for Emergency Medicine, No. 29, Xi Ba He Nan Li, Chaoyang District, Beijing 100020, China
| |
Collapse
|
2
|
Wen M, Guo X, Zhang J, Li Y, Li J, Fan Z, Ren W. Non-coding RNA in cartilage regeneration: regulatory mechanism and therapeutic strategies. Front Bioeng Biotechnol 2025; 13:1522303. [PMID: 40206827 PMCID: PMC11979253 DOI: 10.3389/fbioe.2025.1522303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The pathogenesis of cartilage injury and degeneration is exceptionally complex. In addition to being associated with osteoarthritis and trauma, factors such as age, gender, obesity, inflammation, and apoptosis of chondrocytes are also considered significant influencing factors. Due to the lack of direct blood supply, lymphatic circulation, and neural innervation, coupled with low metabolic activity, the self-repair capability of cartilage after injury is extremely limited, making its treatment quite challenging. Recent research indicated that ncRNA, a class of RNA transcribed from the genome that does not encode proteins, played a crucial regulatory role in various disease processes. Particularly noteworthy is its positive regulatory role in cartilage regeneration, achieved through the modulation of the inflammatory microenvironment, promotion of chondrocyte proliferation, inhibition of chondrocyte degradation, and facilitation of the recruitment and differentiation of bone marrow mesenchymal stem cells into chondrocytes. In the earlier phase, we conducted a review and outlook on therapeutic strategies for the regeneration of articular cartilage injuries. This article specifically focuses on summarizing the regulatory roles and research advancements of ncRNA in cartilage regeneration, as well as its contributions to the clinical application of gene therapy for cartilage defects.
Collapse
Affiliation(s)
- Mengnan Wen
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xueqiang Guo
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Jingdi Zhang
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Yunian Li
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jixiang Li
- Junji College of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| | - Zhenlin Fan
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Wenjie Ren
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
3
|
Qin X, Gong H, Jin L, Wang Y, Dang K, Li H, Zheng Q. Long-term glucosamine supplementation aggravates atrial fibrillation susceptibility by impairing AMPK signaling. Life Sci 2025; 362:123380. [PMID: 39788416 DOI: 10.1016/j.lfs.2025.123380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
AIMS Glucosamine, a widely used dietary supplement, has been linked to potential cardiovascular risks, including atrial fibrillation (AF). This study aimed to investigate the effects of long-term glucosamine supplementation on AF susceptibility and the underlying mechanisms. MATERIALS AND METHODS C57BL/6 J mice were treated with low-dose (15 mg/kg/day) or high-dose (250 mg/kg/day) glucosamine via drinking water for 6 weeks. AF susceptibility was assessed through transesophageal electrical stimulation. Atrial remodeling was characterized through electrophysiological and echocardiography studies, histological analysis, and molecular examination. AMP-activated protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR) was used to validation the underlying mechanism in mice and isolated neonatal atrial cardiomyocytes. KEY FINDINGS Long-term high-dose glucosamine supplementation increased AF susceptibility in mice, as indicated by an elevated AF incidence and duration. Glucosamine induced notable electrical remodeling, evidenced by intra-atrial conduction slowing (P wave duration, amplitude, and area), likely attributable to reduced conduction velocity, as confirmed by two-dimensional electrical mapping. Structural remodeling including increased left atrial weight, cardiomyocyte hypertrophy and fibrosis was evident in the atria of glucosamine-treated mice, despite unaffected cardiac function. Mechanistically, glucosamine suppressed atrial AMPK signaling, leading to lipid and glycogen accumulation. Intriguingly, despite impaired atrial AMPK signaling, high-dose glucosamine improved systemic insulin sensitivity. Pharmacological activation of AMPK with AICAR mitigated glucosamine-induced AF susceptibility and associated pathological changes both in vivo and in vitro. SIGNIFICANCE Our findings demonstrate that long-term glucosamine supplementation enhances AF susceptibility, potentially by impairing atrial AMPK signaling, underscoring the importance of caution in the utilization of glucosamine.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| | - Haoyu Gong
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi'an, Shaanxi 710004, China
| | - Lingyan Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi'an, Shaanxi 710004, China
| | - Yixin Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi'an, Shaanxi 710004, China
| | - Kai Dang
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Hui Li
- Department of Cardiology, 986th Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710054, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
4
|
Wang C, Jiang D. Exogenous PRAS40 reduces KLF4 expression and alleviates hypertrophic scar fibrosis and collagen deposition through inhibiting mTORC1. Burns 2024; 50:936-946. [PMID: 38369439 DOI: 10.1016/j.burns.2024.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND To identify the anti-fibrosis effect of PRAS40 in scar, and its potential mechanism. METHODS We constructed a rat model of hypertrophic scarthat was locally injected the PRAS40 overexpression adenoviruses, mTORC1 inhibitor MHY1485 and activator rapamycin, and further observed the pathological changes of skin tissue and the severity of fibrosis by HE, Masson and sirius red staining, and analyzed the deposition of a-SMA and collagen I by western blot and immunofluorescence test. Meanwhile, the co-localization of KLF4 with a-SMA and type I collagen was analyzed, as well as the regulatory effect of PRAS40 on KLF4. In addition, we also verified whether the inhibition of scar fibrosis by PRAS40 is related to mTORC1, and whether the upregulation of KLF4 is related to mTORC1. RESULTS The results showed that the expression of PRAS40 was low and p-PRAS40 was high in scar skin tissue. After local injection of PRAS40 overexpression adenovirus, the expression of PRAS40 in skin tissue was increased. The overexpression of PRAS40 can inhibit scar skin fibrosis and reduce the content of a-SMA and collagen I. Further mechanism analysis confirms that the inhibitory effect of PRAS40 on skin fibrosis is related to mTORC1, and PRAS40 inhibits the activation of mTORC1. The expression of KLF4 is relatively low in scar tissue. PRAS40 administration upregulated the expression of KLF4, which is related to mTORC1 CONCLUSIONS: PRAS40 significantly improves fibrosis of scar skin tissue and increases the expression of KLF4 in scars. The anti-fibrotic effect of PRAS40 depends on mTORC1.
Collapse
Affiliation(s)
- Chao Wang
- Department of Burn and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Duyin Jiang
- Department of Burn and Plastic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
| |
Collapse
|
5
|
Teuma L, Eshwaran R, Tawokam Fongang U, Wieland J, Shao F, Lagana ML, Wang Y, Agaci A, Hammes HP, Feng Y. Glucosamine inhibits extracellular matrix accumulation in experimental diabetic nephropathy. Front Nutr 2022; 9:1048305. [PMID: 36532524 PMCID: PMC9751334 DOI: 10.3389/fnut.2022.1048305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2023] Open
Abstract
INTRODUCTION Glucosamine, the intermediate metabolite of the hexosamine biosynthesis pathway (HBP), is widely used as a supplementary drug in patients with osteoarthritis. However, its consequences in such patients concomitantly suffering from diabetic nephropathy is unknown. METHODS The aim of the study was to investigate the effect of exogenous administration of glucosamine in the diabetic kidney. A mouse model of streptozotocin-induced diabetic nephropathy in vivo and cultured endothelial cells in vitro were used in the study. The mice were treated with glucosamine for 6 months. Renal function was evaluated by metabolic cage, and histology of the kidney was estimated by periodic acid-schiff (PAS) staining. The expression of related genes was assessed by real-time PCR, immunofluorescence staining, immunoblotting and ELISA. RESULTS There was no significant difference in urinary albumin secretion, relative kidney weight, or creatinine clearance between the groups treated with glucosamine and controls. Assessment of the kidney demonstrated reduction in mesangial expansion and fibronectin expression in the diabetic glomeruli treated with glucosamine. Glucosamine treatment significantly decreased α-smooth muscle actin (α-SMA) protein expression in both diabetic and control kidneys, whereas the expression of other fibrosis-related genes and inflammatory factors was unaltered. Moreover, α-SMA colocalized with the endothelial marker CD31 in the diabetic and control kidneys, and glucosamine reduced α-SMA+ ECs in the diabetic glomeruli. In addition, glucosamine suppressed α-SMA expression in endothelial cells treated with or without high glucose. DISCUSSION In summary, this is the first report to show that glucosamine reduces mesangial expansion and inhibits endothelial-mesenchymal transition in diabetic nephropathy. The underlying mechanisms need to be further investigated.
Collapse
Affiliation(s)
- Loic Teuma
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Rachana Eshwaran
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Ulrich Tawokam Fongang
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Johanna Wieland
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Feng Shao
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Maria Luisa Lagana
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Yixin Wang
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Ane Agaci
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Hans-Peter Hammes
- 5th Medical Clinic, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology Mannheim, Medical Faculty Mannheim, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| |
Collapse
|
6
|
Sciriha GG, Sultana J, Borg J. Identifying and categorizing compounds that reduce corneal transforming growth factor beta induced protein levels: a scoping review. Expert Rev Clin Pharmacol 2022; 15:1423-1442. [PMID: 36308770 DOI: 10.1080/17512433.2022.2142560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/27/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Transforming growth factor beta induced (TGFBI) gene mutations have been reported as the cause of a group of genetically inherited, visually debilitating, corneal dystrophies (CD). A scoping literature review to identify and categorize compounds that inhibit corneal TGFBI expression and/or promote TGFBIp degradation was performed. Emphasis was given to their potential to be used as a cost-effective approach via drug repurposing. AREAS COVERED We performed a thorough search of original peer-reviewed literature using electronic bibliographic databases and selected articles according to a set of criteria. The total number of articles retrieved from the search terms applied to the databases was 2344. The number of relevant full-text articles included added up to 19. We identified 16 compounds that can theoretically reduce the levels of mutant TGFBIp in human corneal cells. EXPERT OPINION Currently, the only temporary treatments available for this condition are lubricant drops and surgery. Here, we explored the crosstalk between cascades that regulate TGFBI expression and identified compounds that target these pathways. Compounds that inhibit DNA synthesis and function, increase elimination of TGFBIp or bind to mutant TGFBIp were also explored with the aim of highlighting promising compounds that can be used in future cost-effective drug-repurposing studies.
Collapse
Affiliation(s)
| | - Janet Sultana
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| |
Collapse
|
7
|
Cheng WY, Yang SY, Huang XY, Zi FY, Li HP, Sheng XL. Identification of genetic variants in five chinese families with keratoconus: Pathogenicity analysis and characteristics of parental corneal topography. Front Genet 2022; 13:978684. [PMID: 36276932 PMCID: PMC9583916 DOI: 10.3389/fgene.2022.978684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose: The study aims to identify genetic variants in five Chinese families with Keratoconus (KC) and describe the characteristics of parental corneal topography. Methods: Fifteen participants, including five probands and ten parents from five Chinese families with KC, were recruited for genetic and clinical analyses. Targeted next-generation sequencing using a custom-designed panel for KC was applied on the probands for variant identification. Sanger sequencing and cosegregation analysis of the suspected pathogenic variants were performed on the family members. The pathogenicities of variants were evaluated according to the American College of Medical Genetics and Genomics guidelines (ACMG). Pentacam 3D anterior segment analysis system was applied for keratectasia detection and the Corvis ST for corneal biomechanics measurement. Fifteen parameters were recorded, including nine keratectasia indicators (BAD-D, TP, Kmax, Df, Db, Dp, Dt, Da, ARTH), six corneal biomechanical indicators (CBI, DA ratio, SP-A1, IR, bIOP, TBI). Results: A total of six novel variants, including five missense variants and one frameshift variant, were detected in the HMX1, SLC4A11, TGFBI, PIKFYVE, and ZEB1 genes in five probands, all of which showed co-segregation of genotype and clinical phenotype and were determined to be pathogenic. The genetic model was autosomal dominant (AD) in four families and autosomal recessive (AR) in 1 family. The analysis of keratectasia and corneal biomechanical indicators of the proband’s parents (first-generation relatives) in AD families revealed that there were several abnormal indexes in BAD-D, TP, Kmax, Df, Db, Dp, Dt, Da, CBI, DA ratio, SP-A1, IR, bIOP and TBI test indexes, showing clinical characteristics of incipient KC. Conclusion: Our study shows that variants in HMX1, SLC4A11, TGFBI, PIKFYVE, and ZEB1 were associated with KC. Our study extends the gene spectrum associated with KC, provides novel insights into KC phenotypic assessments, and contributes to early diagnosis for these patients.
Collapse
Affiliation(s)
- Wan-Yu Cheng
- Ningxia Eye Hospital, People’s Hospital of Ningxia Hui Autonomous Region, Third Clinical Medical College of Ningxia Medical University, Yinchuan, China
| | - Shang-Ying Yang
- Ningxia Eye Hospital, People’s Hospital of Ningxia Hui Autonomous Region, Third Clinical Medical College of Ningxia Medical University, Yinchuan, China
| | - Xiao-Yu Huang
- Ningxia Eye Hospital, People’s Hospital of Ningxia Hui Autonomous Region, Third Clinical Medical College of Ningxia Medical University, Yinchuan, China
| | - Fei-Yin Zi
- Ningxia Eye Hospital, People’s Hospital of Ningxia Hui Autonomous Region, Third Clinical Medical College of Ningxia Medical University, Yinchuan, China
| | - Hui-Ping Li
- Ningxia Eye Hospital, People’s Hospital of Ningxia Hui Autonomous Region, Third Clinical Medical College of Ningxia Medical University, Yinchuan, China
- *Correspondence: Hui-Ping Li, ; Xun-Lun Sheng,
| | - Xun-Lun Sheng
- Gansu Aier Ophthalmology and Optometry Hospital, Lanzhou City, China
- *Correspondence: Hui-Ping Li, ; Xun-Lun Sheng,
| |
Collapse
|
8
|
Zhang L, Gao J, Gong A, Dong Y, Hao X, Wang X, Zheng J, Ma W, Song Y, Zhang J, Xu W. The Long Noncoding RNA LINC00963 Inhibits Corneal Fibrosis Scar Formation by Targeting miR-143-3p. DNA Cell Biol 2022; 41:400-409. [PMID: 35262384 PMCID: PMC9063159 DOI: 10.1089/dna.2021.1034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Corneal fibrosis is a complication of severe corneal injury, one of the major causes of vision loss. The formation of myofibroblasts has emerged as a key stimulative factor of corneal fibrosis. In the current study, we focused on the role of LINC00963 in regulating corneal fibrosis. Transforming growth factor β1 (TGF-β1) was used to induce human corneal stromal cells differentiating into corneal myofibroblasts, and the significant increase of α-smooth muscle actin (α-SMA) was verified by quantitative real-time PCR (qRT-PCR), western blot, and immunofluorescence, respectively. LINC00963 was identified to be one-half decreased compared with nonstimulated human corneal stromal cells, indicating that it might play a role in corneal fibrosis. Interestingly, overexpression of LINC00963 resulted in decreased formation of myofibroblasts indicating that it might exhibit an inhibiting effect. Moreover, bioinformatics tool was applied to predict the downstream target of LINC00963. We investigated that LINC00963 suppressed α-SMA induced by TGF-β1 in corneal fibroblasts, at least in part, by downregulating the expression of miR-143-3p. In addition, either LINC00963 promotion or miR-143-3p inhibition could significantly decrease myofibroblast contractility and collagen I and III secretion, which are the key to contribute to corneal fibrosis. Taken together, our study identified LINC00963 as a promising therapeutic target.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China.,Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Anjing Gong
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanhan Dong
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xiaodan Hao
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xuekang Wang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China.,Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jian Zheng
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Wenmeng Ma
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Yiying Song
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Jie Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Wenhua Xu
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Chen Y, Guo H, Li L, Bao D, Gao F, Li Q, Huang Q, Duan X, Xiang Z. Long Non-Coding RNA (lncRNA) Small Nucleolar RNA Host Gene 15 (SNHG15) Alleviates Osteoarthritis Progression by Regulation of Extracellular Matrix Homeostasis. Med Sci Monit 2020; 26:e923868. [PMID: 32643707 PMCID: PMC7370589 DOI: 10.12659/msm.923868] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Growing evidence suggests that long non-coding RNAs (lncRNAs), as decoys of microRNAs (miRNAs), are involved in osteoarthritis (OA) progression, but the potential mechanism of lncRNA SNHG15 in OA remains unknown. Thus, the present study explored the molecular mechanism of SNHG15 in OA progression. MATERIAL AND METHODS OA chondrocytes were created by 20 ng/ml IL-1ß stimulation, and the experimental OA model was created by destabilization of the medial meniscus (DMM) surgery. Cartilage histomorphology was observed by safranin and fast green double dyeing. The relationships between SNHG15 and miR-7, KLF4, and miR-7 were determined by dual-luciferase assay or RNA immunoprecipitation (RIP). Immunofluorescence was used to detect the expressions of Ki67, collagen II, and Aggrecan. Moreover, SNHG15, miR-7, KLF4, MMP3, ADAMTS5, COL2A1, Aggrecan, and ß-catenin expressions were assessed by qRT-PCR or Western blot. The methylation status of SNHG15 promoter was evaluated by MS-PCR. RESULTS Underexpression of KLF4 and SHNG15 and overexpression of miR-7 were found in human OA knee cartilage tissues and IL-1ß-stimulated OA chondrocytes. SHNG15 overexpression significantly inhibited ECM degradation and promoted chondrocyte formation of OA chondrocytes. Furthermore, SNHG15 regulated KLF4 expression by sponging miR-7. Further analysis found that SNHG15 significantly inhibited b-catenin in OA chondrocytes. SNHG15 had a higher level of methylation in human OA tissues than in normal cartilage tissues. CONCLUSIONS Our results revealed that SNHG15 alleviated OA progression by regulating ECM homeostasis, which provides a promising target for OA therapy.
Collapse
Affiliation(s)
- Yunping Chen
- Department of Clinical Laboratory Medicine, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Hongna Guo
- Department of Clinical Laboratory Medicine, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Lang Li
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland).,Department of Orthopedics, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, China (mainland)
| | - Dingsu Bao
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland).,Department of Orthopedics, Hospital of Traditional Chinese Medicine Affiliated to Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Feng Gao
- Department of Orthopedics, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, China (mainland)
| | - Qiang Li
- Department of Orthopedics, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, China (mainland)
| | - Qi Huang
- Department of Orthopedics, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, China (mainland)
| | - Xin Duan
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Zhou Xiang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|