51
|
Wang XB, Wang H, Long HQ, Li DY, Zheng X. LINC00641 regulates autophagy and intervertebral disc degeneration by acting as a competitive endogenous RNA of miR-153-3p under nutrition deprivation stress. J Cell Physiol 2018; 234:7115-7127. [PMID: 30378116 DOI: 10.1002/jcp.27466] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 08/29/2018] [Indexed: 12/13/2022]
Abstract
Emerging evidence supports the involvement of autophagy in the pathogenesis of intervertebral disc degeneration (IDD). MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) play fundamental roles in various cellular processes, including autophagy. However, it remains largely unknown as to how autophagy is regulated by miRNAs and lncRNAs in IDD. Biological functions of miR-153-3p and long intergenic nonprotein coding RNA 641 (LINC00641) were investigated. Luciferase reporter assays was done to validate miR-153-3p targets. To induce nutritional stress, nucleus pulposus (NP) cells were cultured in the normal nutritional condition and the low nutritional condition. Quantitative reverse-transcription polymerase chain reaction (RT-qPCR) was used to analyze miR-153-3p and LINC00641 in response to nutrient deprivation. Autophagic activity was assessed by transmission electron microscopy, western blot analysis and green fluorescent protein-light chain 3 puncta. Pull-down assay and RNA fluorescent in situ hybridization were performed to validate LINC00641 target and the location. MiR-153-3p is downregulated in NP tissues from IDD patients. Further, LINC00641 can affect collagen II and matrix metalloproteinase-3 expressions. Upregulation of LINC00641 and downregulation of miR-153-3p are detected in NP cells under nutritional stress. LINC00641 can regulate autophagic cell death by targeting miR-153-3p and autophagy-related gene 5 (ATG5). MiR-153-3p inhibits autophagy and IDD by targeting ATG5. More important, LINC00641 targets miR-153-3p, and thus affects ATG5 expression, autophagic cell death and IDD. These findings uncover a novel regulatory pathway that is composed of LINC00641, miR-153-3p, and ATG5 in IDD. This mechanism may stimulate to a more understanding of IDD pathogenesis and provide new sights for the treatment of this disorder.
Collapse
Affiliation(s)
- Xiao-Bo Wang
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hua Wang
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hou-Qing Long
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dong-Ya Li
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xin Zheng
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
52
|
Inhibition of miR-449a Promotes Cartilage Regeneration and Prevents Progression of Osteoarthritis in In Vivo Rat Models. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:322-333. [PMID: 30326428 PMCID: PMC6197768 DOI: 10.1016/j.omtn.2018.09.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 09/21/2018] [Accepted: 09/23/2018] [Indexed: 12/20/2022]
Abstract
Traumatic and degenerative lesions of articular cartilage usually progress to osteoarthritis (OA), a leading cause of disability in humans. MicroRNAs (miRNAs) can regulate the differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) and play important roles in the expression of genes related to OA. However, their functional roles in OA remain poorly understood. Here, we have examined miR-449a, which targets sirtuin 1 (SIRT1) and lymphoid enhancer-binding factor-1 (LEF-1), and observed its effects on damaged cartilage. The levels of chondrogenic markers and miR-449a target genes increased during chondrogenesis in anti-miR-449a-transfected hBMSCs. A locked nucleic acid (LNA)-anti-miR-449a increased cartilage regeneration and expression of type II collagen and aggrecan on the regenerated cartilage surface in acute defect and OA models. Furthermore, intra-articular injection of LNA-anti-miR-449a prevented disease progression in the OA model. Our study indicates that miR-449a may be a novel potential therapeutic target for age-related joint diseases like OA.
Collapse
|
53
|
Wang H, Zhang H, Sun Q, Yang J, Zeng C, Ding C, Cai D, Liu A, Bai X. Chondrocyte mTORC1 activation stimulates miR‐483‐5p via HDAC4 in osteoarthritis progression. J Cell Physiol 2018; 234:2730-2740. [PMID: 30145794 DOI: 10.1002/jcp.27088] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/28/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Hua Wang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical CollegeHaikou China
- Department of Cell BiologySchool of Basic Medical Science, Southern Medical UniversityGuangzhou China
| | - Haiyan Zhang
- Department of OrthopedicsAcademy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical UniversityGuangzhou China
| | - Qiuyi Sun
- Department of Cell BiologySchool of Basic Medical Science, Southern Medical UniversityGuangzhou China
| | - Jian Yang
- Department of Biomedical EngineeringMaterials Research Institute, The Huck Institutes of The Life Sciences, The Pennsylvania State University, University ParkPA USA
| | - Chun Zeng
- Department of OrthopedicsAcademy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical UniversityGuangzhou China
| | - Changhai Ding
- Department of Cell BiologySchool of Basic Medical Science, Southern Medical UniversityGuangzhou China
| | - Daozhang Cai
- Department of OrthopedicsAcademy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical UniversityGuangzhou China
| | - Anling Liu
- Department of BiochemistrySchool of Basic Medical Science, Southern Medical UniversityGuangzhou China
| | - Xiaochun Bai
- Department of Cell BiologySchool of Basic Medical Science, Southern Medical UniversityGuangzhou China
| |
Collapse
|
54
|
Xiao L, Xu S, Xu Y, Liu C, Yang B, Wang J, Xu H. TGF-β/SMAD signaling inhibits intermittent cyclic mechanical tension-induced degeneration of endplate chondrocytes by regulating the miR-455-5p/RUNX2 axis. J Cell Biochem 2018; 119:10415-10425. [PMID: 30132981 DOI: 10.1002/jcb.27391] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/09/2018] [Indexed: 12/30/2022]
Abstract
A mechanical stimulation plays a pivotal role in maintaining normal cartilage function. Our objective was to reveal the mechanism of action of the tension-sensitive molecule miR-455-5p in the degeneration of endplate chondrocytes and to identify whether the transforming growth factor beta (TGF-β)/SMAD signaling pathway has a regulatory effect on it. The expression profiles of members of the TGF-β/SMAD pathway, miR-455-5p, and RUNX2 were determined by microRNA microarray analysis, reverse transcription quantitative polymerase chain reaction, luciferase reporter assay, and Western blot analysis. Intermittent cyclic mechanical tension (ICMT) induced the degeneration of endplate chondrocytes without affecting their viability. The tension-sensitive molecule miR-455-5p specifically bound to RUNX2, a gene involved in the degeneration of endplate chondrocytes. Activation of the TGF-β/SMAD signaling pathway upregulated miR-455-5p expression and thus inhibited RUNX2 levels. Therefore, the TGF-β/SMAD signaling pathway inhibits the ICMT-induced degeneration of endplate chondrocytes by regulating the miR-455-5p/RUNX2 axis.
Collapse
Affiliation(s)
- Liang Xiao
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Shujuan Xu
- Department of Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yongming Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Chen Liu
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Bijing Yang
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Jing Wang
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hongguang Xu
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
55
|
Onset and Progression of Human Osteoarthritis-Can Growth Factors, Inflammatory Cytokines, or Differential miRNA Expression Concomitantly Induce Proliferation, ECM Degradation, and Inflammation in Articular Cartilage? Int J Mol Sci 2018; 19:ijms19082282. [PMID: 30081513 PMCID: PMC6121276 DOI: 10.3390/ijms19082282] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative whole joint disease, for which no preventative or therapeutic biological interventions are available. This is likely due to the fact that OA pathogenesis includes several signaling pathways, whose interactions remain unclear, especially at disease onset. Early OA is characterized by three key events: a rarely considered early phase of proliferation of cartilage-resident cells, in contrast to well-established increased synthesis, and degradation of extracellular matrix components and inflammation, associated with OA progression. We focused on the question, which of these key events are regulated by growth factors, inflammatory cytokines, and/or miRNA abundance. Collectively, we elucidated a specific sequence of the OA key events that are described best as a very early phase of proliferation of human articular cartilage (AC) cells and concomitant anabolic/catabolic effects that are accompanied by incipient pro-inflammatory effects. Many of the reviewed factors appeared able to induce one or two key events. Only one factor, fibroblast growth factor 2 (FGF2), is capable of concomitantly inducing all key events. Moreover, AC cell proliferation cannot be induced and, in fact, is suppressed by inflammatory signaling, suggesting that inflammatory signaling cannot be the sole inductor of all early OA key events, especially at disease onset.
Collapse
|
56
|
Panagopoulos PK, Lambrou GI. The Involvement of MicroRNAs in Osteoarthritis and Recent Developments: A Narrative Review. Mediterr J Rheumatol 2018; 29:67-79. [PMID: 32185303 PMCID: PMC7046075 DOI: 10.31138/mjr.29.2.67] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/12/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background: Osteoarthritis (OA) is the most common chronic joint disease and it may progressively cause disability and compromise quality of life. Lately, the role of miRNAs in the pathogenesis of OA has drawn a lot of attention. miRNAs are small, single-stranded, non-coding molecules of RNA which regulate gene expression at post-transcriptional level. The dysregulation of the expression of several miRNAs affects pathways involved in OA pathogenesis. Objective: The purpose of this article is to review the literature on the involvement of miRNAs in the pathogenesis of OA and the implications on its diagnosis and treatment. Materials and Methods: An extensive electronic literature search was conducted by two researchers from January 2008 to August 2017. Titles and abstracts of papers were screened by the authors for further inclusion in the present work. Finally, full texts of the selected articles were retrieved. Results: Abnormally expressed miRNAs enhance the production of cartilage degrading enzymes, inhibit the expression of cartilage matrix components, increase the production of proinflammatory cytokines, facilitate chondrocyte apoptosis, suppress autophagy in chondrocytes and are involved in pain-related pathways. miRNAs are also incorporated in extra-cellular membranous vesicles such as exosomes and participate in the intercellular communication in osteoarthritic joints. Conclusion: Ongoing research on miRNAs has potential implications in the diagnosis and treatment of OA. Their different levels in peripheral blood and synovial fluid between OA patients and healthy population makes them candidates for being used as biomarkers of the disease, while targeting miRNAs may be a novel therapeutic strategy in OA.
Collapse
Affiliation(s)
- Panagiotis K Panagopoulos
- Postgraduate Program "Metabolic Bone Diseases", National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - George I Lambrou
- Postgraduate Program "Metabolic Bone Diseases", National and Kapodistrian University of Athens, Medical School, Athens, Greece.,First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| |
Collapse
|
57
|
Rai MF, Pham CT. Intra-articular drug delivery systems for joint diseases. Curr Opin Pharmacol 2018; 40:67-73. [PMID: 29625332 DOI: 10.1016/j.coph.2018.03.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/24/2018] [Indexed: 12/20/2022]
Abstract
Intra-articular (IA) injections directly deliver high concentrations of therapeutics to the joint space and are routinely used in various musculoskeletal conditions such as osteoarthritis (OA) and rheumatoid arthritis (RA). However, current IA-injected drugs are rapidly cleared and do not significantly affect the course of joint disease. In this review, we highlight recent developments in IA therapy, with a special emphasis on current and emerging therapeutic carriers and their potential to deliver disease-modifying treatment modalities for arthritis. Recent IA approaches concentrate on platforms that are safe with efficient tissue penetration, and readily translatable for controlled and sustained delivery of therapeutic agents. Gene therapy delivered by viral or non-viral vectors and cell-based therapy for cartilage preservation and regeneration are being intensively explored.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, 660 South Euclid Avenue, Box 8233, Saint Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8233, Saint Louis, MO 63110, USA.
| | - Christine Tn Pham
- Department of Medicine, Division of Rheumatology, 660 South Euclid Avenue, Box 8045, Saint Louis, MO 63110, USA.
| |
Collapse
|
58
|
Grol MW, Lee BH. Gene therapy for repair and regeneration of bone and cartilage. Curr Opin Pharmacol 2018; 40:59-66. [PMID: 29621661 DOI: 10.1016/j.coph.2018.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/12/2018] [Indexed: 12/28/2022]
Abstract
Gene therapy refers to the use of viral and non-viral vectors to deliver nucleic acids to tissues of interest using direct (in vivo) or transduced cell-mediated (ex vivo) approaches. Over the past few decades, strategies have been adopted to express therapeutic transgenes at sites of injury to promote or facilitate repair of bone and cartilage. Targets of interest have typically included secreted proteins such as growth factors and anti-inflammatory mediators; however, work has also begun to focus intracellularly on signaling components, transcription factors and small, regulatory nucleic acids such as microRNAs (miRNAs). In recent years, a number of single therapeutic gene approaches (termed 'monotherapies') have proven effective in preclinical models of disease, and several are being evaluated in clinical trials. In particular, an ex vivo TGF-β1 gene therapy was approved in Korea in 2017 for treatment of moderate-to-severe osteoarthritis (OA). The ability to utilize viral vectors for context-specific and combinatorial gene therapy is also being investigated, and these strategies are likely to be important in more robustly addressing the complexities of tissue repair and regeneration in skeletal disease. In this review, we provide an overview of viral gene therapies being developed for treatment of bone and cartilage pathologies, with an emphasis on emerging combinatorial strategies as well as those targeting intracellular mediators such as miRNAs.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
59
|
Trachana V, Ntoumou E, Anastasopoulou L, Tsezou A. Studying microRNAs in osteoarthritis: Critical overview of different analytical approaches. Mech Ageing Dev 2018; 171:15-23. [DOI: 10.1016/j.mad.2018.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/14/2018] [Accepted: 02/25/2018] [Indexed: 12/14/2022]
|
60
|
Abstract
PURPOSE OF REVIEW Epigenomics has emerged as a key player in our rapidly evolving understanding of osteoarthritis. Historical studies implicated epigenetic alterations, particularly DNA methylation, in OA pathogenesis; however, recent technological advances have resulted in numerous epigenome-wide studies examining in detail epigenetic modifications in OA. The purpose of this article is to introduce basic concepts in epigenetics and their recent applications to the study of osteoarthritis development and progression. RECENT FINDINGS Epigenetics describes three major phenomena: DNA modification via methylation, histone sidechain modifications, and short noncoding RNA sequences which work in concert to regulate gene transcription in a heritable fashion. Cartilage has been the most widely studied tissue in OA, and differential methylation of genes involved in inflammation, cell cycle, TGFβ, and HOX genes have been confirmed several times. Bone studies suggest similar findings, and the intriguing possibility of epigenetic changes in subchondral bone during many OA processes. Multiple studies have demonstrated the involvement of certain noncoding RNAs, particularly miR-140, in OA development via modulation of key catabolic factors. Although much work has been done, much is still unknown. Future epigenomic studies will no doubt continue to widen our understanding of extraarticular tissues and OA pathogenesis, and studies in animal models may offer glimpses into epigenome alterations in the earliest stages of OA.
Collapse
|
61
|
夏 颖, 周 雪, 古 文, 赵 岩, 肖 潇, 白 晓, 刘 俊, 李 明. [A method for efficient transduction of miR-483-5p in the kidney of mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:141-147. [PMID: 29502051 PMCID: PMC6743870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To establish a method for gene delivery in murine renal tissue using lentivirus vector encoding miR-483-5p. METHODS Thirty-five C57BL/6J mice were randomly divided into control group, low-dose treatment group (5 µL each kidney) , and high?dose treatment group (20 µL each kidney), and in the latter two groups, the lentivirus vector encoding miR-483-5p were injected in the renal cortex. The tissue samples were collected at 7 and 21 days after the injection. A transgenic mouse model with inducible systemic overexpression of miR-483-5p was established in TG483 mice. The Cre-loxp system was used to create a mouse model with renal tubule-specific expression of miR-483-5p. The levels of BUN in the mice were detected and HE staining and fluorometric TUNEL assay were used to observe the morphological changes of the kidneys; real-time qPCR was used to detect miR-483-5p expression in the renal cortex. RESULTS The mice with overexpression of miR-483-5p had normal renal function without obvious pathological changes or apoptosis in the renal tissue. Renal cortex injection of 20 µL lentivirus resulted in obviously increased level of miR-483-5p at 21 days (1.2∓0.43 vs 8.6∓1.09, P<0.001). miR-483-5p showed a low expression (0.9∓0.09 vs 1.7∓0.19, P<0.05) in TG483 mice and a high expression in the kidney of the transgenic mice established using the Cre-loxp system (1.6∓1.13 vs 12.36∓3.89, P<0.05). CONCLUSION The transgenic mice with renal tubule-specific expression of miR-483-5p show normal renal function, and this model facilitates further study of the role of miR-483-5p in the kidney.
Collapse
Affiliation(s)
- 颖 夏
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 雪娟 周
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 文清 古
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 岩岩 赵
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 潇 肖
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 晓春 白
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 俊 刘
- 广州军区广州总医院泌尿外科,广东 广州 510010Department of Urology, General Hospital of Guangzhou Military Region, Guangzhou 510010, China
| | - 明 李
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
62
|
夏 颖, 周 雪, 古 文, 赵 岩, 肖 潇, 白 晓, 刘 俊, 李 明. [A method for efficient transduction of miR-483-5p in the kidney of mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:141-147. [PMID: 29502051 PMCID: PMC6743870 DOI: 10.3969/j.issn.1673-4254.2018.02.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To establish a method for gene delivery in murine renal tissue using lentivirus vector encoding miR-483-5p. METHODS Thirty-five C57BL/6J mice were randomly divided into control group, low-dose treatment group (5 µL each kidney) , and high?dose treatment group (20 µL each kidney), and in the latter two groups, the lentivirus vector encoding miR-483-5p were injected in the renal cortex. The tissue samples were collected at 7 and 21 days after the injection. A transgenic mouse model with inducible systemic overexpression of miR-483-5p was established in TG483 mice. The Cre-loxp system was used to create a mouse model with renal tubule-specific expression of miR-483-5p. The levels of BUN in the mice were detected and HE staining and fluorometric TUNEL assay were used to observe the morphological changes of the kidneys; real-time qPCR was used to detect miR-483-5p expression in the renal cortex. RESULTS The mice with overexpression of miR-483-5p had normal renal function without obvious pathological changes or apoptosis in the renal tissue. Renal cortex injection of 20 µL lentivirus resulted in obviously increased level of miR-483-5p at 21 days (1.2∓0.43 vs 8.6∓1.09, P<0.001). miR-483-5p showed a low expression (0.9∓0.09 vs 1.7∓0.19, P<0.05) in TG483 mice and a high expression in the kidney of the transgenic mice established using the Cre-loxp system (1.6∓1.13 vs 12.36∓3.89, P<0.05). CONCLUSION The transgenic mice with renal tubule-specific expression of miR-483-5p show normal renal function, and this model facilitates further study of the role of miR-483-5p in the kidney.
Collapse
Affiliation(s)
- 颖 夏
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 雪娟 周
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 文清 古
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 岩岩 赵
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 潇 肖
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 晓春 白
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - 俊 刘
- 广州军区广州总医院泌尿外科,广东 广州 510010Department of Urology, General Hospital of Guangzhou Military Region, Guangzhou 510010, China
| | - 明 李
- 南方医科大学基础医学院细胞生物学教研室,广东 广州 510515Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
63
|
Zhou ZB, Du D, Huang GX, Chen A, Zhu L. Circular RNA Atp9b, a competing endogenous RNA, regulates the progression of osteoarthritis by targeting miR-138-5p. Gene 2018; 646:203-209. [PMID: 29305974 DOI: 10.1016/j.gene.2017.12.064] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/21/2017] [Accepted: 12/29/2017] [Indexed: 10/18/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease and is mainly characterized by degradation of the articular cartilage. Recently, circular RNAs (circRNAs), novel noncoding RNAs with different biological functions and pathological implications, have been reported to be closely associated with various diseases. Growing evidence indicates that circRNAs act as competing endogenous RNAs (ceRNAs) that bind with microRNAs (miRNAs) and regulate their downstream functions. Here, we identified a new circRNA, circRNA_Atp9b, and further investigated its function in OA using a well-established mouse chondrocyte model. We demonstrated that circRNA_Atp9b expression was significantly up-regulated in mouse chondrocytes after stimulation with interleukin-1 beta (IL-1β), and that knockdown of circRNA_Atp9b promoted the expression of type II collagen while inhibiting the generation of MMP13, COX-2 and IL-6. Moreover, there was a negative correlation between the expression levels of circRNA_Atp9b and microRNA (miR)-138-5p, indicating that miR-138-5p also played a role in IL-1β-induced chondrocytes. Bioinformatics analysis predicted circRNA_Atp9b directly target miR-138-5p, which was validated by dual-luciferase assay. Further functional experiments revealed that down-regulation of miR-138-5p partly reversed the effects of circRNA_Atp9b on extracellular matrix (ECM) catabolism and inflammation. Taken together, these results suggest that circRNA_Atp9b regulates OA progression by modulating ECM catabolism and inflammation in chondrocytes via sponging miR-138-5p. Our findings provide novel insight into the regulatory mechanism of circRNA_Atp9b in OA and may contribute to establishing potential therapeutic strategies.
Collapse
Affiliation(s)
- Zhi-Bin Zhou
- Department of Orthopaedic Trauma Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Di Du
- Department of Orthopaedic Trauma Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Gao-Xiang Huang
- Department of Pathology, No. 181 Hospital of PLA, Guilin, Guangxi 541002, China
| | - Aimin Chen
- Department of Orthopaedic Trauma Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Lei Zhu
- Department of Orthopaedic Trauma Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| |
Collapse
|
64
|
Qu R, Chen X, Wang W, Qiu C, Ban M, Guo L, Vasilev K, Chen J, Li W, Zhao Y. Ghrelin protects against osteoarthritis through interplay with Akt and NF‐κB signaling pathways. FASEB J 2018; 32:1044-1058. [DOI: 10.1096/fj.201700265r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ruize Qu
- Department of PathologyShandong UniversityJinanChina
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Xiaomin Chen
- Department of PathologyShandong UniversityJinanChina
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Wenhan Wang
- Department of OrthopedicsQilu HospitalShandong UniversityJinanChina
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Cheng Qiu
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Miaomiao Ban
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Linlin Guo
- Medical School of Shandong UniversityShandong UniversityJinanChina
| | - Krasimir Vasilev
- School of EngineeringUniversity of South AustraliaMawson LakesSouth AustraliaAustralia
| | - Jianying Chen
- Institute of Biopharmaceuticals of Shandong ProvinceJinanChina
| | - Weiwei Li
- Department of PathologyShandong UniversityJinanChina
| | - Yunpeng Zhao
- Department of OrthopedicsQilu HospitalShandong UniversityJinanChina
| |
Collapse
|
65
|
McAlinden A, Im GI. MicroRNAs in orthopaedic research: Disease associations, potential therapeutic applications, and perspectives. J Orthop Res 2018; 36:33-51. [PMID: 29194736 PMCID: PMC5840038 DOI: 10.1002/jor.23822] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function to control many cellular processes by their ability to suppress expression of specific target genes. Tens to hundreds of target genes may be affected by one miRNA, thereby resulting in modulation of multiple pathways in any given cell type. Therefore, altered expression of miRNAs (i.e., during tissue development or in scenarios of disease or cellular stress) can have a profound impact on processes regulating cell differentiation, metabolism, proliferation, or apoptosis, for example. Over the past 5-10 years, thousands of reports have been published on miRNAs in cartilage and bone biology or disease, thus highlighting the significance of these non-coding RNAs in regulating skeletal development and homeostasis. For the purpose of this review, we will focus on miRNAs or miRNA families that have demonstrated function in vivo within the context of cartilage, bone or other orthopaedic-related tissues (excluding muscle). Specifically, we will discuss studies that have utilized miRNA transgenic mouse models or in vivo approaches to target a miRNA with the aim of altering conditions such as osteoarthritis, osteoporosis and bone fractures in rodents. We will not discuss miRNAs in the context skeletal cancers since this topic is worthy of a review of its own. Overall, we aim to provide a comprehensive description of where the field currently stands with respect to the therapeutic potential of specific miRNAs to treat orthopaedic conditions and current technologies to target and modify miRNA function in vivo. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:33-51, 2018.
Collapse
Affiliation(s)
- Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, Missouri 63110
| | - Gun-Il Im
- Department of Orthopaedic Surgery, Dongguk University Ilsan Hospital, 814 Siksa-Dong, Goyang, Korea
| |
Collapse
|
66
|
Anderson BA, McAlinden A. miR-483 targets SMAD4 to suppress chondrogenic differentiation of human mesenchymal stem cells. J Orthop Res 2017; 35:2369-2377. [PMID: 28244607 PMCID: PMC5573664 DOI: 10.1002/jor.23552] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/18/2017] [Indexed: 02/04/2023]
Abstract
MicroRNAs (miRNAs) can regulate cellular differentiation processes by modulating multiple pathways simultaneously. Previous studies to analyze in vivo miRNA expression patterns in developing human limb cartilage tissue identified significant downregulation of miR-483 in hypertrophic chondrocytes relative to proliferating and differentiated chondrocytes. To test the function of miR-483 during chondrogenesis, lentiviral strategies were used to overexpress miR-483 during in vitro chondrogenesis of human bone marrow-derived mesenchymal stem cells (hBM-MSCs). While the in vivo expression patterns led us to hypothesize that miR-483 may enhance chondrogenesis or suppress hypertrophic marker expression, surprisingly, miR-483 overexpression reduced chondrocyte gene expression and cartilage matrix production. In addition, cell death was induced at later stages of the chondrogenesis assay. Mechanistic studies revealed that miR-483 overexpression resulted in downregulation of the TGF-β pathway member SMAD4, a known direct target of miR-483-3p. From these studies, we conclude that constitutive overexpression of miR-483 in hBM-MSCs inhibits chondrogenesis of these cells and does not represent an effective strategy to attempt to enhance chondrocyte differentiation and anabolism in this system in vitro. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2369-2377, 2017.
Collapse
Affiliation(s)
- Britta A. Anderson
- Department of Orthopaedic Surgery, Washington University School of Medicine, 600 S. Euclid Ave., St. Louis, MO 63110
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, 600 S. Euclid Ave., St. Louis, MO 63110,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO,Corresponding author:: , Phone: (314) 454-8860
| |
Collapse
|
67
|
Abstract
Although the potential effect of aberrant expression of catabolic and
anabolic genes on the development of osteoarthritis (OA) is well-documented, the
regulatory mechanism for the expression of these genes in articular chondrocytes
remains to be elucidated. The recent advances in epigenetic studies have
identified microRNA (miRNA) as one of the epigenetic mechanisms for the
regulation of gene expression. This mini review highlights the role of miRNA in
the regulation of gene expression in articular chondrocytes and its significance
in the pathogenesis of OA, with a discussion on the potential of miRNA as a new
biomarker and therapeutic target for OA. Further investigations are required to
determine the specificity, sensitivity, and efficacy of miRNA for clinical
applications.
Collapse
Affiliation(s)
- Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kate Lygrisse
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|