1
|
Xu Y, Shi X, Lin H, Li S, Zhang Z, Wei F, Chen Y. GelMA/HA-NB hydrogel encapsulating adipose-derived chondrogenic exosomes enhances enthesis regeneration in chronic rotator cuff tears. Int J Biol Macromol 2025; 309:142800. [PMID: 40185430 DOI: 10.1016/j.ijbiomac.2025.142800] [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: 01/17/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Chronic rotator cuff tears (RCTs) often lead to poor surgical outcomes, requiring innovative therapies. This study explores the potential of exosomes from chondrogenic stem/progenitor cells (CSPCs), encapsulated in a GelMA/HA-NB hydrogel, to improve rotator cuff healing. Adipose-derived stem cells (ASCs) were isolated and sorted to obtain CSPCs, from which exosomes (sub-Exos) were extracted and characterized. Unsorted ASCs exosomes (un-Exos) were also isolated for comparison. The hydrogel-exosome system was evaluated for biocompatibility, chondrogenic differentiation, and sustained release in vitro and in a chronic RCT rat model. 112 rats were divided into four groups: control, hydrogel alone, un-Exos with hydrogel, and sub-Exos with hydrogel. Healing was assessed at 4 and 8 weeks using micro-CT, histology, and biomechanical testing. In vitro, sub-Exos with hydrogel demonstrated excellent biocompatibility and enhanced chondrogenic potential. In vivo, sub-Exos were retained at the injury site for up to 14 days, significantly improving histological scores, bone mineral density, bone volume/total volume, and trabecular thickness. Biomechanical tests revealed superior failure load and stiffness in the sub-Exos group. These findings demonstrate that localized delivery of GelMA/HA-NB hydrogel-encapsulated sub-Exos significantly enhances enthesis healing, offering a promising cell-free therapeutic strategy for chronic RCTs.
Collapse
Affiliation(s)
- Yan Xu
- Department of Orthopaedics, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, China
| | - Xin Shi
- Department of Orthopaedics, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, China; Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Haofeng Lin
- Department of Orthopaedics, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, China
| | - Siqi Li
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Zhiyuan Zhang
- Department of Orthopaedics, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, China
| | - Fuxin Wei
- Department of Orthopaedics, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, China.
| | - Yang Chen
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China; Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
2
|
An S, Intini C, O'Shea D, Dixon JE, Zheng Y, O'Brien FJ. A miR-activated hydrogel for the delivery of a pro-chondrogenic microRNA-221 inhibitor as a minimally invasive therapeutic approach for articular cartilage repair. Mater Today Bio 2025; 30:101382. [PMID: 39759843 PMCID: PMC11699623 DOI: 10.1016/j.mtbio.2024.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Articular cartilage has limited capacity for repair (or for regeneration) under pathological conditions, given its non-vascularized connective tissue structure and low cellular density. Our group has successfully developed an injectable hydrogel for cartilage repair, composed of collagen type I (Col I), collagen type II (Col II), and methacrylated-hyaluronic acid (MeHA), capable of supporting chondrogenic differentiation of mesenchymal stem cells (MSCs) towards articular cartilage-like phenotypes. Recent studies have demonstrated that silencing miR-221 may be an effective approach in promoting improved MSC chondrogenesis. Thus, this study aimed to develop a miR-activated hydrogel capable of offering a more effective and less invasive therapeutic approach to articular cartilage repair by delivering a pro-chondrogenic miR-221 inhibitor to MSCs using our MeHA-Col I/Col II hydrogel. The MeHA-Col I/Col II hydrogel was cast as previously shown and incorporated with cells transfected with miR-221 inhibitor (using a non-viral peptide delivery vector) to produce the miR-activated hydrogel. Down-regulation of miR-221 did not affect cell viability and enhanced MSCs-mediated chondrogenesis, as evidenced by significantly upregulated expression of key pro-chondrogenic articular cartilage genes (COL2A1 and ACAN) without promoting hypertrophic events (RUNX2 and COL10A1). Furthermore, miR-221 down-regulation improved cartilage-like matrix formation in the MeHA-Col I/Col II hydrogel, with significantly higher levels of sulfated glycosaminoglycans (sGAG) and Col II produced by MSCs in the hydrogel. These results provide evidence of the potential of the miR-activated hydrogel as a minimally invasive therapeutic strategy for articular cartilage repair.
Collapse
Affiliation(s)
- Shan An
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- College of Pharmaceutical Sciences, Soochow University, China
| | - Claudio Intini
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Donagh O'Shea
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - James E. Dixon
- Regenerative Medicine & Cellular Therapies (RMCT), Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, China
| | - Fergal J. O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, TCD, Ireland
| |
Collapse
|
3
|
Kwon DG, Kim MK, Jeon YS, Nam YC, Park JS, Ryu DJ. State of the Art: The Immunomodulatory Role of MSCs for Osteoarthritis. Int J Mol Sci 2022; 23:1618. [PMID: 35163541 PMCID: PMC8835711 DOI: 10.3390/ijms23031618] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) has generally been introduced as a degenerative disease; however, it has recently been understood as a low-grade chronic inflammatory process that could promote symptoms and accelerate the progression of OA. Current treatment strategies, including corticosteroid injections, have no impact on the OA disease progression. Mesenchymal stem cells (MSCs) based therapy seem to be in the spotlight as a disease-modifying treatment because this strategy provides enlarged anti-inflammatory and chondroprotective effects. Currently, bone marrow, adipose derived, synovium-derived, and Wharton's jelly-derived MSCs are the most widely used types of MSCs in the cartilage engineering. MSCs exert immunomodulatory, immunosuppressive, antiapoptotic, and chondrogenic effects mainly by paracrine effect. Because MSCs disappear from the tissue quickly after administration, recently, MSCs-derived exosomes received the focus for the next-generation treatment strategy for OA. MSCs-derived exosomes contain a variety of miRNAs. Exosomal miRNAs have a critical role in cartilage regeneration by immunomodulatory function such as promoting chondrocyte proliferation, matrix secretion, and subsiding inflammation. In the future, a personalized exosome can be packaged with ideal miRNA and proteins for chondrogenesis by enriching techniques. In addition, the target specific exosomes could be a gamechanger for OA. However, we should consider the off-target side effects due to multiple gene targets of miRNA.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong Jin Ryu
- Orthopedic Surgery, Inha University Hospital, 22332 Inhang-ro 27, Jung-gu, Incheon 22332, Korea; (D.G.K.); (M.K.K.); (Y.S.J.); (Y.C.N.); (J.S.P.)
| |
Collapse
|
4
|
Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP, Law JX. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals (Basel) 2021; 14:1093. [PMID: 34832875 PMCID: PMC8618513 DOI: 10.3390/ph14111093] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are the small extracellular vesicles secreted by cells for intercellular communication. Exosomes are rich in therapeutic cargos such as microRNA (miRNA), long non-coding RNA (lncRNA), small interfering RNA (siRNA), DNA, protein, and lipids. Recently, many studies have focused on miRNAs as a promising therapeutic factor to support cartilage regeneration. Exosomes are known to contain a substantial amount of a variety of miRNAs. miRNAs regulate the post-transcriptional gene expression by base-pairing with the target messenger RNA (mRNA), leading to gene silencing. Several exosomal miRNAs have been found to play a role in cartilage regeneration by promoting chondrocyte proliferation and matrix secretion, reducing scar tissue formation, and subsiding inflammation. The exosomal miRNA cargo can be modulated using techniques such as cell transfection and priming as well as post-secretion modifications to upregulate specific miRNAs to enhance the therapeutic effect. Exosomes are delivered to the joints through direct injection or via encapsulation within a scaffold for sustained release. To date, exosome therapy for cartilage injuries has yet to be optimized as the ideal cell source for exosomes, and the dose and method of delivery have yet to be identified. More importantly, a deeper understanding of the role of exosomal miRNAs in cartilage repair is paramount for the development of more effective exosome therapy.
Collapse
Affiliation(s)
- Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Qi Hao Looi
- My Cytohealth Sdn. Bhd., D353a, Menara Suezcap 1, KL Gateway, no. 2, Jalan Kerinchi, Gerbang Kerinchi Lestari, Kuala Lumpur 59200, Malaysia;
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Sau Har Lee
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| | - Pei Pei Chong
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
5
|
Potter ML, Hill WD, Isales CM, Hamrick MW, Fulzele S. MicroRNAs are critical regulators of senescence and aging in mesenchymal stem cells. Bone 2021; 142:115679. [PMID: 33022453 PMCID: PMC7901145 DOI: 10.1016/j.bone.2020.115679] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 01/10/2023]
Abstract
MicroRNAs (miRNAs) have recently come under scrutiny for their role in various age-related diseases. Similarly, cellular senescence has been linked to disease and aging. MicroRNAs and senescence likely play an intertwined role in driving these pathologic states. In this review, we present the connection between these two drivers of age-related disease concerning mesenchymal stem cells (MSCs). First, we summarize key miRNAs that are differentially expressed in MSCs and other musculoskeletal lineage cells during senescence and aging. Additionally, we also reviewed miRNAs that are regulated via traditional senescence-associated secretory phenotype (SASP) cytokines in MSC. Lastly, we summarize miRNAs that have been found to target components of the cell cycle arrest pathways inherently activated in senescence. This review attempts to highlight potential miRNA targets for regenerative medicine applications in age-related musculoskeletal disease.
Collapse
Affiliation(s)
- Matthew L Potter
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
6
|
Esmaeili A, Hosseini S, Baghaban Eslaminejad M. Engineered-extracellular vesicles as an optimistic tool for microRNA delivery for osteoarthritis treatment. Cell Mol Life Sci 2021; 78:79-91. [PMID: 32601714 PMCID: PMC11072722 DOI: 10.1007/s00018-020-03585-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/13/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Worldwide, osteoarthritis (OA) is one of the most common chronic diseases. In OA, profiling gene expression changes occur and cartilage tissue homeostasis is lost. Suggestions for OA treatment include regulation of gene expressions via the use of microRNAs (miRNAs). However, problems exist with the use of miRNAs, the most important of which is the delivery of sufficient amounts of effective miRNAs to save cartilage tissue. The engineering of extracellular vesicles (EVs) with the use of advanced techniques would be an efficient OA treatment. Therefore, we discuss the importance of miRNAs in terms of cartilage tissue regeneration and review recent advances in production of enriched EVs and miRNA delivery by EVs for future clinical applications.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
West-Livingston LN, Park J, Lee SJ, Atala A, Yoo JJ. The Role of the Microenvironment in Controlling the Fate of Bioprinted Stem Cells. Chem Rev 2020; 120:11056-11092. [PMID: 32558555 PMCID: PMC7676498 DOI: 10.1021/acs.chemrev.0c00126] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering and regenerative medicine has made numerous advances in recent years in the arena of fabricating multifunctional, three-dimensional (3D) tissue constructs. This can be attributed to novel approaches in the bioprinting of stem cells. There are expansive options in bioprinting technology that have become more refined and specialized over the years, and stem cells address many limitations in cell source, expansion, and development of bioengineered tissue constructs. While bioprinted stem cells present an opportunity to replicate physiological microenvironments with precision, the future of this practice relies heavily on the optimization of the cellular microenvironment. To fabricate tissue constructs that are useful in replicating physiological conditions in laboratory settings, or in preparation for transplantation to a living host, the microenvironment must mimic conditions that allow bioprinted stem cells to proliferate, differentiate, and migrate. The advances of bioprinting stem cells and directing cell fate have the potential to provide feasible and translatable approach to creating complex tissues and organs. This review will examine the methods through which bioprinted stem cells are differentiated into desired cell lineages through biochemical, biological, and biomechanical techniques.
Collapse
Affiliation(s)
- Lauren N. West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
8
|
Drug delivery in intervertebral disc degeneration and osteoarthritis: Selecting the optimal platform for the delivery of disease-modifying agents. J Control Release 2020; 328:985-999. [PMID: 32860929 DOI: 10.1016/j.jconrel.2020.08.041] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) and intervertebral disc degeneration (IVDD) as major cause of chronic low back pain represent the most common degenerative joint pathologies and are leading causes of pain and disability in adults. Articular cartilage (AC) and intervertebral discs are cartilaginous tissues with a similar biochemical composition and pathophysiological aspects of degeneration. Although treatments directed at reversing these conditions are yet to be developed, many promising disease-modifying drug candidates are currently under investigation. Given the localized nature of these chronic diseases, drug delivery systems have the potential to enhance therapeutic outcomes by providing controlled and targeted release of bioactives, minimizing the number of injections needed and increasing drug concentration in the affected areas. This review provides a comprehensive overview of the currently most promising disease-modifying drugs as well as potential drug delivery systems for OA and IVDD therapy.
Collapse
|
9
|
Wang R, Jiang W, Zhang L, Xie S, Zhang S, Yuan S, Jin Y, Zhou G. Intra-articular delivery of extracellular vesicles secreted by chondrogenic progenitor cells from MRL/MpJ superhealer mice enhances articular cartilage repair in a mouse injury model. Stem Cell Res Ther 2020; 11:93. [PMID: 32122385 PMCID: PMC7052980 DOI: 10.1186/s13287-020-01594-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/21/2019] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background Chondrogenic progenitor cells (CPCs) have high self-renewal capacity and chondrogenic potential. Intra-articular delivery of purified mesenchymal stem cells (MSCs) from MRL/MpJ “superhealer” mice increased bone volume during repair and prevents post-traumatic arthritis. Recently, although extracellular vesicles released from MSCs have been used widely for treating OA, the application of extracellular vesicles secreted by CPCs from MRL/MpJ mice in OA therapy has never been reported. In this study, we evaluated the effects of extracellular vesicles secreted by CPCs from control CBA (CBA-EVs) and MRL/MpJ mice (MRL-EVs) on proliferation and migration of murine chondrocytes. We also determined here if weekly intra-articular injections of CBA-EVs and MRL-EVs would repair and regenerate surgically induced model in mice. Methods CPC surface markers were detected by flow cytometry. CBA-EVs and MRL-EVs were isolated using an ultrafiltration method. Nanoparticle tracking analysis, transmission electron microscopy, and western blots were used to identify extracellular vesicles. CBA-EVs and MRL-EVs were injected intra-articularly in a mouse model of surgical destabilization of the medial meniscus (DMM)-induced OA, and histological and immunohistochemistry analyses were used to assess the efficacy of exosome injections. We used miRNA-seq analysis to analyze the expression profiles of exosomal miRNAs derived from CBA-EVs as well as MRL-EVs. Cell-counting and scratch assays were used to evaluate the effects of CBA-EVs and MRL-EVs on proliferation and migration of murine chondrocytes, respectively. Meanwhile, a specific RNA inhibitor assesses the roles of the candidate miRNAs in CPC-EV-induced regulation of function of chondrocytes. Results Both CBA-EVs and MRL-EVs stimulated chondrocyte proliferation and migration, but MRL-EVs exerted a stronger effect than CBA-EVs. The similar result was also observed in in vivo study, which indicated that injecting either CBA-EVs or MRL-EVs attenuated OA, but MRL-EVs showed a superior therapeutic effect in comparison with CBA-EVs. The results of bioinformatics analyses revealed that the differentially expressed exosomal miRNAs participated in multiple biological processes. We identified 80 significantly upregulated and 100 downregulated miRNAs. Moreover, we found that the top 20 differentially expressed exosomal miRNAs connected OA repair to processes such as AMPK signaling, regulation of autophagy, and insulin signaling. Notably, miRNA 221-3p were highly enriched in MRL-Exos and treatment with miR 221-3p inhibitor markedly decreased chondrocyte proliferation and migration induced by CBA-EVs or MRL-EVs in vitro. Conclusions This is the first study to demonstrate MRL-EVs had a greater therapeutic effect on the treatment of OA than CBA-EVs. This study will hopefully provide new insight into the pathogenesis, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, People's Republic of China.,Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory for Anti-ageing and Regenerative Medicine and Guangdong Key Laboratory for Genome Stability and Disease Prevention, Health Science Center, Shenzhen University , Shenzhen, 518060, People's Republic of China
| | - Wei Jiang
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory for Anti-ageing and Regenerative Medicine and Guangdong Key Laboratory for Genome Stability and Disease Prevention, Health Science Center, Shenzhen University , Shenzhen, 518060, People's Republic of China
| | - Lang Zhang
- Jiangxi Provincial Children's Hospital, Nanchang, 330006, People's Republic of China
| | - Saisai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Shuai Zhang
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory for Anti-ageing and Regenerative Medicine and Guangdong Key Laboratory for Genome Stability and Disease Prevention, Health Science Center, Shenzhen University , Shenzhen, 518060, People's Republic of China
| | - Shun Yuan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, People's Republic of China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory for Anti-ageing and Regenerative Medicine and Guangdong Key Laboratory for Genome Stability and Disease Prevention, Health Science Center, Shenzhen University , Shenzhen, 518060, People's Republic of China.
| |
Collapse
|
10
|
Abstract
The transcription factor Snai2, encoded by the SNAI2 gene, is an evolutionarily conserved C2H2 zinc finger protein that orchestrates biological processes critical to tissue development and tumorigenesis. Initially characterized as a prototypical epithelial-to-mesenchymal transition (EMT) transcription factor, Snai2 has been shown more recently to participate in a wider variety of biological processes, including tumor metastasis, stem and/or progenitor cell biology, cellular differentiation, vascular remodeling and DNA damage repair. The main role of Snai2 in controlling such processes involves facilitating the epigenetic regulation of transcriptional programs, and, as such, its dysregulation manifests in developmental defects, disruption of tissue homeostasis, and other disease conditions. Here, we discuss our current understanding of the molecular mechanisms regulating Snai2 expression, abundance and activity. In addition, we outline how these mechanisms contribute to disease phenotypes or how they may impact rational therapeutic targeting of Snai2 dysregulation in human disease.
Collapse
Affiliation(s)
- Wenhui Zhou
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kayla M Gross
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
11
|
Pasquini G, Kunej T. A Map of the microRNA Regulatory Networks Identified by Experimentally Validated microRNA-Target Interactions in Five Domestic Animals: Cattle, Pig, Sheep, Dog, and Chicken. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:448-456. [PMID: 31381467 DOI: 10.1089/omi.2019.0082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Domestic animals are members of the broader ecological context, in which humans are situated. Yet, genomics and systems science research have lagged behind and been relatively underappreciated in domestic animals compared to human genetics/genomics. Harnessing big data calls for omics data mapping studies in a broad range of mammals. To this end, microRNAs (miRNAs) regulate posttranscriptional expression of target genes, hence, governing different biological pathways and physiological processes. The knowledge of miRNA regulatory networks and maps is important for understanding regulation of gene expression and functions in both humans and domestic animals. However, complete miRNA regulatory networks have not yet been described in all species, particularly in domestic animals. We report here an original analysis so as to map the miRNA regulatory networks in domestic animals based on miRNA-target interactions (MTIs). Validated MTIs for five species; cattle, pig, sheep, dog, and chicken were extracted from the miRTarBase. miRNA regulomes were visualized using the Cytoscape software. The data in cattle, chicken, and pig were sufficient to visualize networks, identify central molecules, and subnetworks associated with the same phenotype; however, the MTI data in dog and sheep are still limited. We found several hub genes with large number of interactions, for example, 1 miRNA (bta-miR-17-5p) interacting with 27 genes and 7 miRNAs interacting with the same gene (tumor necrosis factor [TNF]) in cattle. In addition, two single-nucleotide polymorphisms were identified within the seed region of a previously demonstrated MTI, namely, between HMGB3 (high mobility group box 3) gene and bta-miR-17-5p. In summary, this miRNA regulome mapping study will enable and guide further studies of genome function in mammals with a view to applications in human as well as veterinary medicine. Furthermore, these miRNA regulomes can help to clarify fundamental pathways in cell biology and reveal molecular insights on phenotypic trait variability in common complex diseases and response phenotypes of drugs or other health interventions for precision medicine in the future.
Collapse
Affiliation(s)
- Giacomo Pasquini
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domzale, Slovenia
| | - Tanja Kunej
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domzale, Slovenia
| |
Collapse
|
12
|
Penolazzi L, Bonaccorsi G, Gafà R, Ravaioli N, Gabriele D, Bosi C, Lanza G, Greco P, Piva R. SLUG/HIF1-α/miR-221 regulatory circuit in endometrial cancer. Gene 2019; 711:143938. [PMID: 31220580 DOI: 10.1016/j.gene.2019.06.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE The pathogenesis of endometrial cancer (EC) involves many regulatory pathways including transcriptional regulatory networks supported by transcription factors and microRNAs only in part known. The aim of this retrospective study was to explore the possible correlation in the EC microenvironment between master regulators of complex phenomena such as steroid responsiveness through estrogen receptor alpha (ERα) and progesterone receptor (PR), epithelial-to-mesenchymal transition (supported by SLUG transcription factor), hypoxia (with hypoxia inducible factor-1 alpha, HIF-1α), and obesity that has been recognized as a EC risk factor. METHODS Formalin-Fixed Paraffin-Embedded (FFPE) blocks from University of Ferrara Pathology Archive were used and allocated into 2 groups according to their immunohistochemical positivity to ERα and PR, distinguishing the samples with a more benign prognosis (ERα+/PR+) from those with a poorer prognosis (ERα-/PR-). Immunohistochemistry for HIF1-α and SLUG was also performed. Body mass index (BMI) was registered at the time of diagnosis: patients with BMI ≥ 30 kg/m2 were defined obese (OB). Total RNA was isolated for miR-221 analysis. RESULTS We showed a comparable percentage of HIF1-α and SLUG positive samples in the ERα+/PR+ and ERα-/PR- groups. However, the obesity factor impacted more in the ERα+/PR+ group since the ratio between OB and non-obese (NOB) patients with high expression of HIF1-α and SLUG was higher in ERα+/PR+ than in the ERα-/PR- group. miR-221 levels were significantly higher in the OB than NOB patients, and, also in this case, obesity impacted more in the ERα+/PR+ group. CONCLUSIONS A molecular circuit of mutual regulation between ERα, PR, HIF1-α, SLUG and miR-221 is feasible in the EC and was firstly suggested by our research. In this interplay miR-221 seems to be in a nodal point of the regulatory system that is particularly strengthened by the metabolic changes in obesity.
Collapse
Affiliation(s)
- Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Gloria Bonaccorsi
- Section of Obstetrics and Gynecology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Roberta Gafà
- Section of Anatomic Pathology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Noemi Ravaioli
- Section of Obstetrics and Gynecology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Deborah Gabriele
- Section of Anatomic Pathology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Cristina Bosi
- Section of Anatomic Pathology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Giovanni Lanza
- Department of Medical Sciences, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Pantaleo Greco
- Section of Obstetrics and Gynecology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, S. Anna University Hospital, Via Aldo Moro, 8, 44124 Cona, Ferrara, Italy
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
13
|
Armiento AR, Alini M, Stoddart MJ. Articular fibrocartilage - Why does hyaline cartilage fail to repair? Adv Drug Deliv Rev 2019; 146:289-305. [PMID: 30605736 DOI: 10.1016/j.addr.2018.12.015] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/07/2018] [Accepted: 12/27/2018] [Indexed: 12/12/2022]
Abstract
Once damaged, articular cartilage has a limited potential to repair. Clinically, a repair tissue is formed, yet, it is often mechanically inferior fibrocartilage. The use of monolayer expanded versus naïve cells may explain one of the biggest discrepancies in mesenchymal stromal/stem cell (MSC) based cartilage regeneration. Namely, studies utilizing monolayer expanded MSCs, as indicated by numerous in vitro studies, report as a main limitation the induction of type X collagen and hypertrophy, a phenotype associated with endochondral bone formation. However, marrow stimulation and transfer studies report a mechanically inferior collagen I/II fibrocartilage as the main outcome. Therefore, this review will highlight the collagen species produced during the different therapeutic approaches. New developments in scaffold design and delivery of therapeutic molecules will be described. Potential future directions towards clinical translation will be discussed. New delivery mechanisms are being developed and they offer new hope in targeted therapeutic delivery.
Collapse
Affiliation(s)
| | - Mauro Alini
- AO Research Institute Davos, 7270 Davos Platz, Switzerland.
| | | |
Collapse
|
14
|
Oh SE, Park HJ, He L, Skibiel C, Junn E, Mouradian MM. The Parkinson's disease gene product DJ-1 modulates miR-221 to promote neuronal survival against oxidative stress. Redox Biol 2018; 19:62-73. [PMID: 30107296 PMCID: PMC6092527 DOI: 10.1016/j.redox.2018.07.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/28/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
DJ-1 is a highly conserved protein that protects neurons against oxidative stress and whose loss of function mutations are linked to recessively inherited Parkinson's disease (PD). While a number of signaling pathways have been shown to be regulated by DJ-1, its role in controlling cell survival through non-coding RNAs remains poorly understood. Here, using a microarray screen, we found that knocking down DJ-1 in human neuroblastoma cells results in down-regulation of microRNA-221 (miR-221). This is one of the most abundant miRNAs in the human brain and promotes neurite outgrowth and neuronal differentiation. Yet the molecular mechanism linking miR-221 to genetic forms of PD has not been studied. Consistent with the microarray data, miR-221 expression is also decreased in DJ-1-/- mouse brains. Re-introduction of wild-type DJ-1, but not its PD-linked pathogenic M26I mutant, restores miR-221 expression. Notably, over-expression of miR-221 is protective against 1-methyl-4-phenylpyridinium (MPP+)-induced cell death, while inhibition of endogenous miR-221 sensitizes cells to this toxin. Additionally, miR-221 down-regulates the expression of several pro-apoptotic proteins at basal conditions and prevents oxidative stress-induced up-regulation of bcl-2-like protein 11 (BIM). Accordingly, miR-221 protects differentiated DJ-1 knock-down ReNcell VM human dopaminergic neuronal cells from MPP+-induced neurite retraction and cell death. DJ-1 is a known activator of the mitogen-activated protein kinase (MAPK)/extracellular-regulated kinase (ERK) pathway and may modulate miR-221 levels in part through this pathway. We found that inhibiting ERK1/2 decreases miR-221 levels, whereas over-expressing ERK1 in DJ-1 knock-down cells increases miR-221 levels. These findings point to a new cytoprotective mechanism by which DJ-1 may increase miR-221 expression through the MAPK/ERK pathway, subsequently leading to repression of apoptotic molecules. The inability of a pathogenic DJ-1 mutant to modulate miR-221 further supports the relevance of this mechanism in neuronal health and its failure in DJ-1-linked PD.
Collapse
Affiliation(s)
- Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA
| | - Hye-Jin Park
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA
| | - Liqiang He
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA
| | - Catherine Skibiel
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA
| | - Eunsung Junn
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Room 180, Piscataway, NJ 08854, USA.
| |
Collapse
|
15
|
Extracellular vesicles: A new therapeutic strategy for joint conditions. Biochem Pharmacol 2018; 153:134-146. [PMID: 29427625 DOI: 10.1016/j.bcp.2018.02.004] [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: 11/27/2017] [Accepted: 02/05/2018] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are attracting increasing interest since they might represent a more convenient therapeutic tool with respect to their cells of origin. In the last years much time and effort have been expended to determine the biological properties of EVs from mesenchymal stem cells (MSCs) and other sources. The immunoregulatory, anti-inflammatory and regenerative properties of MSC EVs have been demonstrated in in vitro studies and animal models of rheumatoid arthritis or osteoarthritis. This cell-free approach has been proposed as a possible better alternative to MSC therapy in autoimmune conditions and tissue regeneration. In addition, EVs show great potential as biomarkers of disease or delivery systems for active molecules. The standardization of isolation and characterization methods is a key step for the development of EV research. A better understanding of EV mechanisms of action and efficacy is required to establish the potential therapeutic applications of this new approach in joint conditions.
Collapse
|
16
|
Bellavia D, Veronesi F, Carina V, Costa V, Raimondi L, De Luca A, Alessandro R, Fini M, Giavaresi G. Gene therapy for chondral and osteochondral regeneration: is the future now? Cell Mol Life Sci 2018; 75:649-667. [PMID: 28864934 PMCID: PMC11105387 DOI: 10.1007/s00018-017-2637-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022]
Abstract
Gene therapy might represent a promising strategy for chondral and osteochondral defects repair by balancing the management of temporary joint mechanical incompetence with altered metabolic and inflammatory homeostasis. This review analysed preclinical and clinical studies on gene therapy for the repair of articular cartilage defects performed over the last 10 years, focussing on expression vectors (non-viral and viral), type of genes delivered and gene therapy procedures (direct or indirect). Plasmids (non-viral expression vectors) and adenovirus (viral vectors) were the most employed vectors in preclinical studies. Genes delivered encoded mainly for growth factors, followed by transcription factors, anti-inflammatory cytokines and, less frequently, by cell signalling proteins, matrix proteins and receptors. Direct injection of the expression vector was used less than indirect injection of cells, with or without scaffolds, transduced with genes of interest and then implanted into the lesion site. Clinical trials (phases I, II or III) on safety, biological activity, efficacy, toxicity or bio-distribution employed adenovirus viral vectors to deliver growth factors or anti-inflammatory cytokines, for the treatment of osteoarthritis or degenerative arthritis, and tumour necrosis factor receptor or interferon for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Daniele Bellavia
- Rizzoli Orthopedic Institute, Bologna, Italy.
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy.
| | - F Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - V Carina
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - V Costa
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - L Raimondi
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - A De Luca
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - R Alessandro
- Biology and Genetics Unit, Department of Biopathology and Medical Biotechnology, University of Palermo, Palermo, Italy
| | - M Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - G Giavaresi
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
17
|
miR-27b promotes type II collagen expression by targetting peroxisome proliferator-activated receptor-γ2 during rat articular chondrocyte differentiation. Biosci Rep 2018; 38:BSR20171109. [PMID: 29187585 PMCID: PMC6435457 DOI: 10.1042/bsr20171109] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs (miRNAs) play an essential role in articular cartilage development and growth. However, the exact mechanisms involved in this process remain unknown. In the present study, we investigated the biological functions of miR-27b during hypertrophic differentiation of rat articular chondrocytes. Based on in situ hybridization and immunohistochemistry, we report that miR-27b expression is reduced in the hypertrophic zone of articular cartilage, but expression of peroxisome proliferator-activated receptor γ (Pparγ) is increased. Dual-luciferase reporter gene assay and Western blot analysis demonstrated that Pparγ2 is a target of miR-27b Overexpression of miR-27b inhibited expression of Pparγ2, as well as type X collagen (Col10a1) and matrix metalloproteinase 13 (Mmp13), while significantly promoting the expression of Sex-determining Region-box 9 (Sox9) and type II collagen (Col2a1) at both the mRNA and protein levels. Rosiglitazone, a Pparγ agonist, suppressed Col2a1 expression, while promoting expression of runt-related transcription factor 2 (Runx2) and Col10a1 in a concentration-dependent manner. siRNA-mediated knockdown of Pparγ2 caused an increase in protein levels of Col2a1. The present study demonstrates that miR-27b regulates chondrocyte hypertrophy in part by targetting Pparγ2, and that miR-27b may have important therapeutic implications in cartilage diseases.
Collapse
|
18
|
Budd E, Waddell S, de Andrés MC, Oreffo ROC. The Potential of microRNAs for Stem Cell-based Therapy for Degenerative Skeletal Diseases. ACTA ACUST UNITED AC 2017; 3:263-275. [PMID: 29214143 PMCID: PMC5700219 DOI: 10.1007/s40610-017-0076-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of Review Degenerative skeletal disorders including osteoarthritis (OA) and osteoporosis (OP) are the result of attenuation of tissue regeneration and lead to painful conditions with limited treatment options. Preventative measures to limit the onset of OA and OP remain a significant unmet clinical need. MicroRNAs (miRNAs) are known to be involved in the differentiation of stem cells, and in combination with stem cell therapy could induce skeletal regeneration and potentially prevent OA and OP onset. Recent Findings The combination of stem cells and miRNA has been successful at regenerating the bone and cartilage in vivo. MiRNAs, including miR-146b known to be involved in chondrogenic differentiation, could provide innovative targets for stem cell-based therapy, for the repair of articular cartilage defects forestalling the onset of OA or in the generation of a stem cell-based therapy for OP. Summary This review discusses the combination of skeletal stem cells (SSCs) and candidate miRNAs for application in a cell-based therapy approach for skeletal regenerative medicine.
Collapse
Affiliation(s)
- Emma Budd
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Shona Waddell
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - María C de Andrés
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Richard O C Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
19
|
Gu YL, Rong XX, Wen LT, Zhu GX, Qian MQ. miR-195 inhibits the proliferation and migration of chondrocytes by targeting GIT1. Mol Med Rep 2016; 15:194-200. [PMID: 27922692 DOI: 10.3892/mmr.2016.5982] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/13/2016] [Indexed: 11/05/2022] Open
Abstract
Previous studies have demonstrated that G-protein coupled receptor kinase interacting protein-1 (GIT1) and microRNAs (miRNAs) serve an important role in chondrocyte proliferation and migration. However, a limited number of studies conducted thus far have investigated the association between GIT1 and miRNAs. In the present study, putative miR‑195 binding sites in the GIT1 3'‑untranslated region were identified using common bioinformatic algorithms (miRanda, TargetScan, miRBase and miRWalk), and it was demonstrated that they may be involved in regulating GIT1 expression. Following transfection of miR‑195 mimics in chondrocytes, the expression of GIT1 was significantly reduced, whereas the expression was significantly increased following transfection with miR‑195 inhibitors. In addition, the results of the current study demonstrated that increased miR‑195 expression may downregulate chondrocyte proliferation and reduce cell migration. However, chondrocyte proliferation and migration was enhanced following suppression of miR‑195 expression. Furthermore, upon co‑transfection of miR‑195 and GIT1 expression vectors, the inhibitory effect of miR‑195 on chondrocyte proliferation and migration was attenuated. Therefore, miR‑195 may affect chondrocyte proliferation and migration via targeted regulation of GIT1 expression. The results of the current study provide novel evidence for the regulatory mechanisms of miRNAs in bone and cartilage tissues, which may facilitate further research and provide a greater understanding of different osteoarticular diseases.
Collapse
Affiliation(s)
- Yang-Lin Gu
- Department of Orthopedics, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Xiao-Xu Rong
- Department of Orthopedics, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Li-Ting Wen
- Department of Orthopedics, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Guo-Xing Zhu
- Department of Orthopedics, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Ming-Quan Qian
- Department of Orthopedics, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| |
Collapse
|
20
|
Toh WS, Lai RC, Hui JHP, Lim SK. MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment. Semin Cell Dev Biol 2016; 67:56-64. [PMID: 27871993 DOI: 10.1016/j.semcdb.2016.11.008] [Citation(s) in RCA: 356] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell (MSC) therapies have demonstrated efficacy in cartilage repair in animal and clinical studies. The efficacy of MSC-based therapies which was previously predicated on the chondrogenic potential of MSC is increasingly attributed to the paracrine secretion, particularly exosomes. Exosomes are thought to function primarily as intercellular communication vehicles to transfer bioactive lipids, nucleic acids (mRNAs and microRNAs) and proteins between cells to elicit biological responses in recipient cells. For MSC exosomes, many of these biological responses translated to a therapeutic outcome in injured or diseased cells. Here, we review the current understanding of MSC exosomes, discuss the possible mechanisms of action in cartilage repair within the context of the widely reported immunomodulatory and regenerative potency of MSC exosomes, and provide new perspectives for development of an off-the-shelf and cell-free MSC therapy for treatment of cartilage injuries and osteoarthritis.
Collapse
Affiliation(s)
- Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore; Tissue Engineering Program, Life Sciences Institute National University of Singapore, Singapore.
| | - Ruenn Chai Lai
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - James Hoi Po Hui
- Tissue Engineering Program, Life Sciences Institute National University of Singapore, Singapore; Cartilage Repair Program, Therapeutic Tissue Engineering Laboratory, Department of Orthopaedic Surgery, National University Health System, National University of Singapore, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
21
|
Seeliger C, Balmayor ER, van Griensven M. miRNAs Related to Skeletal Diseases. Stem Cells Dev 2016; 25:1261-81. [PMID: 27418331 DOI: 10.1089/scd.2016.0133] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
miRNAs as non-coding, short, double-stranded RNA segments are important for cellular biological functions, such as proliferation, differentiation, and apoptosis. miRNAs mainly contribute to the inhibition of important protein translations through their cleavage or direct repression of target messenger RNAs expressions. In the last decade, miRNAs got in the focus of interest with new publications on miRNAs in the context of different diseases. For many types of cancer or myocardial damage, typical signatures of local or systemically circulating miRNAs have already been described. However, little is known about miRNA expressions and their molecular effect in skeletal diseases. An overview of published studies reporting miRNAs detection linked with skeletal diseases was conducted. All regulated miRNAs were summarized and their molecular interactions were illustrated. This review summarizes the involvement and interaction of miRNAs in different skeletal diseases. Thereby, 59 miRNAs were described to be deregulated in tissue, cells, or in the circulation of osteoarthritis (OA), 23 miRNAs deregulated in osteoporosis, and 107 miRNAs deregulated in osteosarcoma (OS). The molecular influences of miRNAs regarding OA, osteoporosis, and OS were illustrated. Specific miRNA signatures for skeletal diseases are described in the literature. Some overlapped, but also unique ones for each disease exist. These miRNAs may present useful targets for the development of new therapeutic approaches and are candidates for diagnostic evaluations.
Collapse
Affiliation(s)
- Claudine Seeliger
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| | - Elizabeth R Balmayor
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| | - Martijn van Griensven
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| |
Collapse
|
22
|
Yeh CH, Jin L, Shen F, Balian G, Li X. miR-221 attenuates the osteogenic differentiation of human annulus fibrosus cells. Spine J 2016; 16:896-904. [PMID: 26997108 PMCID: PMC4970913 DOI: 10.1016/j.spinee.2016.03.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 01/30/2016] [Accepted: 03/11/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND In the moderate and end stages of intervertebral disc (IVD) degeneration, endochondral ossifications are found in the IVD. PURPOSE The aim of this study was to investigate whether endochondral ossification in the late stages of disc degeneration is due to the differentiation of resident progenitor cell in the annulus fibrosus (AF) and the potential signaling pathways in vitro. STUDY DESIGN This is an in vitro study of AF cell osteogenic differentiation and possible mechanisms METHODS Normal annulus fibrosus (NAF) and degenerated annulus fibrosus (DAF) cells were isolated from tissue removed surgically from juvenile patients with idiopathic scoliosis and adult patients with degenerative scoliosis. Osteogenic differentiation was investigated using quantitative reverse transcription polymerase chain reaction (RT-PCR) and histology. The effects of miR-221 on osteogenesis were measured by overexpression of miR-221 with lentivirus. BMP2 and phospho-Smad proteins were detected by Western blotting. RESULTS Both NAF and DAF cells underwent osteogenic differentiation, which was confirmed by detecting mineralization of the cell cultures and by an increase in the expression mRNAs for BMP2, runx2, alkaline phosphatase (ALP), and osteocalcin. DAF cells exhibited increased osteogenic differentiation potential over the NAF cells. By contrast to the elevated phospho-Smads, the basal level of miR-221 significantly decreased in DAF cells compared with that in NAF cells. Cultures of both cell types in osteogenic medium showed a decrease in miR-221 expression, and overexpression of miR-221 markedly decreased the level of BMP2, phospho-Smads, and the expression of osteogenic genes in DAF cells. The osteogenic potential of DAF cells diminished by the overexpression of miR-221. CONCLUSION Compared with NAF cells, AF cells from degenerated discs have a greater tendency for osteogenic differentiation, which involves the BMP-Smad pathways and can be regulated by miR-221. These observations may be developed into a therapeutic to prevent the endochondral ossification.
Collapse
Affiliation(s)
| | | | | | | | - Xudong Li
- Corresponding Author: Dr. Xudong Li, Mailing Address: Orthopaedic Surgery Laboratory, University of Virginia, Charlottesville, VA 22908, USA, , Tel: 434-982-4135, Fax: 434-922-1691
| |
Collapse
|
23
|
Li YP, Wei XC, Li PC, Chen CW, Wang XH, Jiao Q, Wang DM, Wei FY, Zhang JZ, Wei L. The Role of miRNAs in Cartilage Homeostasis. Curr Genomics 2016; 16:393-404. [PMID: 27019614 PMCID: PMC4765526 DOI: 10.2174/1389202916666150817203144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/18/2015] [Accepted: 06/26/2015] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is an age-related disease with poorly understood pathogenesis. Recent studies have demonstrated that miRNA might play a key role in OA initiation and development. We reviewed recent publications and elucidated the connection between miRNA and OA cartilage anabolic and catabolic signals, including four signaling pathways: TGF-β/Smads and BMPs signaling, associated with cartilage anabolism; and MAPK and NF-KB signaling, associated with cartilage catabolism. We also explored the relationships with MMP, ADAMTS and NOS (NitricOxide Synthases) families, as well as with the catabolic cytokines IL-1 and TNF-α. The potential role of miRNAs in biological processes such as cartilage degeneration, chondrocyte proliferation, and differentiation is discussed. Collective evidence indicates that miRNAs play a critical role in cartilage degeneration. These findings will aid in understanding the molecular network that governs articular cartilage homeostasis and in to elucidate the role of miRNA in the pathogenesis of OA.
Collapse
Affiliation(s)
- Yong Ping Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xiao Chun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Peng Cu Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Chun Wei Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xiao Hu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Qiang Jiao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Dong Ming Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Fang Yuan Wei
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian Zhong Zhang
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lei Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China;; Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China; ; Department of Orthopaedics, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin Street, Providence, RI 02903, USA
| |
Collapse
|
24
|
Lolli A, Narcisi R, Lambertini E, Penolazzi L, Angelozzi M, Kops N, Gasparini S, van Osch GJ, Piva R. Silencing of Antichondrogenic MicroRNA-221 in Human Mesenchymal Stem Cells Promotes Cartilage Repair In Vivo. Stem Cells 2016; 34:1801-11. [DOI: 10.1002/stem.2350] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Andrea Lolli
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Roberto Narcisi
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Marco Angelozzi
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| | - Nicole Kops
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Simona Gasparini
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
- Department of Otorhinolaryngology; Erasmus MC, University Medical Center; CN Rotterdam The Netherlands
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences; University of Ferrara; Ferrara Italy
| |
Collapse
|
25
|
Gabler J, Ruetze M, Kynast KL, Grossner T, Diederichs S, Richter W. Stage-Specific miRs in Chondrocyte Maturation: Differentiation-Dependent and Hypertrophy-Related miR Clusters and the miR-181 Family. Tissue Eng Part A 2015; 21:2840-51. [DOI: 10.1089/ten.tea.2015.0352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jessica Gabler
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Ruetze
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina L. Kynast
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Giovannini C, Minguzzi M, Baglioni M, Fornari F, Giannone F, Ravaioli M, Cescon M, Chieco P, Bolondi L, Gramantieri L. Suppression of p53 by Notch3 is mediated by Cyclin G1 and sustained by MDM2 and miR-221 axis in hepatocellular carcinoma. Oncotarget 2015; 5:10607-20. [PMID: 25431954 PMCID: PMC4279397 DOI: 10.18632/oncotarget.2523] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/24/2014] [Indexed: 02/07/2023] Open
Abstract
To successfully target Notch receptors as part of a multidrug anticancer strategy, it will be essential to fully characterize the factors that are modulated by Notch signaling. We recently reported that Notch3 silencing in HCC results in p53 up-regulation in vitro and, therefore, we focused on the mechanisms that associate Notch3 to p53 protein expression. We explored the regulation of p53 by Notch3 signalling in three HCC cell lines HepG2, SNU398 and Hep3B.We found that Notch3 regulates p53 at post-transcriptional level controlling both Cyclin G1 expression and the feed-forward circuit involving p53, miR-221 and MDM2. Moreover, our results were validated in human HCCs and in a rat model of HCC treated with Notch3 siRNAs. Our findings are becoming an exciting area for further in-depth research toward targeted inactivation of Notch3 receptor as a novel therapeutic approach for increasing the drug-sensitivity, and thereby improving the treatment outcome of patients affected by HCC. Indeed, we proved that Notch3 silencing strongly increases the effects of Nutilin-3.With regard to therapeutic implications, Notch3-specific drugs could represent a valuable strategy to limit Notch signaling in the context of hepatocellular carcinoma over-expressing this receptor.
Collapse
Affiliation(s)
- Catia Giovannini
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Manuela Minguzzi
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Michele Baglioni
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Francesca Fornari
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Ferdinando Giannone
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Matteo Ravaioli
- Department of Medical and Surgical Sciences, General and Transplant Surgery Unit, University of Bologna, Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, General and Transplant Surgery Unit, University of Bologna, Bologna, Italy
| | - Pasquale Chieco
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Luigi Bolondi
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy. Department of Medical and Surgical Sciences University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Center for Applied Biomedical Research (CRBA), S.Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
27
|
Abstract
Preclinical Research Bone is a rigid and dynamic organ that undergoes continuous turnover. Bone homeostasis is maintained by osteoclast-mediated bone resorption and osteoblast-mediated bone formation. The interruption of this balance can cause various diseases, including osteoporosis a public health issue due to the rate of hip fracture, the most serious outcome of osteoporosis. The bone loss in osteoporosis results from an increase in bone resorption versus bone formation. Thus, regulation of osteoblast and osteoclast activity is a main focus in the treatment of osteoporosis. MicroRNAs (miRNAs) are a class of single stranded noncoding RNAs consisting of 18-22 nucleotides that have an important role in cell differentiation, cell fate, apoptosis, and pathogenesis in various disease states. The potential therapeutic and biomarker function of miRNAs in treating bone disorders is receiving more attention. The current review summarizes the role of miRNAs in bone function at a cellular level in the context of their therapeutic potential.
Collapse
Affiliation(s)
- Junying Chen
- Department of Pathology, 324 Hospital of People's Liberation Army, Chongqing, China
| | - Min Qiu
- Department of Pathology, 324 Hospital of People's Liberation Army, Chongqing, China
| | - Ce Dou
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Zhen Cao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| |
Collapse
|
28
|
Gori M, Trombetta M, Santini D, Rainer A. Tissue engineering and microRNAs: future perspectives in regenerative medicine. Expert Opin Biol Ther 2015. [DOI: 10.1517/14712598.2015.1071349] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
29
|
Georgi N, Taipaleenmaki H, Raiss CC, Groen N, Portalska KJ, van Blitterswijk C, de Boer J, Post JN, van Wijnen AJ, Karperien M. MicroRNA Levels as Prognostic Markers for the Differentiation Potential of Human Mesenchymal Stromal Cell Donors. Stem Cells Dev 2015; 24:1946-55. [PMID: 25915705 DOI: 10.1089/scd.2014.0534] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The ability of human mesenchymal stromal/stem cells (hMSCs) to differentiate into various mesenchymal cell lineages makes them a promising cell source for the use in tissue repair strategies. Since the differentiation potential of hMSCs differs between donors, it is necessary to establish biomarkers for the identification of donors with high differentiation potential. In this study, we show that microRNA (miRNA) expression levels are effective for distinguishing donors with high differentiation potential from low differentiation potential. Twenty hMSC donors were initially tested for marker expression and differentiation potential. In particular, the chondrogenic differentiation potential was evaluated on the basis of histological matrix formation, mRNA expression levels of chondrogenic marker genes, and quantitative glycosaminoglycan deposition. Three donors out of twenty were identified as donors with high chondrogenic potential, whereas nine showed moderate and eight showed low chondrogenic potential. Expression profiles of miRNAs involved in chondrogenesis and cartilage homeostasis were used for the distinction between high-performance hMSCs and low-performance hMSCs. Global mRNA expression profiles of the donors before the onset of chondrogenic differentiation revealed minor differences in gene expression between low and high chondrogenic performers. However, analysis of miRNA expression during a 7-day differentiation period identified miR-210 and miR-630 as positive regulators of chondrogenesis. In contrast, miR-181 and miR-34a, which are negative regulators of chondrogenesis, were upregulated during differentiation in low-performing donors. In conclusion, profiling of hMSC donors for a specific panel of miRNAs may have a prognostic value for selecting donors with high differentiation potential to improve hMSC-based strategies for tissue regeneration.
Collapse
Affiliation(s)
- Nicole Georgi
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Hanna Taipaleenmaki
- 2 Heisenberg-Group for Molecular Skeletal Biology, Department of Trauma-, Hand- and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Christian C Raiss
- 3 Nanobiophysics Group, Faculty of Science and Technology, MESA+Institute for Nanotechnology, University of Twente , Enschede, the Netherlands
| | - Nathalie Groen
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Karolina Janaeczek Portalska
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Clemens van Blitterswijk
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Jan de Boer
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Janine N Post
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Andre J van Wijnen
- 5 Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| |
Collapse
|
30
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
31
|
Alečković M, Kang Y. Bone marrow stroma-derived miRNAs as regulators, biomarkers and therapeutic targets of bone metastasis. BONEKEY REPORTS 2015; 4:671. [PMID: 25908970 PMCID: PMC4398005 DOI: 10.1038/bonekey.2015.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are short, endogenous RNA molecules that have essential roles in regulating gene expression. They control numerous physiological and cellular processes, including normal bone organogenesis and homeostasis, by enhancing or inhibiting bone marrow cell growth, differentiation, functional activity and crosstalk of the multiple cell types within the bone. Hence, elucidating miRNA targets in bone marrow stromal cells has revealed novel regulations during bone development and maintenance. Moreover, recent studies have detailed the capacity for bone stromal miRNAs to influence bone metastasis from a number of primary carcinomas by interfering with bone homeostasis or by directly influencing metastatic tumor cells. Owing to the current lack of good diagnostic biomarkers of bone metastases, such changes in bone stromal miRNA expression in the presence of metastatic lesions may become useful biomarkers, and may even serve as therapeutic targets. In particular, cell-free and exosomal miRNAs shed from bone stromal cells into circulation may be developed into novel biomarkers that can be routinely measured in easily accessible samples. Taken together, these findings reveal the significant role of bone marrow stroma-derived miRNAs in the regulation of bone homeostasis and bone metastasis.
Collapse
Affiliation(s)
- Maša Alečković
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
32
|
Hoffman Y, Pilpel Y, Oren M. microRNAs and Alu elements in the p53-Mdm2-Mdm4 regulatory network. J Mol Cell Biol 2015; 6:192-7. [PMID: 24868102 DOI: 10.1093/jmcb/mju020] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
p53 is a transcription factor that governs numerous stress response pathways within the cell. Maintaining the right levels of p53 is crucial for cell survival and proper cellular homeostasis. The tight regulation of p53 involves many cellular components, most notably its major negative regulators Mdm2 and Mdm4, which maintain p53 protein amount and activity in tight check. microRNAs (miRNAs) are small non-coding RNAs that target specific mRNAs to translational arrest and degradation. miRNAs are also key components of the normal p53 pathway, joining forces with Mdm2 and Mdm4 to maintain proper p53 activity. Here we review the current knowledge of miRNAs targeting Mdm2 and Mdm4, and their importance in different tissues and in pathological states such as cancer. In addition, we address the role of Alu sequences-highly abundant retroelements spread throughout the human genome, and their impact on gene regulation via the miRNA machinery. Alus occupy a significant portion of genes' 3'UTR, and as such they have the potential to impact mRNA regulation. Since Alus are primate-specific, they introduce a new regulatory layer into primate genomes. Alus can influence and alter gene regulation, creating primate-specific cancer-preventive regulatory mechanisms to sustain the transition to longer life span in primates. We review the possible influence of Alu sequences on miRNA functionality in general and specifically within the p53 network.
Collapse
Affiliation(s)
- Yonit Hoffman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yitzhak Pilpel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
33
|
Umeda M, Terao F, Miyazaki K, Yoshizaki K, Takahashi I. MicroRNA-200a Regulates the Development of Mandibular Condylar Cartilage. J Dent Res 2015; 94:795-802. [PMID: 25784252 DOI: 10.1177/0022034515577411] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mandibular condylar cartilage (MCC) is classified as secondary cartilage, the histologic structure of which is unique from that of primary cartilage. MicroRNA (miRNA) is a small noncoding RNA that binds to the messenger RNA (mRNA) target to repress its translation and plays an important role in cell differentiation, proliferation, and death. Microarray analysis revealed that miR-200a was characteristically expressed during embryonic development. We hypothesized that miR-200a may be involved in regulating the formation of cartilage during MCC growth. We investigated the function of miR-200a by transfecting an inhibitor or mimic into MCC organ and cell cultures. A histologic examination revealed the localized inhibitory effects of the miR-200a mimic and widespread enhancing effects of the inhibitor on chondrocytic differentiation in the MCC organ culture system. An immunohistochemical examination and gene expression analysis demonstrated that the miR-200a inhibitor enhanced chondrogenesis, while the mimic had the opposite effect by enhancing cell proliferation. Quantitative reverse transcription polymerase chain reaction analysis revealed that miR-200a downregulated the gene expression of chondrocyte markers. Moreover, transfection of the miR-200a mimic into ATDC5 cells repressed the formation of the cartilaginous matrix. These results indicate that miR-200a contributed to chondrogenesis in developing MCC by controlling proliferation and differentiation in MCC cells.
Collapse
Affiliation(s)
- M Umeda
- Department of Orthodontics, Kyushu University Hospital, Fukuoka, Japan
| | - F Terao
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - K Miyazaki
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University Graduate School of Dentistry, Fukuoka, Japan
| | - K Yoshizaki
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - I Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| |
Collapse
|
34
|
Gurianova V, Stroy D, Kruzliak P, Kyrichenko V, Moibenko A, Dosenko V. Does proteasome regulate the level of microRNA-1 in cardiomyocytes? Application to anoxia-reoxygenation. Mol Cell Biochem 2015; 404:45-51. [PMID: 25724682 DOI: 10.1007/s11010-015-2365-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/21/2015] [Indexed: 11/30/2022]
Abstract
Proteasome and microRNAs play a critical role in almost all processes in a living organism, including pathology of the heart; however, their interaction is still in question. In the present study, we have found that proteasome inhibitor provoked increase of mature but not immature microRNA-1 in cultured cardiomyocytes, and tested the hypothesis that mature microRNA-1 can be a substrate for endonuclease activity of proteasome. In our in vitro experiments, we have found that proteasome fraction II is able to degrade both mature and primary but not precursor microRNA-1. However, this in vitro effect was not abolished by chemical inhibitor of proteolytic activities of proteasome. These data let us summarize that proteasome has the complex effect on the level of microRNA-1.
Collapse
Affiliation(s)
- Veronika Gurianova
- Bogomoletz Institute of Physiology, National Academy of Science, Kiev, Ukraine
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
microRNAs (miRNAs) regulate gene expression mainly at the posttranscriptional level. Many different miRNAs are expressed in chondrocytes, and each individual miRNA can regulate hundreds of target genes, creating a complex gene regulatory network. Experimental evidence suggests that miRNAs play significant roles in various aspects of cartilage development, homeostasis, and pathology. The possibility that miRNAs can be novel therapeutic targets for cartilage diseases led to vigorous investigations to understand the role of individual miRNAs in skeletal tissues. Here, we summarize our current understanding of miRNAs in chondrocytes and cartilage. In the first part, we discuss roles of miRNAs in growth plate development and chondrocyte differentiation. In the second part, we put a particular focus on articular cartilage and discuss the significance of variety of findings in the context of osteoarthritis, the most common degenerative joint disease.
Collapse
Affiliation(s)
- Fatemeh Mirzamohammadi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, MA 02114, USA
| | - Garyfallia Papaioannou
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, MA 02114, USA
| | - Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
36
|
MicroRNA processing machinery in the developing chick embryo. Gene Expr Patterns 2014; 16:114-21. [DOI: 10.1016/j.gep.2014.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/12/2014] [Accepted: 09/24/2014] [Indexed: 12/21/2022]
|
37
|
MicroRNAs involved in bone formation. Cell Mol Life Sci 2014; 71:4747-61. [PMID: 25108446 DOI: 10.1007/s00018-014-1700-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/31/2014] [Accepted: 08/04/2014] [Indexed: 12/21/2022]
Abstract
During skeletal development, mesenchymal progenitor cells undergo a multistage differentiation process in which they proliferate and become bone- and cartilage-forming cells. This process is tightly regulated by multiple levels of regulatory systems. The small non-coding RNAs, microRNAs (miRNAs), post-transcriptionally regulate gene expression. Recent studies have demonstrated that miRNAs play significant roles in all stages of bone formation, suggesting the possibility that miRNAs can be novel therapeutic targets for skeletal diseases. Here, we review the role and mechanism of action of miRNAs in bone formation. We discuss roles of specific miRNAs in major types of bone cells, osteoblasts, chondrocytes, osteoclasts, and their progenitors. Except a few, the current knowledge about miRNAs in bone formation has been obtained mainly by in vitro studies; further validation of these findings in vivo is awaited. We also discuss about several miRNAs of particular interest in the light of future therapies of bone diseases.
Collapse
|
38
|
Meza-Sosa KF, Pedraza-Alva G, Pérez-Martínez L. microRNAs: key triggers of neuronal cell fate. Front Cell Neurosci 2014; 8:175. [PMID: 25009466 PMCID: PMC4070303 DOI: 10.3389/fncel.2014.00175] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 06/06/2014] [Indexed: 01/31/2023] Open
Abstract
Development of the central nervous system (CNS) requires a precisely coordinated series of events. During embryonic development, different intra- and extracellular signals stimulate neural stem cells to become neural progenitors, which eventually irreversibly exit from the cell cycle to begin the first stage of neurogenesis. However, before this event occurs, the self-renewal and proliferative capacities of neural stem cells and neural progenitors must be tightly regulated. Accordingly, the participation of various evolutionary conserved microRNAs is key in distinct central nervous system (CNS) developmental processes of many organisms including human, mouse, chicken, frog, and zebrafish. microRNAs specifically recognize and regulate the expression of target mRNAs by sequence complementarity within the mRNAs 3′ untranslated region and importantly, a single microRNA can have several target mRNAs to regulate a process; likewise, a unique mRNA can be targeted by more than one microRNA. Thus, by regulating different target genes, microRNAs let-7, microRNA-124, and microRNA-9 have been shown to promote the differentiation of neural stem cells and neural progenitors into specific neural cell types while microRNA-134, microRNA-25 and microRNA-137 have been characterized as microRNAs that induce the proliferation of neural stem cells and neural progenitors. Here we review the mechanisms of action of these two sets of microRNAs and their functional implications during the transition from neural stem cells and neural progenitors to fully differentiated neurons. The genetic and epigenetic mechanisms that regulate the expression of these microRNAs as well as the role of the recently described natural RNA circles which act as natural microRNA sponges regulating post-transcriptional microRNA expression and function during the early stages of neurogenesis is also discussed.
Collapse
Affiliation(s)
- Karla F Meza-Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México Cuernavaca, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México Cuernavaca, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México Cuernavaca, México
| |
Collapse
|
39
|
Lolli A, Lambertini E, Penolazzi L, Angelozzi M, Morganti C, Franceschetti T, Pelucchi S, Gambari R, Piva R. Pro-Chondrogenic Effect of miR-221 and Slug Depletion in Human MSCs. Stem Cell Rev Rep 2014; 10:841-55. [DOI: 10.1007/s12015-014-9532-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
40
|
Wu C, Tian B, Qu X, Liu F, Tang T, Qin A, Zhu Z, Dai K. MicroRNAs play a role in chondrogenesis and osteoarthritis (review). Int J Mol Med 2014; 34:13-23. [PMID: 24736803 DOI: 10.3892/ijmm.2014.1743] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 04/08/2014] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is one of the most widespread degenerative joint diseases affecting the elderly. Research into the regulatory mechanisms underlying the pathogenesis of OA is therefore warranted, and over the past decade, there has been an increased focus on the functional role of microRNAs (miRNAs or miRs). In this systematic review, we aimed to review the evidence implicating miRNAs in the pathogenesis of chondrogenesis and OA. Systematic reviews of PubMed and Embase were performed to search for studies using strings of miRNAs, non-coding RNAs, cartilage, chondrocytes, chondrogenesis, chondrocytogenesis and OA. The identified studies were retrieved, and the references provided were searched. The selected studies were required to focus on the role of miRNAs in chondrogenesis and OA. The results of this review indicated that more than 25 miRNAs have been implicated in chondrogenesis and OA. In particular, chondrocytogenesis, chondrogenic differentiation, chondrocyte proliferation, chondrocyte hypertrophy, endochondral ossification, and proteolytic enzyme regulation are targeted or facilitated by more than 1 miRNA. To date, limited efforts have been performed to evaluate translational applications for this knowledge. Novel therapeutic strategies have been developed and are under investigation to selectively modulate miRNAs, which could potentially enable personalized OA therapy. miRNAs appear to be important modulators of chondrogenesis and OA. Their expression is frequently altered in OA, and many are functionally implicated in the pathogenesis of the disease. The translational roles and therapeutic potential of miRNAs remains to be evaluated.
Collapse
Affiliation(s)
- Chuanlong Wu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Bo Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Xinhua Qu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Fengxiang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - An Qin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Zhenan Zhu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
41
|
Karlsen TA, Jakobsen RB, Mikkelsen TS, Brinchmann JE. microRNA-140 targets RALA and regulates chondrogenic differentiation of human mesenchymal stem cells by translational enhancement of SOX9 and ACAN. Stem Cells Dev 2014; 23:290-304. [PMID: 24063364 DOI: 10.1089/scd.2013.0209] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lesions of articular cartilage do not heal spontaneously. One treatment strategy would be to make cartilage in the laboratory by directed chondrogenic differentiation of mesenchymal stem cells (MSCs). To promote our understanding of the molecular control of chondrogenesis, we have compared the changes in microRNAs (miRNAs) during in vitro chondrogenesis of MSCs with those observed in uncultured and dedifferentiated articular chondrocytes (ACs). Several miRNAs showed a reciprocal relationship during the differentiation of MSCs and dedifferentiation of ACs. miR-140-5p and miR-140-3p changed the most during in vitro chondrogenesis, they were the miRNAs most highly expressed in tissue-engineered chondrocytes, and they were also among the miRNAs most highly expressed in uncultured ACs. There was a 57% overlap for the 100 most highly expressed miRNAs in differentiated MSCs and uncultured ACs, but for other miRNAs, the expression pattern was quite different. We transiently and stably inhibited and overexpressed miR-140-5p and miR-140-3p in differentiating MSCs and dedifferentiating ACs, respectively, to describe global effects and identify and validate new targets. Surprisingly, SOX9 and aggrecan proteins were found to be downregulated in anti-miR-140 transduced differentiating MSCs despite unchanged mRNA levels. This suggests that miR-140 stimulates in vitro chondrogenesis by the upregulation of these molecules at the protein level. RALA, a small GTPase, was identified as a miR-140 target and knockdown experiments showed that RALA regulated SOX9 at the protein level. These observations shed new light on the effect of miR-140 for chondrogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Tommy A Karlsen
- 1 Norwegian Center for Stem Cell Research, Oslo University Hospital , Rikshospitalet, Oslo, Norway
| | | | | | | |
Collapse
|
42
|
Fornari F, Milazzo M, Galassi M, Callegari E, Veronese A, Miyaaki H, Sabbioni S, Mantovani V, Marasco E, Chieco P, Negrini M, Bolondi L, Gramantieri L. p53/mdm2 feedback loop sustains miR-221 expression and dictates the response to anticancer treatments in hepatocellular carcinoma. Mol Cancer Res 2013; 12:203-16. [PMID: 24324033 DOI: 10.1158/1541-7786.mcr-13-0312-t] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED The overexpression of microRNA-221 (miR-221) is reported in several human cancers including hepatocellular carcinoma, and its targeting by tailored treatments has been proposed. The evidence supporting the role of miR-221 in cancer is growing and has been mainly focused on the discovery of miR-221 targets as well as on its possible therapeutic exploitations. However, the mechanism sustaining miR-221 aberrant expression remains to be elucidated. In this study, MDM2 (E3 ubiquitin-protein ligase homolog), a known p53 (TP53) modulator, is identified as a direct target of miR-221, and a feed-forward loop is described that sustains miR-221 aberrant expression. Interestingly, miR-221 can activate the p53/mdm2 axis by inhibiting MDM2 and, in turn, p53 activation contributes to miR-221 enhanced expression. Moreover, by modulating the p53 axis, miR-221 impacts cell-cycle progression and apoptotic response to doxorubicin in hepatocellular carcinoma-derived cell lines. Finally, CpG island methylation status was assessed as a causative event associated with miR-221 upregulation in hepatocellular carcinoma cells and primary tumor specimens. In hepatocellular carcinoma-derived cell lines, pharmacologically induced DNA hypomethylation potentiated a significant increase in miR-221 expression. These data were confirmed in clinical specimens of hepatocellular carcinoma in which elevated miR-221 expression was associated with the simultaneous presence of wild-type p53 and DNA hypomethylation. IMPLICATIONS These findings reveal a novel miR-221-sustained regulatory loop that determines a p53-context-specific response to doxorubicin treatment in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Francesca Fornari
- St. Orsola-Malpighi University Hospital, via Albertoni, 15, 40138 Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shi K, Ma C. Reply to "On microRNA-214 suppressing osteogenic differentiation of C2C12 myoblast cells by targeting Osterix". Bone 2013; 57:328-34. [PMID: 23907033 DOI: 10.1016/j.bone.2013.07.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 07/09/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Kaikai Shi
- Department of Developmental Genetics, Nanjing Medical University, Nanjing 210029, China
| | | |
Collapse
|
44
|
Shang J, Liu H, Zhou Y. Roles of microRNAs in prenatal chondrogenesis, postnatal chondrogenesis and cartilage-related diseases. J Cell Mol Med 2013; 17:1515-24. [PMID: 24373548 PMCID: PMC3914653 DOI: 10.1111/jcmm.12161] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 09/09/2013] [Indexed: 12/15/2022] Open
Abstract
Cartilage has limited repair and regeneration capacity, thus damage of cartilage often results in its dysfunction and even chronic diseases like osteoarthritis (OA). Chondrogenesis induced by tissue-engineering methods is essential to treating cartilage-related diseases. MicroRNAs (miRNAs) are a class of small non-coding single-stranded RNAs which exert their biological effects by binding to the target messenger RNAs (mRNAs), resulting in decay or translation suppression of target mRNAs. There are emerging evidence indicating that miRNAs may play important roles in regulating both prenatal and postnatal chondrogenesis. During embryonic skeletal development, prenatal chondrogenesis is thought to be a precondition for formation of cartilage in developing limbs. Plenty of studies on different types of stem cells have undoubtedly proven their capacity of differentiating into chondrocytes. MiRNAs are found to comprehensively modulate these processes by establishing an interaction network with target genes, transcription factors and cytokines et al. In addition, translational application of miRNA technology has also been explored. In this review, we focus on the up-dated progress on regulatory mechanisms of miRNAs in prenatal and postnatal chondrogenesis. In addition, several miRNA target genes and roles of miRNAs in cartilage-related diseases are also discussed. This will contribute to studies of chondrogenesis mechanisms and development of new treating methods.
Collapse
Affiliation(s)
- Jin Shang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
45
|
Choi E, Choi E, Hwang KC. MicroRNAs as novel regulators of stem cell fate. World J Stem Cells 2013; 5:172-187. [PMID: 24179605 PMCID: PMC3812521 DOI: 10.4252/wjsc.v5.i4.172] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/13/2013] [Accepted: 08/17/2013] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence in stem cell biology has shown that microRNAs (miRNAs) play a crucial role in cell fate specification, including stem cell self-renewal, lineage-specific differentiation, and somatic cell reprogramming. These functions are tightly regulated by specific gene expression patterns that involve miRNAs and transcription factors. To maintain stem cell pluripotency, specific miRNAs suppress transcription factors that promote differentiation, whereas to initiate differentiation, lineage-specific miRNAs are upregulated via the inhibition of transcription factors that promote self-renewal. Small molecules can be used in a similar manner as natural miRNAs, and a number of natural and synthetic small molecules have been isolated and developed to regulate stem cell fate. Using miRNAs as novel regulators of stem cell fate will provide insight into stem cell biology and aid in understanding the molecular mechanisms and crosstalk between miRNAs and stem cells. Ultimately, advances in the regulation of stem cell fate will contribute to the development of effective medical therapies for tissue repair and regeneration. This review summarizes the current insights into stem cell fate determination by miRNAs with a focus on stem cell self-renewal, differentiation, and reprogramming. Small molecules that control stem cell fate are also highlighted.
Collapse
|
46
|
Le LTT, Swingler TE, Clark IM. Review: the role of microRNAs in osteoarthritis and chondrogenesis. ACTA ACUST UNITED AC 2013; 65:1963-74. [PMID: 23666813 DOI: 10.1002/art.37990] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 04/23/2013] [Indexed: 12/21/2022]
|
47
|
Mackenzie NCW, Staines KA, Zhu D, Genever P, Macrae VE. miRNA-221 and miRNA-222 synergistically function to promote vascular calcification. Cell Biochem Funct 2013; 32:209-16. [PMID: 24604335 PMCID: PMC4158883 DOI: 10.1002/cbf.3005] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/30/2013] [Accepted: 08/28/2013] [Indexed: 01/28/2023]
Abstract
Vascular calcification shares many similarities with skeletal mineralisation and involves the phenotypic trans-differentiation of vascular smooth muscle cells (VSMCs) to osteoblastic cells within a calcified environment. Various microRNAs (miRs) are known to regulate cell differentiation; however, their role in mediating VSMC calcification is not fully understood. miR-microarray analysis revealed the significant down-regulation of a range of miRs following nine days in culture, including miR-199b, miR-29a, miR-221, miR-222 and miR-31 (p < 0.05). Subsequent studies investigated the specific role of the miR-221/222 family in VSMC calcification. Real-time quantitative polymerase chain reaction data confirmed the down-regulation of miR-221 (32.4%; p < 0.01) and miR-222 (15.7%; p < 0.05). VSMCs were transfected with mimics of miR-221 and miR-222, individually and in combination. Increased calcium deposition was observed in the combined treatment (two-fold; p < 0.05) but not in individual treatments. Runx2 and Msx2 expression was increased during calcification, but no difference in expression was observed following transfection with miR mimics. Interestingly, miR-221 and miR-222 mimics induced significant changes in ectonucleotide phosphodiesterase 1 (Enpp1) and Pit-1 expression, suggesting that these miRs may modulate VSMC calcification through cellular inorganic phosphate and pyrophosphate levels. © 2013 The Authors. Cell Biochemistry and Function published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- N C W Mackenzie
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | | | | | | |
Collapse
|
48
|
Abstract
Extensive regeneration of the vertebrate body plan is found in salamander and fish species. In these organisms, regeneration takes place through reprogramming of differentiated cells, proliferation, and subsequent redifferentiation of adult tissues. Such plasticity is rarely found in adult mammalian tissues, and this has been proposed as the basis of their inability to regenerate complex structures. Despite their importance, the mechanisms underlying the regulation of the differentiated state during regeneration remain unclear. Here, we analyzed the role of the tumor-suppressor p53 during salamander limb regeneration. The activity of p53 initially decreases and then returns to baseline. Its down-regulation is required for formation of the blastema, and its up-regulation is necessary for the redifferentiation phase. Importantly, we show that a decrease in the level of p53 activity is critical for cell cycle reentry of postmitotic, differentiated cells, whereas an increase is required for muscle differentiation. In addition, we have uncovered a potential mechanism for the regulation of p53 during limb regeneration, based on its competitive inhibition by ΔNp73. Our results suggest that the regulation of p53 activity is a pivotal mechanism that controls the plasticity of the differentiated state during regeneration.
Collapse
|
49
|
Qin L, Chen Y, Liu X, Ye S, Yu K, Huang Z, Yu J, Zhou X, Chen H, Mo D. Integrative analysis of porcine microRNAome during skeletal muscle development. PLoS One 2013; 8:e72418. [PMID: 24039761 PMCID: PMC3770649 DOI: 10.1371/journal.pone.0072418] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/09/2013] [Indexed: 02/07/2023] Open
Abstract
Pig is an important agricultural animal for meat production and provides a valuable model for many human diseases. Functional studies have demonstrated that microRNAs (miRNAs) play critical roles in almost all aspects of skeletal muscle development and disease pathogenesis. To investigate the miRNAs involved in regulating different periods of skeletal muscle development, we herein performed a comprehensive research for porcine microRNAome (miRNAome) during 10 skeletal muscle developmental stages including 35, 49, 63, 77, 91 dpc (days post coitum) and 2, 28, 90, 120, 180 dpn (days postnatal) using Solexa sequencing technology. Our results extend the repertoire of pig miRNAome to 247 known miRNAs processed from 210 pre-miRNAs and 297 candidate novel miRNAs through comparison with known miRNAs in the miRBase. Expression analysis of the 15 most abundant miRNAs in every library indicated that functional miRNAome may be smaller and tend to be highly expressed. A series of muscle-related miRNAs summarized in our study present different patterns between myofibers formation phase and muscle maturation phase, providing valuable reference for investigation of functional miRNAs during skeletal muscle development. Analysis of temporal profiles of miRNA expression identifies 18 novel candidate myogenic miRNAs in pig, which might provide new insight into regulation mechanism mediated by miRNAs underlying muscle development.
Collapse
Affiliation(s)
- Lijun Qin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Sanxing Ye
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Kaifan Yu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Zheng Huang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jingwei Yu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Xingyu Zhou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Hu Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, P. R. China
| |
Collapse
|
50
|
Perez CJ, Rundhaug JE, Johnson DG, Oberyszyn TM, Tober KL, Kusewitt DF. Slug expression in mouse skin and skin tumors is not regulated by p53. J Invest Dermatol 2013; 134:566-568. [PMID: 24008423 PMCID: PMC3947144 DOI: 10.1038/jid.2013.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Carlos J Perez
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Joyce E Rundhaug
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - David G Johnson
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Tatiana M Oberyszyn
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Kathleen L Tober
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Donna F Kusewitt
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA.
| |
Collapse
|