1
|
Shrestha S, Tieu T, Wojnilowicz M, Voelcker NH, Forsythe JS, Frith JE. Delivery of miRNAs Using Porous Silicon Nanoparticles Incorporated into 3D Hydrogels Enhances MSC Osteogenesis by Modulation of Fatty Acid Signaling and Silicon Degradation. Adv Healthc Mater 2024; 13:e2400171. [PMID: 38657207 DOI: 10.1002/adhm.202400171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Strategies incorporating mesenchymal stromal cells (MSC), hydrogels and osteoinductive signals offer promise for bone repair. Osteoinductive signals such as growth factors face challenges in clinical translation due to their high cost, low stability and immunogenicity leading to interest in microRNAs as a simple, inexpensive and powerful alternative. The selection of appropriate miRNA candidates and their efficient delivery must be optimised to make this a reality. This study evaluated pro-osteogenic miRNAs and used porous silicon nanoparticles modified with polyamidoamine dendrimers (PAMAM-pSiNP) to deliver these to MSC encapsulated within gelatin-PEG hydrogels. miR-29b-3p, miR-101-3p and miR-125b-5p are strongly pro-osteogenic and are shown to target FASN and ELOVL4 in the fatty acid biosynthesis pathway to modulate MSC osteogenesis. Hydrogel delivery of miRNA:PAMAM-pSiNP complexes enhanced transfection compared to 2D. The osteogenic potential of hBMSC in hydrogels with miR125b:PAMAM-pSiNP complexes is evaluated. Importantly, a dual-effect on osteogenesis occurred, with miRNAs increasing expression of alkaline phosphatase (ALP) and Runt-related transcription factor 2 (RUNX2) whilst the pSiNPs enhanced mineralisation, likely via degradation into silicic acid. Overall, this work presents insights into the role of miRNAs and fatty acid signalling in osteogenesis, providing future targets to improve bone formation and a promising system to enhance bone tissue engineering.
Collapse
Affiliation(s)
- Surakshya Shrestha
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Terence Tieu
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Marcin Wojnilowicz
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
2
|
Obesity Affects the Proliferative Potential of Equine Endometrial Progenitor Cells and Modulates Their Molecular Phenotype Associated with Mitochondrial Metabolism. Cells 2022; 11:cells11091437. [PMID: 35563743 PMCID: PMC9100746 DOI: 10.3390/cells11091437] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/04/2022] [Accepted: 04/20/2022] [Indexed: 02/01/2023] Open
Abstract
The study aimed to investigate the influence of obesity on cellular features of equine endometrial progenitor cells (Eca EPCs), including viability, proliferation capacity, mitochondrial metabolism, and oxidative homeostasis. Eca EPCs derived from non-obese (non-OB) and obese (OB) mares were characterized by cellular phenotype and multipotency. Obesity-induced changes in the activity of Eca EPCs include the decline of their proliferative activity, clonogenic potential, mitochondrial metabolism, and enhanced oxidative stress. Eca EPCs isolated from obese mares were characterized by an increased occurrence of early apoptosis, loss of mitochondrial dynamics, and senescence-associated phenotype. Attenuated metabolism of Eca EPCs OB was related to increased expression of pro-apoptotic markers (CASP9, BAX, P53, P21), enhanced expression of OPN, PI3K, and AKT, simultaneously with decreased signaling stabilizing cellular homeostasis (including mitofusin, SIRT1, FOXP3). Obesity alters functional features and the self-renewal potential of endometrial progenitor cells. The impaired cytophysiology of progenitor cells from obese endometrium predicts lower regenerative capacity if used as autologous transplants.
Collapse
|
3
|
Modulation of miR-204 Expression during Chondrogenesis. Int J Mol Sci 2022; 23:ijms23042130. [PMID: 35216245 PMCID: PMC8874780 DOI: 10.3390/ijms23042130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/03/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
RUNX2 and SOX9 are two pivotal transcriptional regulators of chondrogenesis. It has been demonstrated that RUNX2 and SOX9 physically interact; RUNX2 transactivation may be inhibited by SOX9. In addition, RUNX2 exerts reciprocal inhibition on SOX9 transactivity. Epigenetic control of gene expression plays a major role in the alternative differentiation fates of stem cells; in particular, it has been reported that SOX9 can promote the expression of miRNA (miR)-204. Our aim was therefore to investigate the miR-204-5p role during chondrogenesis and to identify the relationship between this miR and the transcription factors plus downstream genes involved in chondrogenic commitment and differentiation. To evaluate the role of miR-204 in chondrogenesis, we performed in vitro transfection experiments by using Mesenchymal Stem Cells (MSCs). We also evaluated miR-204-5p expression in zebrafish models (adults and larvae). By silencing miR-204 during the early differentiation phase, we observed the upregulation of SOX9 and chondrogenic related genes compared to controls. In addition, we observed the upregulation of COL1A1 (a RUNX2 downstream gene), whereas RUNX2 expression of RUNX2 was slightly affected compared to controls. However, RUNX2 protein levels increased in miR-204-silenced cells. The positive effects of miR204 silencing on osteogenic differentiation were also observed in the intermediate phase of osteogenic differentiation. On the contrary, chondrocytes’ maturation was considerably affected by miR-204 downregulation. In conclusion, our results suggest that miR-204 negatively regulates the osteochondrogenic commitment of MSCs, while it positively regulates chondrocytes’ maturation.
Collapse
|
4
|
lncRNAs MALAT1 and LINC00657 upstream to miR-214-3p/BMP2 regulate osteogenic differentiation of human mesenchymal stem cells. Mol Biol Rep 2022; 49:6847-6857. [DOI: 10.1007/s11033-022-07136-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
|
5
|
Lu CH, Chen YA, Ke CC, Chiu SJ, Jeng FS, Chen CC, Hsieh YJ, Yang BH, Chang CW, Wang FS, Liu RS. Multiplexed Molecular Imaging Strategy Integrated with RNA Sequencing in the Assessment of the Therapeutic Effect of Wharton's Jelly Mesenchymal Stem Cell-Derived Extracellular Vesicles for Osteoporosis. Int J Nanomedicine 2021; 16:7813-7830. [PMID: 34880610 PMCID: PMC8646890 DOI: 10.2147/ijn.s335757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/05/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction Osteoporosis is a result of an imbalance in bone remodeling. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been considered as a potentially promising treatment for osteoporosis. However, the therapeutic effect, genetic alterations, and in vivo behavior of exogenous EVs for osteoporosis in mice models remain poorly understood. Methods A multiplexed molecular imaging strategy was constructed by micro-positron emission tomography (µPET)/computed tomography (CT), µCT, and optical imaging modality which reflected the osteoblastic activity, microstructure, and in vivo behavior of EVs, respectively. RNA sequencing was used to analyze the cargo of EVs, and the bone tissues of ovariectomized (OVX) mice post EV treatment. Results The result of [18F]NaF µPET showed an increase in osteoblastic activity in the distal femur of EV-treated mice, and the bone structural parameters derived from µCT were also improved. In terms of in vivo behavior of exogenous EVs, fluorescent dye-labeled EVs could target the distal femur of mice, whereas the uptakes of bone tissues were not significantly different between OVX mice and healthy mice. RNA sequencing demonstrated upregulation of ECM-related genes, which might associate with the PI3K/AKT signaling pathway, in line with the results of microRNA analysis showing that mir-21, mir-29, mir-221, and let-7a were enriched in Wharton’s jelly-MSC-EVs and correlated to the BMP and PI3K/AKT signaling pathways. Conclusion The therapeutic effect of exogenous WJ-MSC-EVs in the treatment of osteoporosis was successfully assessed by a multiplexed molecular imaging strategy. The RNA sequencing demonstrated the possible molecular targets in the regulation of bone remodeling. The results highlight the novelty of diagnostic and therapeutic strategies of EV-based treatment for osteoporosis.
Collapse
Affiliation(s)
- Cheng-Hsiu Lu
- Industrial Ph.D. Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Core Facility for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-An Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Chien-Chih Ke
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Sain-Jhih Chiu
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Fong-Shya Jeng
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Chao-Cheng Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Ju Hsieh
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Bang-Hung Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,PET Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Wei Chang
- PET Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Feng-Sheng Wang
- Core Facility for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ren-Shyan Liu
- Industrial Ph.D. Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,PET Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Nuclear Medicine, Cheng Hsin Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Lu CH, Chen YA, Ke CC, Liu RS. Mesenchymal Stem Cell-Derived Extracellular Vesicle: A Promising Alternative Therapy for Osteoporosis. Int J Mol Sci 2021; 22:12750. [PMID: 34884554 PMCID: PMC8657894 DOI: 10.3390/ijms222312750] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is the chronic metabolic bone disease caused by the disturbance of bone remodeling due to the imbalance of osteogenesis and osteoclastogenesis. A large population suffers from osteoporosis, and most of them are postmenopausal women or older people. To date, bisphosphonates are the main therapeutic agents in the treatment of osteoporosis. However, limited therapeutic effects with diverse side effects caused by bisphosphonates hindered the therapeutic applications and decreased the quality of life. Therefore, an alternative therapy for osteoporosis is still needed. Stem cells, especially mesenchymal stem cells, have been shown as a promising medication for numerous human diseases including many refractory diseases. Recently, researchers found that the extracellular vesicles derived from these stem cells possessed the similar therapeutic potential to that of parental cells. To date, a number of studies demonstrated the therapeutic applications of exogenous MSC-EVs for the treatment of osteoporosis. In this article, we reviewed the basic back ground of EVs, the cargo and therapeutic potential of MSC-EVs, and strategies of engineering of MSC-EVs for osteoporosis treatment.
Collapse
Affiliation(s)
- Cheng-Hsiu Lu
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Yi-An Chen
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan;
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chien-Chih Ke
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ren-Shyan Liu
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan;
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei 112, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- PET Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| |
Collapse
|
7
|
Nasirishargh A, Kumar P, Ramasubramanian L, Clark K, Hao D, Lazar SV, Wang A. Exosomal microRNAs from mesenchymal stem/stromal cells: Biology and applications in neuroprotection. World J Stem Cells 2021; 13:776-794. [PMID: 34367477 PMCID: PMC8316862 DOI: 10.4252/wjsc.v13.i7.776] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/29/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are extensively studied as cell-therapy agents for neurological diseases. Recent studies consider exosomes secreted by MSCs as important mediators for MSCs' neuroprotective functions. Exosomes transfer functional molecules including proteins, lipids, metabolites, DNAs, and coding and non-coding RNAs from MSCs to their target cells. Emerging evidence shows that exosomal microRNAs (miRNAs) play a key role in the neuroprotective properties of these exosomes by targeting several genes and regulating various biological processes. Multiple exosomal miRNAs have been identified to have neuroprotective effects by promoting neurogenesis, neurite remodeling and survival, and neuroplasticity. Thus, exosomal miRNAs have significant therapeutic potential for neurological disorders such as stroke, traumatic brain injury, and neuroinflammatory or neurodegenerative diseases and disorders. This review discusses the neuroprotective effects of selected miRNAs (miR-21, miR-17-92, miR-133, miR-138, miR-124, miR-30, miR146a, and miR-29b) and explores their mechanisms of action and applications for the treatment of various neurological disease and disorders. It also provides an overview of state-of-the-art bioengineering approaches for isolating exosomes, optimizing their yield and manipulating the miRNA content of their cargo to improve their therapeutic potential.
Collapse
Affiliation(s)
- Aida Nasirishargh
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Lalithasri Ramasubramanian
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States
| | - Kaitlin Clark
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Dake Hao
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Sabrina V Lazar
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States.
| |
Collapse
|
8
|
Zhao YJ, Gao ZC, He XJ, Li J. The let-7f-5p-Nme4 pathway mediates tumor necrosis factor α-induced impairment in osteogenesis of bone marrow-derived mesenchymal stem cells. Biochem Cell Biol 2021; 99:488-498. [PMID: 34297624 DOI: 10.1139/bcb-2020-0281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although tumor necrosis factor α (TNF-α)-mediated inflammation significantly impacts osteoporosis, the mechanisms underlying the osteogenic differentiation defects of bone marrow-derived mesenchymal stem cells (BM-MSCs) caused by TNF-α remain poorly understood. We found that TNF-α stimulation of murine BM-MSCs significantly upregulated the expression levels of several microRNAs (miRNAs), including let-7f-5p, but this increase was significantly reversed by treatment with the kinase inhibitor BAY 11-7082. To study gain- or loss of function, we transfected cells with an miRNA inhibitor or miRNA mimic. We then demonstrated that let-7f-5p impaired osteogenic differentiation of BM-MSCs in the absence and presence of TNF-α, as evidenced by alkaline phosphatase and alizarin red staining as well as quantitative assays of the mRNA levels of bone formation marker genes in differentiated BM-MSCs. Moreover, let-7f-5p targets the 3' untranslated region of Nucleoside diphosphate kinase 4 (Nme4) mRNA and negatively regulates Nme4 expression in mouse BM-MSCs. Ectopic expression of Nme4 completely reversed the inhibitory effects of the let-7f-5p mimic on osteogenic differentiation of mouse BM-MSCs. Furthermore, inhibition of let-7f-5p or overexpression of Nme4 in BM-MSCs restored in-vivo bone formation in an ovariectomized animal model. Collectively, our work indicates that let-7f-5p is involved in TNF-α-mediated reduction of BM-MSC osteogenesis via targeting Nme4.
Collapse
Affiliation(s)
- Ying-Jie Zhao
- Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng-Chao Gao
- Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xi-Jing He
- Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jing Li
- Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
9
|
Phan TH, Divakarla SK, Yeo JH, Lei Q, Tharkar P, Pansani TN, Leslie KG, Tong M, Coleman VA, Jämting Å, Du Plessis MD, New EJ, Kalionis B, Demokritou P, Woo HK, Cho YK, Chrzanowski W. New Multiscale Characterization Methodology for Effective Determination of Isolation-Structure-Function Relationship of Extracellular Vesicles. Front Bioeng Biotechnol 2021; 9:669537. [PMID: 34164385 PMCID: PMC8215393 DOI: 10.3389/fbioe.2021.669537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have been lauded as next-generation medicines, but very few EV-based therapeutics have progressed to clinical use. Limited clinical translation is largely due to technical barriers that hamper our ability to mass produce EVs, i.e., to isolate, purify, and characterize them effectively. Technical limitations in comprehensive characterization of EVs lead to unpredicted biological effects of EVs. Here, using a range of optical and non-optical techniques, we showed that the differences in molecular composition of EVs isolated using two isolation methods correlated with the differences in their biological function. Our results demonstrated that the isolation method determines the composition of isolated EVs at single and sub-population levels. Besides the composition, we measured for the first time the dry mass and predicted sedimentation of EVs. These parameters were likely to contribute to the biological and functional effects of EVs on single cell and cell cultures. We anticipate that our new multiscale characterization approach, which goes beyond traditional experimental methodology, will support fundamental understanding of EVs as well as elucidate the functional effects of EVs in in vitro and in vivo studies. Our findings and methodology will be pivotal for developing optimal isolation methods and establishing EVs as mainstream therapeutics and diagnostics. This innovative approach is applicable to a wide range of sectors including biopharma and biotechnology as well as to regulatory agencies.
Collapse
Affiliation(s)
- Thanh Huyen Phan
- Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Shiva Kamini Divakarla
- Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Jia Hao Yeo
- School of Chemistry, The University of Sydney, Camperdown, NSW, Australia
| | - Qingyu Lei
- Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Priyanka Tharkar
- Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Taisa Nogueira Pansani
- Department of Dental Materials and Prosthodontics, Araraquara School of Dentistry, UNESP-Universidade Estadual Paulista, Araraquara, Brazil
| | - Kathryn G Leslie
- School of Chemistry, The University of Sydney, Camperdown, NSW, Australia
| | - Maggie Tong
- School of Chemistry, The University of Sydney, Camperdown, NSW, Australia
| | - Victoria A Coleman
- Nanometrology Section, National Measurement Institute Australia, Lindfield, NSW, Australia
| | - Åsa Jämting
- Nanometrology Section, National Measurement Institute Australia, Lindfield, NSW, Australia
| | - Mar-Dean Du Plessis
- Nanometrology Section, National Measurement Institute Australia, Lindfield, NSW, Australia
| | - Elizabeth J New
- School of Chemistry, The University of Sydney, Camperdown, NSW, Australia.,School of Chemistry, Faculty of Science, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Bill Kalionis
- Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's Hospital, and Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
| | - Philip Demokritou
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Hyun-Kyung Woo
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, South Korea.,Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, South Korea.,Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Wojciech Chrzanowski
- Sydney School of Pharmacy, Faculty of Medicine and Health, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
10
|
Xiang J, Bian Y. PWAR6 interacts with miR‑106a‑5p to regulate the osteogenic differentiation of human periodontal ligament stem cells. Mol Med Rep 2021; 23:268. [PMID: 33576453 PMCID: PMC7893692 DOI: 10.3892/mmr.2021.11907] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) associated with bone regeneration serve an important role in the treatment of periodontal disease. Long non-coding RNAs are involved in the osteogenesis of multiple stem cells and can act as a sponge of microRNAs (miRs). The present study aimed to investigate the interaction between Prader Willi/Angelman region RNA 6 (PWAR6) and miR-106a-5p, as well as their influences on the osteogenic differentiation of hPDLSCs. hPDLSCs were isolated and cultured in osteogenic medium (OM) or growth medium (GM) for 7 days prior to transfection with PWAR6 overexpression vector, short hairpin RNA PWAR6 or miR-106a-5p mimic. The expression levels of runt-related transcription factor 2, osteocalcin and bone morphogenetic protein 2 (BMP2) were detected by western blotting and reverse transcription-quantitative PCR (RT-qPCR), and the expression levels of PWAR6, miR-106a-5p and alkaline phosphatase (ALP) were determined by RT-qPCR. ALP activity assays and Alizarin red staining were performed to detect osteogenesis and mineralization, respectively. Luciferase activities of wild-type and mutant PWAR6 and BMP2 were assessed by conducting a dual-luciferase reporter assay. The results indicated that PWAR6 expression was upregulated in OM-incubated hPDLSCs compared with GM-incubated hPDLSCs, and PWAR6 overexpression increased the osteogenic differentiation and mineralization of hPDLSCs compared with the corresponding control group. By contrast, miR-106a-5p expression was downregulated in OM-incubated hPDLSCs compared with GM-incubated hPDLSCs. PWAR6 acted as a sponge of miR-106a-5p and PWAR6 overexpression promoted the osteogenesis of miR-106a-5p mimic-transfected hPDLSCs. BMP2 was predicted as a target gene of miR-106a-5p. Collectively, the results indicated that PWAR6 displayed a positive influence on the osteogenic differentiation of hPDLSCs. The results of the present study demonstrated that the PWAR6/miR-106a-5p interaction network may serve as a potential regulatory mechanism underlying hPDLSCs osteogenesis.
Collapse
Affiliation(s)
- Juan Xiang
- Department of Oral and Maxillofacial Surgery, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Ying Bian
- Department of Oral and Maxillofacial Surgery, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
11
|
Hu C, Zhao L, Li L. Genetic modification by overexpression of target gene in mesenchymal stromal cell for treating liver diseases. J Mol Med (Berl) 2021; 99:179-192. [PMID: 33388882 DOI: 10.1007/s00109-020-02031-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/02/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022]
Abstract
Different hepatoxic factors cause irreversible liver injury, leading to liver failure, cirrhosis, and cancer in mammals. Liver transplantation is the only effective strategy, which can improve the prognosis of patients with end-stage liver diseases, but it is limited by liver donor shortage, expensive costs, liver graft rejection and dysfunction, and recurring liver failure. Recently, mesenchymal stromal cells (MSCs) isolated from various tissues are regarded as the main stem cell type with therapeutic effects in liver diseases because of their hepatogenic differentiation, anti-inflammatory, immuoregulatory, anti-apoptotic, antifibrotic, and antitumor capacities. To further improve the therapeutic effects of MSCs, multiple studies showed that genetically engineered MSCs have increased regenerative capacities and are able to more effectively inhibit cell death. Moreover, they are able to secrete therapeutic proteins for attenuating liver injury in liver diseases. In this review, we mainly focus on gene overexpression for reprogramming MSCs to increase their therapeutic effects in treating various liver diseases. We described the potential mechanisms of MSCs with gene overexpression in attenuating liver injury, and we recommend further expansion of experiments to discover more gene targets and optimized gene delivery methods for MSC-based regenerative medicine. We also discussed the potential hurdles in genetic engineering MSCs. In conclusion, we highlight that we need to overcome all scientific hurdles before genetically modified MSC therapy can be translated into clinical practices for patients with liver diseases.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
12
|
Ataei A, Poorebrahim M, Rajabpour A, Rizvanov A, Shahriar Arab S. Topological Analysis of Regulatory Networks Reveals Functionally Key Genes and miRNAs Involved in the Differentiation of Mesenchymal Stem Cells. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2565. [PMID: 34179189 PMCID: PMC8217530 DOI: 10.30498/ijb.2021.2565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background The details of molecular mechanisms underlying the differentiation of Mesenchymal Stem Cells (MSCs) into specific lineages are not well understood. Objectives We aimed to construct the interactome network and topology analysis of bone marrow mesenchymal stem cell of CAGE data. Applying the enrichment results, we wanted to introduce the common genes and hub-microRNA and hub-genes of these giant network. Materials and Methods In this study, we constructed gene regulatory networks for each non-mesenchymal cell lineage according to their gene expression profiles obtained from FANTOM5 database. The putative interactions of TF-gene and protein-protein were determined using TRED, STRING, HPRD and GeneMANIA servers. In parallel, a regulatory network including corresponding miRNAs and total differentially expressed genes (DEGs) was constructed for each cell lineage. Results The results indicated that analysis of networks' topology can significantly distinguish the hub regulatory genes and miRNAs involved in the differentiation of MSCs. The functional annotation of identified hub genes and miRNAs revealed that several signal transduction pathways i.e. AKT, WNT and TGFβ and cell proliferation related pathways play a pivotal role in the regulation of MSCs differentiation. We also classified cell lineages into two groups based on their predicted miRNA profiles. Conclusions In conclusion, we found a number of hub genes and miRNAs which seem to have key regulatory functions during differentiation of MSCs. Our results also introduce a number of new regulatory genes and miRNAs which can be considered as the new candidates for genetic manipulation of MSCs in vitro.
Collapse
Affiliation(s)
- Atousa Ataei
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Equal contribution
| | - Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran.,Equal contribution
| | - Azam Rajabpour
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
13
|
Hu HF, Xu WW, Zhang WX, Yan X, Li YJ, Li B, He QY. Identification of miR-515-3p and its targets, vimentin and MMP3, as a key regulatory mechanism in esophageal cancer metastasis: functional and clinical significance. Signal Transduct Target Ther 2020; 5:271. [PMID: 33243974 PMCID: PMC7693265 DOI: 10.1038/s41392-020-00275-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 02/08/2023] Open
Abstract
Metastasis is the main factor of treatment failure in cancer patients, but the underlying mechanism remains to be elucidated and effective new treatment strategies are urgently needed. This study aims to explore novel key metastasis-related microRNAs (miRNAs) in esophageal squamous cell carcinoma (ESCC). By comparing miRNA profiles of the highly metastatic ESCC cell sublines, we established through serial in vivo selection with the parental cells, we found that the expression level of miR-515-3p was lower in ESCC tumor tissues than adjacent normal tissues, further decreased in metastatic tumors, and moreover, markedly associated with advanced stage, metastasis and patient survival. The in vitro and in vivo assays suggested that miR-515-3p could increase the expression of the epithelial markers as well as decrease the expression of the mesenchymal markers, and more importantly, suppress invasion and metastasis of ESCC cells. Mechanistically, we revealed that miR-515-3p directly regulated vimentin and matrix metalloproteinase-3 (MMP3) expression by binding to the coding sequence and 3'untranslated region, respectively. In addition, the data from whole-genome methylation sequencing and methylation-specific PCR indicated that the CpG island within miR-515-3p promoter was markedly hypermethylated in ESCC cell lines and ESCC tumor tissues, which may lead to deregulation of miR-515-3p expression in ESCC. Furthermore, our preclinical experiment provides solid evidence that systemic delivery of miR-515-3p oligonucleotide obviously suppressed the metastasis of ESCC cells in nude mice. Taken together, this study demonstrates that miR-515-3p suppresses tumor metastasis and thus represents a promising prognostic biomarker and therapeutic strategy in ESCC.
Collapse
Affiliation(s)
- Hui-Fang Hu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Wen Wen Xu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine and MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wei-Xia Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Xin Yan
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Yang-Jia Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Bin Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China.
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou, China.
| |
Collapse
|
14
|
MicroRNAomic Transcriptomic Analysis Reveal Deregulation of Clustered Cellular Functions in Human Mesenchymal Stem Cells During in Vitro Passaging. Stem Cell Rev Rep 2020; 16:222-238. [PMID: 31848878 DOI: 10.1007/s12015-019-09924-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Clinical trials using human mesenchymal stem/stromal cells (hMSCs) for cell replacement therapy showed varied outcomes, where cells' efficacy has been perceived as the limiting factor. In particular, the quality and number of the expanded cells in vitro. In this study, we aimed to determine molecular signatures of hMSCs derived from the pulp of extracted deciduous teeth (SHED) and Wharton's jelly (WJSCs) that associated with cellular ageing during in vitro passaging. We observed distinct phenotypic changes resembling proliferation reduction, cell enlargement, an increase cell population in G2/M phase, and differentially expressed of tumor suppressor p53 in passage (P) 6 as compared to P3, which indicating in vitro cell senescence. The subsequent molecular analysis showed a set of diverse differentially expressed miRNAs and mRNAs involved in maintaining cell proliferation and stemness properties. Considering the signaling pathway related to G2/M DNA damage regulation is widely recognized as part of anti-proliferation mechanism controlled by p53, we explored possible miRNA-mRNA interaction in this regulatory pathway based on genomic coordinates retrieved from miRanda. Our work reveals the potential reason for SHED underwent proliferation arrest due to the direct impinge on the expression of CKS1 by miRNAs specifically miR-22 and miR-485-5p which lead to down regulation of CDK1 and Cyclin B. It is intended that our study will contribute to the understanding of these miRNA/mRNA driving the biological process and regulating different stages of cell cycle is beneficial in developing effective rejuvenation strategies in order to obtain quality stem cells for transplantation.
Collapse
|
15
|
Asgarpour K, Shojaei Z, Amiri F, Ai J, Mahjoubin-Tehran M, Ghasemi F, ArefNezhad R, Hamblin MR, Mirzaei H. Exosomal microRNAs derived from mesenchymal stem cells: cell-to-cell messages. Cell Commun Signal 2020; 18:149. [PMID: 32917227 PMCID: PMC7488404 DOI: 10.1186/s12964-020-00650-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
Exosomes are extracellular vesicles characterized by their size, source, release mechanism and contents. MicroRNAs (miRNAs) are single stranded non-coding RNAs transcribed from DNA. Exosomes and miRNAs are widespread in eukaryotic cells, especially in mesenchymal stem cells (MSCs). MSCs are used for tissue regeneration, and also exert paracrine, anti-inflammatory and immunomodulatory effects. However, the use of MSCs is controversial, especially in the presence or after the remission of a tumor, due to their secretion of growth factors and their migration ability. Instead of intact MSCs, MSC-derived compartments or substances could be used as practical tools for diagnosis, follow up, management and monitoring of diseases. Herein, we discuss some aspects of exosomal miRNAs derived from MSCs in the progression, diagnosis and treatment of various diseases. Video Abstract.
Collapse
Affiliation(s)
- Kasra Asgarpour
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Zahra Shojaei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amiri
- School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Reza ArefNezhad
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran.
| |
Collapse
|
16
|
Abstract
Mesenchymal stem cells (MSCs) represent a promising source of cell-based therapies for treatment of a wide variety of injuries and diseases. Their tropism and migration to the damaged sites, which are elicited by cytokines secreted from tissues around pathology, are the prerequisite for tissue repair and regeneration. Better understanding of the elicited-migration of MSCs and discovering conditions that elevate their migration ability, will help to increase their homing to pathologies and improve therapeutic efficacy. It is increasingly recognized that microRNAs are important regulators of cell migration. Here we summarize current understanding of the microRNA-regulated migration of MSCs.
Collapse
|
17
|
Wijaya JC, Khanabdali R, Georgiou HM, Kalionis B. Ageing in human parturition: impetus of the gestation clock in the decidua†. Biol Reprod 2020; 103:695-710. [PMID: 32591788 DOI: 10.1093/biolre/ioaa113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Despite sharing many common features, the relationship between ageing and parturition remains poorly understood. The decidua is a specialized lining of endometrial tissue, which develops in preparation for pregnancy. The structure and location of the decidua support its role as the physical scaffold for the growing embryo and placenta, and thus, it is vital to sustain pregnancy. Approaching term, the physical support properties of the decidua are naturally weakened to permit parturition. In this review, we hypothesize that the natural weakening of decidual tissue at parturition is promoted by the ageing process. Studies of the ageing-related functional and molecular changes in the decidua at parturition are reviewed and classified using hallmarks of ageing as the framework. The potential roles of decidual mesenchymal stem/stromal cell (DMSC) ageing in labor are also discussed because, although stem cell exhaustion is also a hallmark of ageing, its role in labor is not completely understood. In addition, the potential roles of extracellular vesicles secreted by DMSCs in labor, and their parturition-related miRNAs, are reviewed to gain further insight into this research area. In summary, the literature supports the notion that the decidua ages as the pregnancy progresses, and this may facilitate parturition, suggesting that ageing is the probable impetus of the gestational clocks in the decidua. This conceptual framework was developed to provide a better understanding of the natural ageing process of the decidua during parturition as well as to encourage future studies of the importance of healthy ageing for optimal pregnancy outcomes.
Collapse
Affiliation(s)
- Joan C Wijaya
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Ramin Khanabdali
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Process Development, Exopharm Limited, Melbourne, Victoria, Australia
| | - Harry M Georgiou
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Kim SY, Joglekar MV, Hardikar AA, Phan TH, Khanal D, Tharkar P, Limantoro C, Johnson J, Kalionis B, Chrzanowski W. Placenta Stem/Stromal Cell-Derived Extracellular Vesicles for Potential Use in Lung Repair. Proteomics 2020; 19:e1800166. [PMID: 31318160 DOI: 10.1002/pmic.201800166] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 06/26/2019] [Indexed: 12/28/2022]
Abstract
Many acute and chronic lung injuries are incurable and rank as the fourth leading cause of death globally. While stem cell treatment for lung injuries is a promising approach, there is growing evidence that the therapeutic efficacy of stem cells originates from secreted extracellular vesicles (EVs). Consequently, EVs are emerging as next-generation therapeutics. While EVs are extensively researched for diagnostic applications, their therapeutic potential to promote tissue repair is not fully elucidated. By housing and delivering tissue-repairing cargo, EVs refine the cellular microenvironment, modulate inflammation, and ultimately repair injury. Here, the potential use of EVs derived from two placental mesenchymal stem/stromal cell (MSC) lines is presented; a chorionic MSC line (CMSC29) and a decidual MSC cell line (DMSC23) for applications in lung diseases. Functional analyses using in vitro models of injury demonstrate that these EVs have a role in ameliorating injuries caused to lung cells. It is also shown that EVs promote repair of lung epithelial cells. This study is fundamental to advancing the field of EVs and to unlock the full potential of EVs in regenerative medicine.
Collapse
Affiliation(s)
- Sally Yunsun Kim
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| | - Mugdha V Joglekar
- Islet Biology and Diabetes Group, National Health and Medical Research Council Clinical Trials Center, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, 2050, Australia
| | - Anandwardhan A Hardikar
- Islet Biology and Diabetes Group, National Health and Medical Research Council Clinical Trials Center, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, 2050, Australia
| | - Thanh Huyen Phan
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| | - Dipesh Khanal
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| | - Priyanka Tharkar
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| | - Christina Limantoro
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| | - Jancy Johnson
- Department of Maternal fetal Medicine, Royal Women's Hospital, Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bill Kalionis
- Department of Maternal fetal Medicine, Royal Women's Hospital, Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Wojciech Chrzanowski
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, New South Wales, 2006, Australia.,Nano Institute, The University of Sydney, New South Wales, 2006, Australia
| |
Collapse
|
19
|
Jedari B, Rahmani A, Naderi M, Nadri S. MicroRNA‐7 promotes neural differentiation of trabecular meshwork mesenchymal stem cell on nanofibrous scaffold. J Cell Biochem 2019; 121:2818-2827. [DOI: 10.1002/jcb.29513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/08/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Behrouz Jedari
- Department of Medical BiotechnologyZanjan University of Medical SciencesZanjan Iran
| | - Ali Rahmani
- Department of Medical NanotechnologyZanjan University of Medical SciencesZanjan Iran
| | - Mahmood Naderi
- Cell‐Based Therapies Research Center, Digestive Disease Research InstituteTehran University of Medical SciencesTehran Iran
| | - Samad Nadri
- Department of Medical NanotechnologyZanjan University of Medical SciencesZanjan Iran
- Zanjan Metabolic Diseases Research CenterZanjan University of Medical SciencesZanjan Iran
- Zanjan Pharmaceutical Nanotechnology Research CenterZanjan University of Medical SciencesZanjan Iran
| |
Collapse
|
20
|
Li X, Li N, Chen K, Nagasawa S, Yoshizawa M, Kagami H. Around 90° Contact Angle of Dish Surface Is a Key Factor in Achieving Spontaneous Spheroid Formation. Tissue Eng Part C Methods 2019; 24:578-584. [PMID: 30234440 DOI: 10.1089/ten.tec.2018.0188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Following the discovery of the primary culture of neural stem cells, the spheroid culture has been recognized as one of the selective culture methods for somatic stem cells. Since then, various methods were reported to generate spheroids, which can enrich the potent stem cell population. However, the fundamental factors affecting spheroid formation remain unclear. In this study, we focused on the surface property of the culture dishes, in particular, hydrophobicity. Primary mouse skin culture cells were prepared with conventional two-dimensional culture, and then, the cells were transferred to culture dishes with varying hydrophobicity, which was confirmed with the water contact angles. Of these, a culture dish possessing an almost 90° water contact angle was the only one that successfully exhibited spheroid formation. The spheroid formation was spontaneous, efficient, and stable. Since this outcome was achieved with a conventional culture medium with serum, but without any additives such as epidermal growth factor, basic fibroblast growth factor, and B27, the spheroid formation from this process was not affected by serum and was also not dependent on additives. The results from immunofluorescence and quantitative real-time polymerase chain reaction testing showed the expression of embryonic stem cell markers such as SSEA-1, SOX2, OCT4, and Nanog, which confirmed that the spheroids with this method are comparable to those from other methods. This outcome was reproducible and could be applied not only to skin-derived cells but also to oral mucosa-derived cells, cortical bone-derived cells, and 3T3 cells, also suggesting the generality and robustness of this phenomenon.
Collapse
Affiliation(s)
- Xianqi Li
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University , Shiojiri, Japan .,2 Institute of Oral Science, Matsumoto Dental University , Shiojiri, Japan .,3 Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University , Shiojiri, Japan
| | - Ni Li
- 3 Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University , Shiojiri, Japan
| | - Kai Chen
- 3 Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University , Shiojiri, Japan
| | - Sakae Nagasawa
- 4 Department of Dental Material Science, School of Dentistry, Matsumoto Dental University , Shiojiri, Japan
| | - Michiko Yoshizawa
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University , Shiojiri, Japan .,3 Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University , Shiojiri, Japan
| | - Hideaki Kagami
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University , Shiojiri, Japan .,2 Institute of Oral Science, Matsumoto Dental University , Shiojiri, Japan .,3 Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University , Shiojiri, Japan .,5 Department of General Medicine, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| |
Collapse
|
21
|
Spontaneously Formed Spheroids from Mouse Compact Bone-Derived Cells Retain Highly Potent Stem Cells with Enhanced Differentiation Capability. Stem Cells Int 2019; 2019:8469012. [PMID: 31191686 PMCID: PMC6525826 DOI: 10.1155/2019/8469012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/07/2023] Open
Abstract
The results from our recent study showed the presence of two distinct spheroid-forming mechanisms, i.e., spontaneous and mechanical. In this study, we focused on the spontaneously formed spheroids, and the character of spontaneously formed spheroids from mouse compact bone-derived cells (CBDCs) was explored. Cells from (C57BL/6J) mouse leg bones were isolated, and compact bone-derived cells were cultured after enzymatic digestion. Spontaneous spheroid formation was achieved on a culture plate with specific water contact angle as reported. The expression levels of embryonic stem cell markers were analyzed using immunofluorescence and quantitative reverse transcription polymerase chain reaction. Then, the cells from spheroids were induced into osteogenic and neurogenic lineages. The spontaneously formed spheroids from CBDCs were positive for ES cell markers such as SSEA1, Sox2, Oct4, and Nanog. Additionally, the expressions of fucosyltransferase 4/FUT4 (SSEA1), Sox2, and Nanog were significantly higher than those in monolayer cultured cells. The gene expression of mesenchymal stem cell markers was almost identical in both spheroids and monolayer-cultured cells, but the expression of Sca-1 was higher in spheroids. Spheroid-derived cells showed significantly higher osteogenic and neurogenic marker expression than monolayer-cultured cells after induction. Spontaneously formed spheroids expressed stem cell markers and showed enhanced osteogenic and neurogenic differentiation capabilities than cells from the conventional monolayer culture, which supports the superior stemness.
Collapse
|
22
|
Lee JE, Yin Y, Lim SY, Kim ES, Jung J, Kim D, Park JW, Lee MS, Jeong JH. Enhanced Transfection of Human Mesenchymal Stem Cells Using a Hyaluronic Acid/Calcium Phosphate Hybrid Gene Delivery System. Polymers (Basel) 2019; 11:polym11050798. [PMID: 31060246 PMCID: PMC6571843 DOI: 10.3390/polym11050798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 12/18/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) show enormous potential in regenerative medicine and tissue engineering. However, current use of hMSCs in clinics is still limited because there is no appropriate way to control their behavior in vivo, such as differentiation to a desired cell type. Genetic modification may provide an opportunity to control the cells in an active manner. One of the major hurdles for genetic manipulation of hMSCs is the lack of an efficient and safe gene delivery system. Herein, biocompatible calcium phosphate (CaP)-based nanoparticles stabilized with a catechol-derivatized hyaluronic acid (dopa-HA) conjugate were used as a carrier for gene transfection to hMSCs for improved differentiation. Owing to the specific interactions between HA and CD44 of bone marrow-derived hMSCs, dopa-HA/CaP showed significantly higher transfection in hMSCs than branched polyethylenimine (bPEI, MW 25 kDa) with no cytotoxicity. The co-delivery of a plasmid DNA encoding bone morphogenetic protein 2 (BMP-2 pDNA) and micro RNA 148b (miRNA-148b) by dopa-HA/CaP achieved significantly improved osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Jung Eun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Yue Yin
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Su Yeon Lim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - E Seul Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Jaeback Jung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Dahwun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Ji Won Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Min Sang Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| |
Collapse
|
23
|
Mashayekhi P, Noruzinia M, Zeinali S, Khodaverdi S. Endometriotic Mesenchymal Stem Cells Epigenetic Pathogenesis: Deregulation of miR-200b, miR-145, and let7b in A Functional Imbalanced Epigenetic Disease. CELL JOURNAL 2019; 21:179-185. [PMID: 30825291 PMCID: PMC6397607 DOI: 10.22074/cellj.2019.5903] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/07/2018] [Indexed: 12/31/2022]
Abstract
Objective Stem cell issue is a strong theory in endometriosis pathogenesis. It seems that endometriotic mesenchymal stem cells (MSCs) show different characteristics compared to the normal MSCs. Determined high proliferation and low differentiation/decidualization potential of endometriotic MSCs could be accompanied by their microRNAs deregulation influencing their fate and function. In this study for the first time, we evaluated the expression of miR-200b, miR-145, and let-7b in endometriotic compared to non-endometriotic MSCs. These microRNAs are involved in biological pathways related to proliferation and differentiation of stem cells. Their aberrant expressions can disturb the proliferation/ differentiation balance in stem cells, altering their function and causing various diseases, like endometriosis. Materials and Methods In this experimental study, MSCs were isolated from three endometriotic and three nonendometriotic eutopic endometrium, followed by their characterization and culture. Expression of miR-200b, miR-145, and let-7b was ultimately analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results We found that the expression of miR-200b was up-regulated (P<0.0001) whereas the expression of miR-145 and let-7b was down-regulated (P<0.0001) in endometriotic MSCs in comparison with non-endometriotic normal controls. Conclusion Proliferation and differentiation are important dynamic balanced biological processes, while in equillibrium, they determine a healthy stem cell fate. It seems that they are deregulated in endometriotic MSCs and change their function. miR-200b, miR-145, and let-7b are deregulated during endometriosis and they have pivotal roles in the modulating proliferation and differentiation of stem cells. We found up-regulation of miR-200b and down-regulation of miR-145 and let-7b in endometriotic MSCs. These changes can increase self-renewal and migration, while decreasing differentiation of endometriotic MSCs. Our achievements emphasize previous findings on the importance of proliferation/ differentiation balance in MSCs and clarify the role of microRNAs as main players in faulty endometriotic stem cells development.
Collapse
Affiliation(s)
- Parisa Mashayekhi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehrdad Noruzinia
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran. Electronic Address:
| | - Sirous Zeinali
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sepideh Khodaverdi
- Endometriosis Research Center, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
24
|
Smieszek A, Kornicka K, Szłapka-Kosarzewska J, Androvic P, Valihrach L, Langerova L, Rohlova E, Kubista M, Marycz K. Metformin Increases Proliferative Activity and Viability of Multipotent Stromal Stem Cells Isolated from Adipose Tissue Derived from Horses with Equine Metabolic Syndrome. Cells 2019; 8:E80. [PMID: 30678275 PMCID: PMC6406832 DOI: 10.3390/cells8020080] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, we investigated the influence of metformin (MF) on proliferation and viability of adipose-derived stromal cells isolated from horses (EqASCs). We determined the effect of metformin on cell metabolism in terms of mitochondrial metabolism and oxidative status. Our purpose was to evaluate the metformin effect on cells derived from healthy horses (EqASCHE) and individuals affected by equine metabolic syndrome (EqASCEMS). The cells were treated with 0.5 μM MF for 72 h. The proliferative activity was evaluated based on the measurement of BrdU incorporation during DNA synthesis, as well as population doubling time rate (PDT) and distribution of EqASCs in the cell cycle. The influence of metformin on EqASC viability was determined in relation to apoptosis profile, mitochondrial membrane potential, oxidative stress markers and BAX/BCL-2 mRNA ratio. Further, we were interested in possibility of metformin affecting the Wnt3a signalling pathway and, thus, we determined mRNA and protein level of WNT3A and β-catenin. Finally, using a two-tailed RT-qPCR method, we investigated the expression of miR-16-5p, miR-21-5p, miR-29a-3p, miR-140-3p and miR-145-5p. Obtained results indicate pro-proliferative and anti-apoptotic effects of metformin on EqASCs. In this study, MF significantly improved proliferation of EqASCs, which manifested in increased synthesis of DNA and lowered PDT value. Additionally, metformin improved metabolism and viability of cells, which correlated with higher mitochondrial membrane potential, reduced apoptosis and increased WNT3A/β-catenin expression. Metformin modulates the miRNA expression differently in EqASCHE and EqASCEMS. Metformin may be used as a preconditioning agent which stimulates proliferative activity and viability of EqASCs.
Collapse
Affiliation(s)
- Agnieszka Smieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Jolanta Szłapka-Kosarzewska
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Peter Androvic
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Laboratory of Growth Regulators, Faculty of Science, Palacky University, 78371 Olomouc, Czech Republic.
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
| | - Lucie Langerova
- Gene Core BIOCEV, Průmyslová 595, Vestec 252 50, Czech Republic.
| | - Eva Rohlova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, 128 43 Prague, Czech Republic.
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- TATAA Biocenter AB, 411 03 Gothenburg, Sweden.
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Giessen, Germany.
| |
Collapse
|
25
|
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNA molecules involved in the regulation of gene expression. They are involved in the fine-tuning of fundamental biological processes such as proliferation, differentiation, survival and apoptosis in many cell types. Emerging evidence suggests that miRNAs regulate critical pathways involved in stem cell function. Several miRNAs have been suggested to target transcripts that directly or indirectly coordinate the cell cycle progression of stem cells. Moreover, previous studies have shown that altered expression levels of miRNAs can contribute to pathological conditions, such as cancer, due to the loss of cell cycle regulation. However, the precise mechanism underlying miRNA-mediated regulation of cell cycle in stem cells is still incompletely understood. In this review, we discuss current knowledge of miRNAs regulatory role in cell cycle progression of stem cells. We describe how specific miRNAs may control cell cycle associated molecules and checkpoints in embryonic, somatic and cancer stem cells. We further outline how these miRNAs could be regulated to influence cell cycle progression in stem cells as a potential clinical application.
Collapse
Affiliation(s)
- Michelle M J Mens
- Department of Epidemiology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands. .,Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Gao X, Petricoin EF, Ward KR, Goldberg SR, Duane TM, Bonchev D, Arodz T, Diegelmann RF. Network proteomics of human dermal wound healing. Physiol Meas 2018; 39:124002. [PMID: 30524050 DOI: 10.1088/1361-6579/aaee19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The healing of wounds is critical in protecting the human body against environmental factors. The mechanisms involving protein expression during this complex physiological process have not been fully elucidated. APPROACH Here, we use reverse-phase protein microarrays (RPPA) involving 94 phosphoproteins to study tissue samples from tubes implanted in healing dermal wounds in seven human subjects tracked over two weeks. We compare the proteomic profiles to proteomes of controls obtained from skin biopsies from the same subjects. MAIN RESULTS Compared to previous proteomic studies of wound healing, our approach focuses on wound tissue instead of wound fluid, and has the sensitivity to go beyond measuring only highly abundant proteins. To study the temporal dynamics of networks involved in wound healing, we applied two network analysis methods that integrate the experimental results with prior knowledge about protein-protein physical and regulatory interactions, as well as higher-level biological processes and associated pathways. SIGNIFICANCE We uncovered densely connected networks of proteins that are up- or down-regulated during human wound healing, as well as their relationships to microRNAs and to proteins outside of our set of targets that we measured with proteomic microarrays.
Collapse
Affiliation(s)
- Xi Gao
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Zammit V, Brincat MR, Cassar V, Muscat-Baron Y, Ayers D, Baron B. MiRNA influences in mesenchymal stem cell commitment to neuroblast lineage development. Noncoding RNA Res 2018; 3:232-242. [PMID: 30533571 PMCID: PMC6257889 DOI: 10.1016/j.ncrna.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal Stem Cells (MSCs) are widely used in therapeutic applications. Their plasticity and predisposition to differentiate into a variety of cell types, including those of the neuronal lineage, makes them ideal to study whether a selection of miRNAs may direct the differentiation of MSCs into neuroblasts or neuroblastoma to mature neurons. Following a short-listing, miR-107, 124 and 381 were selected as the most promising candidates for this differentiation. MSCs differentiated into cells of the neural lineage (Conditioned Cells) upon addition of conditioned medium (rich in microvesicles containing miRNAs) obtained from cultured SH-SY5Y neuroblastoma cells. Characterisation of stemness (including SOX2, OCT4, Nanog and HCG) and neural markers (including Nestin, MASH1, TUBB3 and NeuN1) provided insight regarding the neuronal state of each cell type. This was followed by transfection of the three miRNA antagonists and mimics, and quantification of their respective target genes. MiRNA target gene expression following transfection of MSCs with miRNA inhibitors and mimics demonstrated that these three miRNAs were not sufficient to induce differentiation. In conditioned cells the marginal changes in the miRNA target expression levels reflected potential for the modulation of intermediate neural progenitors and immature neuron cell types. Transfection of various combinations of miRNA inhibitors and/or mimics revealed more promise. Undoubtedly, a mix of biomolecules is being released by the SH-SY5Y in culture that induce MSCs to differentiate. Screening for those biomolecules acting synergistically with specific miRNAs will allow further combinatorial testing to elucidate the role of miRNA modulation.
Collapse
Affiliation(s)
- Vanessa Zammit
- National Blood Transfusion Service, St. Luke's Hospital, G'Mangia, PTA1010, Malta.,School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | - Mark R Brincat
- Dept. of Obstetrics & Gynaecology, Mater Dei Hospital, Msida, MSD2090, Malta
| | - Viktor Cassar
- Dept. of Obstetrics & Gynaecology, Mater Dei Hospital, Msida, MSD2090, Malta
| | - Yves Muscat-Baron
- Dept. of Obstetrics & Gynaecology, Mater Dei Hospital, Msida, MSD2090, Malta
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta.,School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| |
Collapse
|
28
|
Dietrich C, Singh M, Kumar N, Singh SR. The Emerging Roles of microRNAs in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:11-26. [PMID: 29754172 DOI: 10.1007/978-3-319-74470-4_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is the continuous loss of tissue and organ function over time. MicroRNAs (miRNAs) are thought to play a vital role in this process. miRNAs are endogenous small noncoding RNAs that control the expression of target mRNA. They are involved in many biological processes such as developmental timing, differentiation, cell death, stem cell proliferation and differentiation, immune response, aging and cancer. Accumulating studies in recent years suggest that miRNAs play crucial roles in stem cell division and differentiation. In the present chapter, we present a brief overview of these studies and discuss their contributions toward our understanding of the importance of miRNAs in normal and aged stem cell function in various model systems.
Collapse
Affiliation(s)
- Catharine Dietrich
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Manish Singh
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Nishant Kumar
- Hospitalist Division, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
| | - Shree Ram Singh
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
29
|
Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci 2018; 6:60-78. [PMID: 29184934 DOI: 10.1039/c7bm00479f] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
Collapse
Affiliation(s)
- I M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | | | | | | | | |
Collapse
|
30
|
Wu Z, Qiu X, Gao B, Lian C, Peng Y, Liang A, Xu C, Gao W, Zhang L, Su P, Rong L, Huang D. Melatonin-mediated miR-526b-3p and miR-590-5p upregulation promotes chondrogenic differentiation of human mesenchymal stem cells. J Pineal Res 2018; 65:e12483. [PMID: 29498095 DOI: 10.1111/jpi.12483] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs), with inherent chondrogenic differentiation potential appear to be ideally suited for therapeutic use in cartilage regeneration. Accumulating evidence has demonstrated that melatonin can promote chondrogenic differentiation in human BMSCs. However, little is known about the mechanism. MicroRNAs (miRNAs) have been shown to regulate the differentiation of BMSCs, but their roles in melatonin-promoted chondrogenic differentiation have not been characterized. Here, we demonstrate that melatonin promoted chondrogenic differentiation of human BMSCs via upregulation of miR-526b-3p and miR-590-5p. Mechanistically, the elevated miR-526b-3p and miR-590-5p enhanced SMAD1 phosphorylation by targeting SMAD7. Additionally, administration of miR-526b-3p mimics or miR-590-5p mimics successfully promoted the chondrogenic differentiation of human BMSCs. Collectively, our study suggests that modification of BMSCs using melatonin or miRNA transduction could be an effective therapy for cartilage damage and degeneration.
Collapse
Affiliation(s)
- Zizhao Wu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bo Gao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Caixia Xu
- Research Centre for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peiqiang Su
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Deciphering synergistic regulatory networks of microRNAs in hESCs and fibroblasts. Int J Biol Macromol 2018; 113:1279-1286. [DOI: 10.1016/j.ijbiomac.2018.03.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/06/2018] [Accepted: 03/11/2018] [Indexed: 12/14/2022]
|
32
|
Inhibition of microRNA-138 enhances bone formation in multiple myeloma bone marrow niche. Leukemia 2018; 32:1739-1750. [PMID: 29925904 DOI: 10.1038/s41375-018-0161-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/25/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022]
Abstract
Myeloma bone disease is a devastating complication of multiple myeloma (MM) and is caused by dysregulation of bone remodeling processes in the bone marrow microenvironment. Previous studies showed that microRNA-138 (miR-138) is a negative regulator of osteogenic differentiation of mesenchymal stromal cells (MSCs) and that inhibiting its function enhances bone formation in vitro. In this study, we explored the role of miR-138 in myeloma bone disease and evaluated the potential of systemically delivered locked nucleic acid (LNA)-modified anti-miR-138 oligonucleotides in suppressing myeloma bone disease. We showed that expression of miR-138 was significantly increased in MSCs from MM patients (MM-MSCs) and myeloma cells compared to those from healthy subjects. Furthermore, inhibition of miR-138 resulted in enhanced osteogenic differentiation of MM-MSCs in vitro and increased the number of endosteal osteoblastic lineage cells (OBCs) and bone formation rate in mouse models of myeloma bone disease. RNA sequencing of the OBCs identified TRPS1 and SULF2 as potential miR-138 targets that were de-repressed in anti-miR-138-treated mice. In summary, these data indicate that inhibition of miR-138 enhances bone formation in MM and that pharmacological inhibition of miR-138 could represent a new therapeutic strategy for treatment of myeloma bone disease.
Collapse
|
33
|
Han S, Kang B, Jang E, Ki J, Kim E, Jeong MY, Huh YM, Son HY, Haam S. Convenient Monitoring System of Intracellular microRNA Expression during Adipogenesis via Mechanical Stimulus-Induced Exocytosis of Lipovesicular miRNA Beacon. Adv Healthc Mater 2018; 7. [PMID: 29280320 DOI: 10.1002/adhm.201701019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/11/2017] [Indexed: 01/01/2023]
Abstract
Noninvasive investigation of microRNAs (miRNAs) expression, which is deeply related to biological phenomena such as stem cell differentiation, in culture soup is particularly useful for monitoring of stem cell differentiation without phototoxicity of living cells, especially when cell morphologies remain unchanged during differentiation. However, real-time detection of miRNA in culture soup is not recommended because of insufficient miRNA amounts in culture soup. In this study, a convenient method is introduced for real-time assessing intracellular miRNA in culture soup by using lipovesicular miRNA beacon (Lipo-mB) and mechanical stimulus-mediated exocytosis. Pipetting-harvest of culture soup induces exocytosis-secretion of fluorescence signal of Lipo-mB from cytoplasm into culture soup. To demonstrate this method, Lipo-mB is applied for monitoring of adipogenesis by analyzing the expression levels of various intracellular miRNAs, which are related to adipogenesis regulators. The fluorescence intensity profile of the culture soup is correlated with the quantitative reverse-transcription-polymerase chain reaction data and absorbance of Oil Red O staining. These results demonstrate that Lipo-mB can successfully monitor stem cell differentiation by sensing changes in miRNA expression from culture soup of living cells. Lipo-mB can be further developed as an accurate sensing system for analyzing subtle differences in genotype, even when changes in phenotype cannot be observed.
Collapse
Affiliation(s)
- Seungmin Han
- Department of Chemical and Biomolecular Engineering; Yonsei University; Seoul 120-749 Republic of Korea
| | - Byunghoon Kang
- Department of Chemical and Biomolecular Engineering; Yonsei University; Seoul 120-749 Republic of Korea
| | - Eunji Jang
- Department of Radiology; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
| | - Jisun Ki
- Department of Chemical and Biomolecular Engineering; Yonsei University; Seoul 120-749 Republic of Korea
| | - Eunjung Kim
- Department of Materials; Department of Bioengineering and Institute for Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Mun-Young Jeong
- Department of Radiology; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
| | - Yong-Min Huh
- Department of Radiology; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
- Severance Biomedical Science Institute; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
- YUHS-KRIBB Medical Convergence Research Institute; Seoul 120-752 Republic of Korea
- Brain Korea 21 Project for Medical Science; Yonsei University College of Medicine; Seoul 120-752 Republic of Korea
| | - Hye-Young Son
- Department of Radiology; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
- Severance Biomedical Science Institute; College of Medicine; Yonsei University; Seoul 120-752 Republic of Korea
- YUHS-KRIBB Medical Convergence Research Institute; Seoul 120-752 Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering; Yonsei University; Seoul 120-749 Republic of Korea
| |
Collapse
|
34
|
Rahmati M, Pennisi CP, Mobasheri A, Mozafari M. Bioengineered Scaffolds for Stem Cell Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:73-89. [DOI: 10.1007/5584_2018_215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
The Effects of BMP-2, miR-31, miR-106a, and miR-148a on Osteogenic Differentiation of MSCs Derived from Amnion in Comparison with MSCs Derived from the Bone Marrow. Stem Cells Int 2017; 2017:7257628. [PMID: 29348760 PMCID: PMC5733904 DOI: 10.1155/2017/7257628] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/13/2017] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) offering valuable anticipations for the treatment of degenerative diseases. They can be found in many tissues including amnion. MSCs from amnion (AM-MSCs) can differentiate into osteoblast similar to that of bone marrow-derived MSCs (BM-MSCs). However, the ability is not much efficient compared to BM-MSCs. This study aimed to examine the effects of BMP-2 and miRNAs on osteogenic differentiation of AM-MSCs compared to those of BM-MSCs. The osteogenic differentiation capacity after miRNA treatment was assessed by ALP expression, ALP activity, and osteogenic marker gene expression. The results showed that the osteogenic differentiation capacity increased after BMP-2 treatment both in AM-MSCs and BM-MSCs. MiR-31, miR-106a, and miR-148a were downregulated during the osteogenic differentiation. After transfection with anti-miRNAs, ALP activity and osteogenic genes were increased over the time of differentiation. The data lead to the potential for using AM-MSCs as an alternative source for bone regeneration. Moreover, the information of miRNA expression and function during osteogenic differentiation may be useful for the development of new therapeutics or enhanced an in vitro culture technique required for stem cell-based therapies in the bone regeneration.
Collapse
|
36
|
Costa V, Raimondi L, Conigliaro A, Salamanna F, Carina V, De Luca A, Bellavia D, Alessandro R, Fini M, Giavaresi G. Hypoxia-inducible factor 1Α may regulate the commitment of mesenchymal stromal cells toward angio-osteogenesis by mirna-675-5P. Cytotherapy 2017; 19:1412-1425. [PMID: 29111380 DOI: 10.1016/j.jcyt.2017.09.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/30/2017] [Accepted: 09/10/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS During bone formation, angiogenesis and osteogenesis are regulated by hypoxia, which is able to induce blood vessel formation, as well as recruit and differentiate human mesenchymal stromal cells (hMSCs). The molecular mechanisms involved in HIF-1α response and hMSC differentiation during bone formation are still unclear. This study aimed to investigate the synergistic role of hypoxia and hypoxia-mimetic microRNA miR-675-5p in angiogenesis response and osteo-chondroblast commitment of hMSCs. METHODS By using a suitable in vitro cell model of hMSCs (maintained in hypoxia or normoxia), the role of HIF-1α and miR-675-5p in angiogenesis and osteogenesis coupling was investigated, using fluorescence-activated cell sorting (FACS), gene expression and protein analysis. RESULTS Hypoxia induced miR-675-5p expression and a hypoxia-angiogenic response, as demonstrated by increase in vascular endothelial growth factor messenger RNA and protein release. MiR-675-5p overexpression in normoxia promoted the down-regulation of MSC markers and the up-regulation of osteoblast and chondroblast markers, as demonstrated by FACS and protein analysis. Moreover, miR-675-5p depletion in a low-oxygen condition partially abolished the hypoxic response, including angiogenesis, and in particular restored the MSC phenotype, demonstrated by cytofluorimetric analysis. In addition, current preliminary data suggest that the expression of miR-675-5p during hypoxia plays an additive role in sustaining Wnt/β-catenin pathways and the related commitment of hMSCs during bone ossification. DISCUSSION MiR-675-5p may trigger complex molecular mechanisms that promote hMSC osteoblastic differentiation through a dual strategy: increasing HIF-1α response and activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Viviana Costa
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy.
| | - Lavinia Raimondi
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy
| | - Alice Conigliaro
- Department of Cellular Biotechnology and Hematology, Sapienza University of Rome, Rome, Italy
| | - Francesca Salamanna
- Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - Valeria Carina
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy
| | - Angela De Luca
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy
| | - Daniele Bellavia
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy
| | - Riccardo Alessandro
- Department of Biopathology and Medical Biotechnologies, Section of Biology and Genetics, University of Palermo, Palermo, Italy; Institute of Biomedicine and Molecular Immunology, National Research Council, Palermo, Italy
| | - Milena Fini
- Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - Gianluca Giavaresi
- Rizzoli Orthopedic Institute, Bologna, Italy; Rizzoli Orthopedic Institute, Innovative Technological Platforms for Tissue Engineering, Theranostic and Oncology, Palermo, Italy; Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| |
Collapse
|
37
|
Fatima F, Ekstrom K, Nazarenko I, Maugeri M, Valadi H, Hill AF, Camussi G, Nawaz M. Non-coding RNAs in Mesenchymal Stem Cell-Derived Extracellular Vesicles: Deciphering Regulatory Roles in Stem Cell Potency, Inflammatory Resolve, and Tissue Regeneration. Front Genet 2017; 8:161. [PMID: 29123544 PMCID: PMC5662888 DOI: 10.3389/fgene.2017.00161] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous populations of nano- and micro-sized vesicles secreted by various cell types. There is mounting evidence that EVs have widespread roles in transporting proteins, lipids, and nucleic acids between cells and serve as mediators of intercellular communication. EVs secreted from stem cells could function as paracrine factors, and appear to mimic and recapitulate several features of their secreting cells. EV-mediated transport of regulatory RNAs provides a novel source of trans-regulation between cells. As such, stem cells have evolved unique forms of paracrine mechanisms for recapitulating their potencies with specialized functions by transporting non-coding RNAs (ncRNAs) via EVs. This includes the dissemination of stem cell-derived EV-ncRNAs and their regulatory effects elicited in differentiation, self-renewal, pluripotency, and the induction of reparative programs. Here, we summarize and discuss the therapeutic effects of mesenchymal stem cell-derived EV-ncRNAs in the induction of intrinsic regenerative programs elicited through regulating several mechanisms. Among them, most noticeable are the EV-mediated enrichment of ncRNAs at the injury sites contributing the regulation of matrix remodeling, epithelial mesenchymal transitions, and attraction of fibroblasts. Additionally, we emphasize EV-mediated transmission of anti-inflammatory RNAs from stem cells to injury site that potentially orchestrate the resolution of the inflammatory responses and immune alleviation to better facilitate healing processes. Collectively, this knowledge indicates a high value and potential of EV-mediated RNA-based therapeutic approaches in regenerative medicine.
Collapse
Affiliation(s)
- Farah Fatima
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Karin Ekstrom
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Irina Nazarenko
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Centre, University of Freiburg, Freiburg, Germany
| | - Marco Maugeri
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Giovanni Camussi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Muhammad Nawaz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Nawaz M. Extracellular vesicle-mediated transport of non-coding RNAs between stem cells and cancer cells: implications in tumor progression and therapeutic resistance. Stem Cell Investig 2017; 4:83. [PMID: 29167804 DOI: 10.21037/sci.2017.10.04] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
Recent years have witnessed intensive progress in studying extracellular vesicles (EVs), both for understanding their basic biology and contribution to variety of diseases, biomarker discovery, and their potential as gene delivery vectors and source of innovative therapies. As such, stem cell-derived EVs have contributed significant knowledge which led to the development of cell-free therapies in regenerative medicine. Although, the role of stem cell-derived EVs in maintaining stemness, differentiation and repairing tissue injuries is relatively well-understood; however, knowledge about the contribution of stem cell-derived EVs in cancer progression is just emerging. The aim of this review is, therefore, to discuss the recent developments in stem cell-derived EVs and tumor progression, placing a particular focus on non-coding RNA (ncRNA) mediated cancer progression and resistance against therapies. This includes the failure of normal hematopoiesis and the progression of myeloid neoplasms, enhanced capacity of cancer cells to proliferate and metastasize, and the conversion of normal cells into cancer cells, activation of angiogenic pathways and dormancy in cancer cells. These processes are shared by mesenchymal stem cells (MSCs), cancer stem like-cells and cancer cells in an intricate intratumoral network in order to create self-strengthening tumor niche. In this context, EV-ncRNAs serve as mediators to relay bystander effects of secreting cancer stem cells (CSCs) into recipient cells for priming a tumor permissive environment and relaying therapeutic resistance. Collectively, this knowledge will improve our understandings and approaches in finding new therapeutic targets in the context of CSCs, which could be benefited through engineering EVs for innovative therapies.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
39
|
Mayer U, Benditz A, Grässel S. miR-29b regulates expression of collagens I and III in chondrogenically differentiating BMSC in an osteoarthritic environment. Sci Rep 2017; 7:13297. [PMID: 29038440 PMCID: PMC5643533 DOI: 10.1038/s41598-017-13567-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is characterized by a slowly progressing, irreversible loss of articular cartilage. Tissue engineering approaches for cartilage regeneration include stem cell-based strategies but not much is known about their repair capacity in an OA microenvironment. The aim of the present study was to identify factors regulating collagen expression during chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSC) in an OA microenvironment. Coculture with OA cartilage induced miR-29b expression in BMSC which inhibited collagen I and III expression. Elevated miR-29b expression resulted in higher caspase 3/7 activity and promoted apoptosis of BMSC in part by directly inhibiting the anti-apoptotic proteins Bcl-2 and Mcl-1. Stimulation with IFN-γ induced miR-29b expression in BMSC. Our results suggest that miR-29b affects BMSC-based OA cartilage regeneration because expression of collagen III, mainly produced by undifferentiated BMSC, and collagen I, a marker for dedifferentiated chondrocytes, are inhibited by miR-29b thus influencing composition of the newly formed ECM. This might be critical to avoid formation of inferior fibrocartilage instead of hyaline cartilage. Furthermore, higher miR-29b expression promotes apoptosis either preventing excessive cell growth or reducing the number of BMSC undergoing chondrogenesis. Thus, miR-29b has both supportive but possibly also unfavourable effects on BMSC-based OA cartilage regeneration.
Collapse
Affiliation(s)
- Ute Mayer
- Department Orthopaedic Surgery, Exp. Orthopaedics, ZMB/Biopark 1, University of Regensburg, Regensburg, Germany.,Department Orthopaedic Surgery, Asklepiosklinikum, Bad Abbach, Germany
| | - Achim Benditz
- Department Orthopaedic Surgery, Asklepiosklinikum, Bad Abbach, Germany
| | - Susanne Grässel
- Department Orthopaedic Surgery, Exp. Orthopaedics, ZMB/Biopark 1, University of Regensburg, Regensburg, Germany. .,Department Orthopaedic Surgery, Asklepiosklinikum, Bad Abbach, Germany.
| |
Collapse
|
40
|
Hao Y, Ge Y, Li J, Hu Y, Wu B, Fang F. Identification of MicroRNAs by Microarray Analysis and Prediction of Target Genes Involved in Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. J Periodontol 2017; 88:1105-1113. [PMID: 28598283 DOI: 10.1902/jop.2017.170079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The roles of microRNAs (miRNAs) in osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) remain largely unexplored. In this study, the underlying molecular mechanism of osteogenic differentiation in hPDLSCs is investigated using miRNA profiling. METHODS The miRNA expression profile during osteogenic differentiation was analyzed using a microarray. Target genes of miRNAs with at least two-fold change in expression (P <0.05) were predicted by bioinformatics. Six miRNAs with osteogenesis-related target genes were validated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS Expression of 116 miRNAs was found to be altered after osteoinduction, with 30 upregulated and 86 downregulated. Thirty-one of these miRNAs (26.7%) had osteogenesis-related target genes. Changes in expression levels of six of the 31 miRNAs (miR-654-3p, miR-4288, miR-34c-5p, miR-218-5p, miR-663a, and miR-874-3p) were validated by qRT-PCR. CONCLUSIONS Significant alterations in miRNA expression profiles were observed during osteogenic differentiation of hPDLSCs. These results imply that miRNAs may have regulatory effects on this process by targeting osteogenesis-related genes.
Collapse
Affiliation(s)
- Yilin Hao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihong Ge
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianjia Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanwei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuchun Fang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Bellayr IH, Kumar A, Puri RK. MicroRNA expression in bone marrow-derived human multipotent Stromal cells. BMC Genomics 2017; 18:605. [PMID: 28800721 PMCID: PMC5553681 DOI: 10.1186/s12864-017-3997-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
Background Multipotent stromal cells (MSCs) are being studied in the field of regenerative medicine for their multi-lineage differentiation and immunoregulatory capacity. MicroRNAs (miRNAs) are short non-coding RNAs that are responsible for regulating gene expression by targeting transcripts, which can impact MSC functions such as cellular proliferation, differentiation, migration and cell death. miRNAs are expressed in MSCs; however, the impact of miRNAs on cellular functions and donor variability is not well understood. Eight MSC lines were expanded to passages 3, 5 and 7, and their miRNA expression was evaluated using microarray technology. Results Statistical analyses of our data revealed that 71 miRNAs out of 939 examined were expressed by this set of MSC lines at all passages and the expression of 11 miRNAs were significantly different between passages 3 and 7, while the expression of 7 miRNAs was significantly different between passages 3 and 5. The expression of these identified miRNAs was evaluated using RT-qPCR for both the first set of MSC lines (n = 6) and a second set of MSC lines (n = 7) expanded from passages 4 to 8. By RT-qPCR only 2 miRNAs, miR-638 and miR-572 were upregulated at passage 7 compared to passage 3 in the first set of MSC lines by 1.71 and 1.54 fold, respectively; and upregulated at passage 8 compared to passage 4 in the second set of MSC lines, 1.35 and 1.59 fold, respectively. Conclusions The expression of miR-638 and miR-572 can distinguish MSCs from two different passages of cell culture. These results may be useful in establishing critical quality attributes of MSCs and determining whether changes in these two miRNAs impact cellular functions. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3997-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ian H Bellayr
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Abhinav Kumar
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Raj K Puri
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
42
|
Hu F, Xu P, Sun B, Xiao Z. Differences in the MicroRNA profiles of subcutaneous adipose-derived stem cells and omental adipose-derived stem cells. Gene 2017; 625:55-63. [PMID: 28483594 DOI: 10.1016/j.gene.2017.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/30/2017] [Accepted: 05/04/2017] [Indexed: 01/09/2023]
Abstract
Adipose-derived stem cells (ASCs) isolated from subcutaneous (SC) and omentum (O) share similar characteristics, but the differences in their microRNA profiles are mostly unknown. In this study, besides significant differences in cell morphology and the differentiation ability of the two types of ASCs, the microRNA expression profiles of the cell lines were determined using SOLiD next-generation sequencing. The in-depth analysis found that miR-214, miR-222, miR-181a, miR-26a and miR-23/27/24 clusters and miR-375 act as "markers" to distinguish the different fat deposit-derived ASCs. Additionally, the global miRNA-mRNA interaction differences were revealed, and the results of the GO term enrichment and KEGG pathway in the DAVID tool showed that the molecular function, biological process and signaling pathways showed some different in the two types of ASCs. Our findings provided a clue to a more thorough understanding of the difference between SC-ASCs and O-ASCs and indicate their different potentials for clinical use.
Collapse
Affiliation(s)
- Feihu Hu
- School of Biological Science and Medical Engineering, State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu, China; Medical School, Southeast University, Nanjing, Jiangsu, China
| | - Peng Xu
- School of Biological Science and Medical Engineering, State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu, China
| | - Bo Sun
- School of Biological Science and Medical Engineering, State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu, China
| | - Zhongdang Xiao
- School of Biological Science and Medical Engineering, State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
43
|
Pulsed Electromagnetic Field Regulates MicroRNA 21 Expression to Activate TGF- β Signaling in Human Bone Marrow Stromal Cells to Enhance Osteoblast Differentiation. Stem Cells Int 2017; 2017:2450327. [PMID: 28512472 PMCID: PMC5420424 DOI: 10.1155/2017/2450327] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/05/2017] [Accepted: 02/12/2017] [Indexed: 12/22/2022] Open
Abstract
Pulsed electromagnetic fields (PEMFs) have been documented to promote bone fracture healing in nonunions and increase lumbar spinal fusion rates. However, the molecular mechanisms by which PEMF stimulates differentiation of human bone marrow stromal cells (hBMSCs) into osteoblasts are not well understood. In this study the PEMF effects on hBMSCs were studied by microarray analysis. PEMF stimulation of hBMSCs' cell numbers mainly affected genes of cell cycle regulation, cell structure, and growth receptors or kinase pathways. In the differentiation and mineralization stages, PEMF regulated preosteoblast gene expression and notably, the transforming growth factor-beta (TGF-β) signaling pathway and microRNA 21 (miR21) were most highly regulated. PEMF stimulated activation of Smad2 and miR21-5p expression in differentiated osteoblasts, and TGF-β signaling was essential for PEMF stimulation of alkaline phosphatase mRNA expression. Smad7, an antagonist of the TGF-β signaling pathway, was found to be miR21-5p's putative target gene and PEMF caused a decrease in Smad7 expression. Expression of Runx2 was increased by PEMF treatment and the miR21-5p inhibitor prevented the PEMF stimulation of Runx2 expression in differentiating cells. Thus, PEMF could mediate its effects on bone metabolism by activation of the TGF-β signaling pathway and stimulation of expression of miR21-5p in hBMSCs.
Collapse
|
44
|
Huleihel L, Sellares J, Cardenes N, Álvarez D, Faner R, Sakamoto K, Yu G, Kapetanaki MG, Kaminski N, Rojas M. Modified mesenchymal stem cells using miRNA transduction alter lung injury in a bleomycin model. Am J Physiol Lung Cell Mol Physiol 2017; 313:L92-L103. [PMID: 28385811 DOI: 10.1152/ajplung.00323.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 03/08/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Although different preclinical models have demonstrated a favorable role for bone marrow-derived mesenchymal stem cells (B-MSC) in preventing fibrosis, this protective effect is not observed with late administration of these cells, when fibrotic changes are consolidated. We sought to investigate whether the late administration of B-MSCs overexpressing microRNAs (miRNAs) let-7d (antifibrotic) or miR-154 (profibrotic) could alter lung fibrosis in a murine bleomycin model. Using lentiviral vectors, we transduced miRNAs (let-7d or miR-154) or a control sequence into human B-MSCs. Overexpression of let-7d or miR-154 was associated with changes in the mesenchymal properties of B-MSCs and in their cytokine expression. Modified B-MSCs were intravenously administered to mice at day 7 after bleomycin instillation, and the mice were euthanized at day 14 Bleomycin-injured animals that were treated with let-7d cells were found to recover quicker from the initial weight loss compared with the other treatment groups. Interestingly, animals treated with miR-154 cells had the lowest survival rate. Although a slight reduction in collagen mRNA levels was observed in lung tissue from let-7d mice, no significant differences were observed in Ashcroft score and OH-proline. However, the distinctive expression in cytokines and CD45-positive cells in the lung suggests that the differential effects observed in both miRNA mice groups were related to an effect on the immunomodulation function. Our results establish the use of miRNA-modified mesenchymal stem cells as a potential future research in lung fibrosis.
Collapse
Affiliation(s)
- Luai Huleihel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel.,Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University, New Haven, Connecticut
| | - Jacobo Sellares
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Servei de Pneumologia, Hospital Clínic, August Pi i Sunyer Biomedical Research Institute, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Institut Investigacions Biomèdiques, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; and
| | - Nayra Cardenes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Diana Álvarez
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rosa Faner
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Institut Investigacions Biomèdiques, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; and
| | - Koji Sakamoto
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University, New Haven, Connecticut
| | - Guoying Yu
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University, New Haven, Connecticut
| | - Maria G Kapetanaki
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University, New Haven, Connecticut
| | - Mauricio Rojas
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; .,The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
45
|
Klingelhöffer C, Codrin C, Ettl T, Reichert T, Morsczeck C. miRNA-101 supports the osteogenic differentiation in human dental follicle cells. Arch Oral Biol 2016; 72:47-50. [PMID: 27541634 DOI: 10.1016/j.archoralbio.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/16/2016] [Accepted: 08/03/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Human dental follicle cells (DFCs) are genuine precursor cells of cementoblasts and alveolar bone osteoblasts. MicroRNAs (miRNAs) represent a class of non-coding endogenous RNAs that silence gene expression post-transcriptionally. miRNA101 actively regulates the osteogenic differentiation of periodontal ligament cells. Therefore the aim of this study was to investigate the role of miRNA101 during the osteogenic differentiation in DFCs. MATERIALS AND METHODS DFCs were isolated, cultivated and osteogenic differentiated in differentiation medium. Total RNA including miRNAs was isolated and the expression of miRNA101 was examined by real-time RT-PCRs. The expression of miRNA101 was induced by miRNA101-mimic transfection and the gene expression of osteogenic transcription factors was obtained by real-time RT-PCRs. Moreover the induction of the osteogenic differentiation was evaluated by the activity of alkaline phosphatase. RESULTS miRNA101 was regulated in DFCs during the osteogenic differentiation. After miRNA101-mimic transfection the alkaline phosphatase was increased and the gene expression of typical osteogenic transcription factors such as SP7 (osterix) was up-regulated. CONCLUSION Our results suggest that miRNA101 sustains the osteogenic differentiation of DFCs.
Collapse
Affiliation(s)
- Christoph Klingelhöffer
- Dept. of Cranio- and Maxillofacial Surgery, Hospital of the University of Regensburg, Regensburg, Germany.
| | - Consuela Codrin
- Dept. of Cranio- and Maxillofacial Surgery, Hospital of the University of Regensburg, Regensburg, Germany
| | - Tobias Ettl
- Dept. of Cranio- and Maxillofacial Surgery, Hospital of the University of Regensburg, Regensburg, Germany
| | - Torsten Reichert
- Dept. of Cranio- and Maxillofacial Surgery, Hospital of the University of Regensburg, Regensburg, Germany
| | - Christian Morsczeck
- Dept. of Cranio- and Maxillofacial Surgery, Hospital of the University of Regensburg, Regensburg, Germany
| |
Collapse
|
46
|
Wang Y, He T, Liu J, Liu H, Zhou L, Hao W, Sun Y, Wang X. Synergistic effects of overexpression of BMP‑2 and TGF‑β3 on osteogenic differentiation of bone marrow mesenchymal stem cells. Mol Med Rep 2016; 14:5514-5520. [PMID: 27878265 PMCID: PMC5355709 DOI: 10.3892/mmr.2016.5961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 08/22/2016] [Indexed: 02/07/2023] Open
Abstract
Bone morphogenetic protein 2 (BMP-2) and transforming growth factor β (TGF-β) isoforms are important in advancing bone regeneration. The aim of the present study was to investigate the positive and reciprocal effect of TGF-β3, one of the three TGF-β isoforms, on BMP-2 in promoting osteogenic differentiation. Exogenous BMP-2 and TGF-β3 genes were separately, and in combination, overexpressed in rabbit bone marrow-derived mesenchymal stem cells (rBMSCs). Expression levels of BMP-2 and TGF-β3 were evaluated using reverse-transcription-polymerase chain reaction (RT-PCR) and Western blotting assays. Furthermore, the osteogenic differentiation capacities of BMSCs were assessed by measuring Alizarin Red S staining, an alkaline phosphatase activity assay, and quantification of the osteogenic-specific genes, Runt-related transcription factor 2 (Runx2) and Osterix (Osx). Using lentiviral-mediated transfection, robust co-transfection efficiency of >90% was achieved. RT-PCR and immunoblotting results indicated a marked elevated expression of BMP-2 and TGF-β3 in rBMSCs undergoing co-transfection, compared with transfection with BMP-2 or TGF-β3 alone, indicating that BMP-2 and TGF-β3 are synergistically expressed in rBMSCs. Furthermore, enhanced osteogenic differentiation was observed in rBMSCs co-transfected with BMP-2/TGF-β3. The present study successfully delivered BMP-2 together with TGF-β3 into rBMSCs with high efficiency for the first time. Furthermore, TGF-β3 overexpression was demonstrated to enhance the osteogenic efficacy of BMP-2 in rBMSCs, and vice versa. This provides a potential clinical therapeutic approach for regenerating the function of osseous tissue, and may present a promising strategy for bone defect healing.
Collapse
Affiliation(s)
- Yilin Wang
- Department of Biochip Laboratory, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Tian He
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Jie Liu
- Department of Biochip Laboratory, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Hongzhi Liu
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Lugang Zhou
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Wei Hao
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Yujie Sun
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| | - Xin Wang
- Department of Orthopedic Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, P.R. China
| |
Collapse
|
47
|
Malcolm DW, Sorrells JE, Van Twisk D, Thakar J, Benoit DSW. Evaluating side effects of nanoparticle-mediated siRNA delivery to mesenchymal stem cells using next generation sequencing and enrichment analysis. Bioeng Transl Med 2016; 1:193-206. [PMID: 27981244 PMCID: PMC5125403 DOI: 10.1002/btm2.10035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
RNA interference has immense potential to modulate cell functions. However, effective delivery of small interfering RNA (siRNA) while avoiding deleterious side effects has proven challenging. This study investigates both intended and unintended effects of diblock copolymer nanoparticle (NP) delivery of siRNA delivery to human mesenchymal stem cells (hMSC). Specifically, siRNA delivery was investigated at a range of NP‐siRNA:hMSC ratios with a focus on the effects of NP‐siRNA treatment on hMSC functions. Additionally, next generation RNA sequencing (RNAseq) was used with enrichment analysis to observe side effects in hMSC gene expression. Results show NP‐siRNA delivery is negatively correlated with hMSC density. However, higher NP‐siRNA:hMSC ratios increased cytotoxicity and decreased metabolic activity. hMSC proliferation was largely unaffected by NP‐siRNA treatment, except for a threefold reduction in hMSCs seeded at 4,000 cells/cm2. Flow cytometry reveals that apoptosis is a function of NP‐siRNA treatment time and seeding density; ∼14% of the treated hMSCs seeded at 8,000 cells/cm2 were annexin V+‐siRNA+ 24 hr after treatment, while 11% of the treated population was annexin V+‐siRNA−. RNAseq shows that NP‐siRNA treatment results in transcriptomic changes in hMSCs, while pathway analysis shows upregulation of apoptosis signaling and downregulation of metabolism, cell cycle, and DNA replication pathways, as corroborated by apoptosis, metabolism, and proliferation assays. Additionally, multiple innate immune signaling pathways such as toll‐like receptor, RIG‐I‐like receptor, and nuclear factor‐κB signaling pathways are upregulated. Furthermore, and consistent with traditional siRNA immune activation, cytokine–cytokine receptor signaling was also upregulated. Overall, this study provides insight into NP‐siRNA:hMSC ratios that are favorable for siRNA delivery. Moreover, NP‐siRNA delivery results in side effects across the hMSC transcriptome that suggest activation of the innate immunity that could alter MSC functions associated with their therapeutic potential.
Collapse
Affiliation(s)
- Dominic W Malcolm
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY14642
| | - Janet E Sorrells
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627
| | - Daniel Van Twisk
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627
| | - Juilee Thakar
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627; Dept. of Biostatistics and Computational Biology University of Rochester Rochester NY 14642
| | - Danielle S W Benoit
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY 14642; Dept. of Chemical Engineering University of Rochester Rochester NY 14627
| |
Collapse
|
48
|
Raisin S, Belamie E, Morille M. Non-viral gene activated matrices for mesenchymal stem cells based tissue engineering of bone and cartilage. Biomaterials 2016; 104:223-37. [PMID: 27467418 DOI: 10.1016/j.biomaterials.2016.07.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 12/22/2022]
|
49
|
Liu X, Xu H, Kou J, Wang Q, Zheng X, Yu T. MiR-9 promotes osteoblast differentiation of mesenchymal stem cells by inhibiting DKK1 gene expression. Mol Biol Rep 2016; 43:939-46. [PMID: 27393149 DOI: 10.1007/s11033-016-4030-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 06/15/2016] [Indexed: 12/28/2022]
Abstract
The aim of this study is to investigate the role of miR-9 and its mechanism on the osteoblast differentiation of mesenchymal stem cells. Real-time PCR and western blotting were used to study gene expression. Assay of Alkaline phosphatase activity and alizarin red staining were used to examine osteoblast differentiation. Transfection of miR-9 mimics or lent-shmiR-9 was used to modulate the level of miR-9 in C2C12. Overexpression of miR-9 in C2C12 cells stimulated alkaline phosphatase activity and osteoblast mineralization, as well as the expression of osteoblast marker genes Col I, Ocn and Bsp. Gene silencing of miR-9 in C2C12 resulted in the suppression of alkaline phosphatase activity and osteoblast mineralization, as well as the expression of Col I, Ocn and Bsp. DKK1 mRNA was not affected by miR-9 overexpression, however, DKK1 protein was significantly decreased. Moreover, DKK1 3'-UTR mediated transcriptional luciferase activity was also significantly suppressed by miR-9 overexpression. DKK1 mRNA was not affected by miR-9 gene silencing, however, DKK1 protein was significantly stimulated. Moreover, DKK1 3'-UTR mediated transcriptional luciferase activity was significantly stimulated by miR-9 gene silencing, and suppressed by miR-9 overexpression, however, DKK1 3'-UTR mutant mediated luciferase activity was unaffected. The siRNA derived gene silencing of DKK1 blocked the inhibiting effect of shmiR-9 on the expression of alkaline phosphatase; and blocked the inhibiting effect of shmiR-9 on the expression of ColI, Ocn and Bsp. MiR-9 promotes osteoblast differentiation of mesenchymal cell C2C12 by suppressing the gene expression of DKK1.
Collapse
Affiliation(s)
- Xiangyun Liu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hao Xu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003, China
| | - Jianqiang Kou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qianqian Wang
- Qingdao Central Blood Station, Qingdao, 266003, China
| | - Xiujun Zheng
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tengbo Yu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
50
|
Gong Y, Lu J, Yu X, Yu Y. Expression of Sp7 in Satb2-induced osteogenic differentiation of mouse bone marrow stromal cells is regulated by microRNA-27a. Mol Cell Biochem 2016; 417:7-16. [PMID: 27142530 DOI: 10.1007/s11010-016-2709-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/19/2016] [Indexed: 11/28/2022]
Abstract
Satb2 is a special AT-rich binding transcription factor essential for osteoblast differentiation and bone formation. Specific microRNAs (miRNAs) have been identified to regulate the complex process of osteogenic differentiation. It remains unclear how miRNA expressions is changed in the Satb2-induced osteogenic differentiation of bone marrow stromal cells (BMSCs). From the miRNA expression profile data collected by us from Satb2-induced osteogenic differentiation of mouse BMSCs, we found that miR-27a was significantly down-regulated relative to non-treated cells 7 days post induction. By in silico analysis, we identified Sp7 as a miR-27a targeting gene and verified the findings by Western blot analysis, qRT-PCR, and luciferase reporter assays. We also analyzed the function of miR-27a in osteogenic differentiation by transfection of exogenous miR-27a into BMSCs. Overexpression of miR-27a remarkably inhibited the expression of Sp7 and attenuated Satb2-induced osteogenic differentiation. Our results suggest that expression of Sp7 during the early stage of Satb2-induced osteogenic differentiation of BMSCs is regulated by miR-27a.
Collapse
Affiliation(s)
- Yiming Gong
- Department of Stomatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Jing Lu
- Shanghai Key Laboratory of Stomatology, Department of Orthodontics, School of Medicine, Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoping Yu
- Department of Stomatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Youcheng Yu
- Department of Stomatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| |
Collapse
|