201
|
Zhao H, Tang J, Zhou D, Weng Y, Qin W, Liu C, Lv S, Wang W, Zhao X. Electrospun Icariin-Loaded Core-Shell Collagen, Polycaprolactone, Hydroxyapatite Composite Scaffolds for the Repair of Rabbit Tibia Bone Defects. Int J Nanomedicine 2020; 15:3039-3056. [PMID: 32431500 PMCID: PMC7200251 DOI: 10.2147/ijn.s238800] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background Electrospinning is a widely used technology that can produce scaffolds with high porosity and surface area for bone regeneration. However, the small pore sizes in electrospun scaffolds constrain cell growth and tissue-ingrowth. In this study, novel drug-loading core-shell scaffolds were fabricated via electrospinning and freeze drying to facilitate the repair of tibia bone defects in rabbit models. Materials and Methods The collagen core scaffolds were freeze-dried containing icariin (ICA)-loaded chitosan microspheres. The shell scaffolds were electrospun using collagen, polycaprolactone and hydroxyapatite materials to form CPH composite scaffolds with the ones containing ICA microspheres named CPHI. The core-shell scaffolds were then cross-linked by genipin. The morphology, microstructure, physical and mechanical properties of the scaffolds were assessed. Rat marrow mesenchymal stem cells from the wistar rat were cultured with the scaffolds. The cell adhesion and proliferation were analysed. Adult rabbit models with tibial plateau defects were used to evaluate the performance of these scaffolds in repairing the bone defects over 4 to 12 weeks. Results The results reveal that the novel drug-loading core-shell scaffolds were successfully fabricated, which showed good physical and chemical properties and appropriate mechanical properties. Furthermore, excellent cells attachment was observed on the CPHI scaffolds. The results from radiography, micro-computed tomography, histological and immunohistochemical analysis demonstrated that abundant new bones were formed on the CPHI scaffolds. Conclusion These new core-shell composite scaffolds have great potential for bone tissue engineering applications and may lead to effective bone regeneration and repair.
Collapse
Affiliation(s)
- Hongbin Zhao
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Junjie Tang
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Dong Zhou
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Yiping Weng
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Wen Qin
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Chun Liu
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Songwei Lv
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China
| | - Wei Wang
- Medical School, Hexi University, Zhangye 730041, People's Republic of China
| | - Xiubo Zhao
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China.,Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
202
|
Salih M, Shaharuddin B, Abdelrazeg S. A Concise Review on Mesenchymal Stem Cells for Tissue Engineering with a Perspective on Ocular Surface Regeneration. Curr Stem Cell Res Ther 2020; 15:211-218. [DOI: 10.2174/1574888x15666200129145251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/27/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
Abstract
Organ and tissue transplantation are limited by the scarcity of donated organs or tissue
sources. The success of transplantation is limited by the risk of disease transmission and immunological-
related rejection. There is a need for new strategies and innovative solutions to make transplantation
readily available, safer and with less complications to increase the success rates. Accelerating progress
in stem cell biology and biomaterials development have pushed tissue and organ engineering to a
higher level. Among stem cells repertoire, Mesenchymal Stem Cells (MSC) are gaining interest and
recognized as a cell population of choice. There is accumulating evidence that MSC growth factors, its
soluble and insoluble proteins are involved in several key signaling pathways to promote tissue development,
cellular differentiation and regeneration. MSC as multipotent non-hematopoietic cells with
paracrine factors is advantageous for regenerative therapies. In this review, we discussed and summarized
the important features of MSC including its immunomodulatory properties, mechanism of homing
in the direction of tissue injury, licensing of MSC and the role of MSC soluble factors in cell-free
therapy. Special consideration is highlighted on the rapidly growing research interest on the roles of
MSC in ocular surface regeneration.
Collapse
Affiliation(s)
- Mohamed Salih
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Bakiah Shaharuddin
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Samar Abdelrazeg
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
203
|
Babaei A, Bannazadeh Baghi H, Nezhadi A, Jamalpoor Z. In Vitro Anti-cancer Activity of Adipose-Derived Mesenchymal Stem Cells Increased after Infection with Oncolytic Reovirus. Adv Pharm Bull 2020; 11:361-370. [PMID: 33880359 PMCID: PMC8046384 DOI: 10.34172/apb.2021.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose: Reovirus type 3 Dearing (ReoT3D), a wild type oncolytic virus (OV) from the Reoviridae family, kills KRAS mutant cancer cells. However, the use of OVs has faced with some limitations such as immune responses, and delivery of OVs to the tumor sites in systemic therapy. To solve this, and also to increase the anti-cancer effects of these OVs, mesenchymal stem cells (MSCs) might be used as an effective vehicle for OVs delivery. In this study, we examined the anti-cancer effects of human adipose derived-MSCs (AD-MSCs) as a vehicle of ReoT3D against human glioblastoma cells. Methods: Here, AD-MSCs were characterized and toxicity of ReoT3D on them was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Then, capability of AD-MSCs for virus production was assessed by real-time polymerase chain reaction (PCR), and different in vitro anti-cancer experiments were applied for our anti-cancer purposes. Results: Our results from toxicity assay revealed that the isolated and provoked AD-MSCs were resistant to nontoxic concentration multiplicity of infection (MOI) >1 pfu/cells of ReoT3D. In addition, the results indicated that AD-MSCs were susceptible for virus life cycle complementation and were capable for production of virus progenies. Furthermore, our results showed that AD-MSCs had oncolysis effects and increased the anti-cancer effects of ReoT3D. Conclusion: AD-MSCs as a susceptible host for oncolytic reovirus could increase the anti-cancer activity of this OV against glioblastoma multiforme (GBM) cell line.
Collapse
Affiliation(s)
- Abouzar Babaei
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akram Nezhadi
- Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran
| |
Collapse
|
204
|
Safarova Y, Umbayev B, Hortelano G, Askarova S. Mesenchymal stem cells modifications for enhanced bone targeting and bone regeneration. Regen Med 2020; 15:1579-1594. [PMID: 32297546 DOI: 10.2217/rme-2019-0081] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In pathological bone conditions (e.g., osteoporotic fractures or critical size bone defects), increasing the pool of osteoblast progenitor cells is a promising therapeutic approach to facilitate bone healing. Since mesenchymal stem cells (MSCs) give rise to the osteogenic lineage, a number of clinical trials investigated the potential of MSCs transplantation for bone regeneration. However, the engraftment of transplanted cells is often hindered by insufficient oxygen and nutrients supply and the tendency of MSCs to home to different sites of the body. In this review, we discuss various approaches of MSCs transplantation for bone regeneration including scaffold and hydrogel constructs, genetic modifications and surface engineering of the cell membrane aimed to improve homing and increase cell viability, proliferation and differentiation.
Collapse
Affiliation(s)
- Yuliya Safarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,School of Engineering & Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bauyrzhan Umbayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Gonzalo Hortelano
- School of Sciences & Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
205
|
Jose J, George T, Thomas AM. Regulation of Stem Cell-Based Research in India in Comparison with the US, EU and other Asian Countries: Current Issues and Future Perspectives. Curr Stem Cell Res Ther 2020; 15:492-508. [PMID: 32250233 DOI: 10.2174/1574888x15666200402134750] [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: 10/11/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 11/22/2022]
Abstract
Stem cell therapy is applicable for repair and replacement of damaged cells and tissues. Apart from transplanting cells to the body, the stem cell therapy directs them to grow new and healthy tissues. Stem cells in the area of regenerative medicines hold tremendous promise that may help to regenerate the damaged tissues and heal various diseases like multiple sclerosis, heart diseases, Parkinson's disease, and so on. To prove the safety, efficacy, and for the requirement of a licence for manufacturing and sale, all the stem cell therapies should pass the required criteria and undergo certain examinations of the regulatory agencies. The regulatory authorities review the manufacturing procedures of products to assure its purity and potency. This review summarizes the comparative critical evaluations of existing regulations and developments on the stem cells research in India, USA, EU and Asian regions and also discusses the challenges that have to be overcome and the important points that should be understood to position India as a source of the perspective nation in stem cells around the world.
Collapse
Affiliation(s)
- Jobin Jose
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| | - Teena George
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| | - Aaron M Thomas
- Department of Pharmaceutical Regulatory Affairs and Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, NITTE Deemed to be University, Paneer, Mangalore 575018, Karnataka, India
| |
Collapse
|
206
|
Sigmarsdóttir Þ, McGarrity S, Rolfsson Ó, Yurkovich JT, Sigurjónsson ÓE. Current Status and Future Prospects of Genome-Scale Metabolic Modeling to Optimize the Use of Mesenchymal Stem Cells in Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:239. [PMID: 32296688 PMCID: PMC7136564 DOI: 10.3389/fbioe.2020.00239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells are a promising source for externally grown tissue replacements and patient-specific immunomodulatory treatments. This promise has not yet been fulfilled in part due to production scaling issues and the need to maintain the correct phenotype after re-implantation. One aspect of extracorporeal growth that may be manipulated to optimize cell growth and differentiation is metabolism. The metabolism of MSCs changes during and in response to differentiation and immunomodulatory changes. MSC metabolism may be linked to functional differences but how this occurs and influences MSC function remains unclear. Understanding how MSC metabolism relates to cell function is however important as metabolite availability and environmental circumstances in the body may affect the success of implantation. Genome-scale constraint based metabolic modeling can be used as a tool to fill gaps in knowledge of MSC metabolism, acting as a framework to integrate and understand various data types (e.g., genomic, transcriptomic and metabolomic). These approaches have long been used to optimize the growth and productivity of bacterial production systems and are being increasingly used to provide insights into human health research. Production of tissue for implantation using MSCs requires both optimized production of cell mass and the understanding of the patient and phenotype specific metabolic situation. This review considers the current knowledge of MSC metabolism and how it may be optimized along with the current and future uses of genome scale constraint based metabolic modeling to further this aim.
Collapse
Affiliation(s)
- Þóra Sigmarsdóttir
- The Blood Bank, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | - Sarah McGarrity
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Óttar Rolfsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Ólafur E. Sigurjónsson
- The Blood Bank, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|
207
|
Thyparambil NJ, Gutgesell LC, Bromet BA, Flowers LE, Greaney S, Day DE, Semon JA. Bioactive borate glass triggers phenotypic changes in adipose stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:35. [PMID: 32206916 DOI: 10.1007/s10856-020-06366-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 02/17/2020] [Indexed: 06/10/2023]
Abstract
A bioactive borate glass, 13-93B3 (B3), has been used successfully in the clinic to treat chronic, nonhealing wounds without scarring. However, the mechanism by which B3 stimulates wound healing is poorly understood. Because adipose stem cells (ASCs) have been shown to have multiple roles in wound repair, we hypothesized that B3 triggers ASCs. In this study, we evaluate the effects of B3 on ASC survival, migration, differentiation, and protein secretion in vitro. In concentrations ≤10 mg/ml, B3 did not affect ASC viability under static conditions. B3 promoted the migration of ASCs but did not increase differentiation into bone or fat. B3 also decreased ASCs secretion of collagen I, PAI-1, MCP-1, DR6, DKK-1, angiogenin, IL-1, IGFBP-6, VEGF, and TIMP-2; increased expression of IL-1R and E-selectin; had a transient decrease in IL-6 secretion; and had a transient increase in bFGF secretion. Together, these results show that B3 alters the protein secretion of ASCs.
Collapse
Affiliation(s)
- Nathan J Thyparambil
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lisa C Gutgesell
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lauren E Flowers
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Samantha Greaney
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Delbert E Day
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA.
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA.
| |
Collapse
|
208
|
Abdal Dayem A, Kim K, Lee SB, Kim A, Cho SG. Application of Adult and Pluripotent Stem Cells in Interstitial Cystitis/Bladder Pain Syndrome Therapy: Methods and Perspectives. J Clin Med 2020; 9:jcm9030766. [PMID: 32178321 PMCID: PMC7141265 DOI: 10.3390/jcm9030766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is a multifactorial, chronic disease without definite etiology characterized by bladder-related pelvic pain. IC/BPS is associated with pain that negatively affects the quality of life. There are various therapeutic approaches against IC/BPS. However, no efficient therapeutic agent against IC/BPS has been discovered yet. Urothelium dysfunction is one of the key factors of IC/BPS-related pathogenicity. Stem cells, including adult stem cells (ASCs) and pluripotent stem cells (PSCs), such as embryonic stem cells (ESCs) and induced PSCs (iPSCs), possess the abilities of self-renewal, proliferation, and differentiation into various cell types, including urothelial and other bladder cells. Therefore, stem cells are considered robust candidates for bladder regeneration. This review provides a brief overview of the etiology, pathophysiology, diagnosis, and treatment of IC/BPS as well as a summary of ASCs and PSCs. The potential of ASCs and PSCs in bladder regeneration via differentiation into bladder cells or direct transplantation into the bladder and the possible applications in IC/BPS therapy are described in detail. A better understanding of current studies on stem cells and bladder regeneration will allow further improvement in the approaches of stem cell applications for highly efficient IC/BPS therapy.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Korea
- Correspondence: (A.K.); (S.-G.C.); Tel.: +82-2-2030-7675 (A.K.); +82-2-450-4207 (S.-G.C.); Fax: +82-2-2030-7748 (A.K.); +82-2-450-4207 (S.-G.C.)
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
- Correspondence: (A.K.); (S.-G.C.); Tel.: +82-2-2030-7675 (A.K.); +82-2-450-4207 (S.-G.C.); Fax: +82-2-2030-7748 (A.K.); +82-2-450-4207 (S.-G.C.)
| |
Collapse
|
209
|
lncRNA-TINCR Functions as a Competitive Endogenous RNA to Regulate the Migration of Mesenchymal Stem Cells by Sponging miR-761. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9578730. [PMID: 32185226 PMCID: PMC7061138 DOI: 10.1155/2020/9578730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/17/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Mounting evidences have indicated that terminal differentiation-induced lncRNA (TINCR) contributes to various cellular processes, such as proliferation, apoptosis, autophagy, migration, invasion, and metastasis. However, the function of TINCR in regulating migration of MSCs is largely unknown. In this study, the effects of TINCR on the migration of rat MSCs from the bone marrow were studied by Transwell assays and wound healing assays. Our results suggested that TINCR positively regulated migration of rMSCs. miR-761 mimics suppressed rMSC migration, whereas miR-761 inhibitor promoted migration. Target prediction analysis tools and dual-luciferase reporter gene assay identified Wnt2 as a direct target of miR-761. miR-761 could inhibit the expression of Wnt2. Further, the investigation about the function of TINCR in miR-761-induced migration of rMSCs was completed. These results demonstrated that TINCR took part in the regulation of miR-761-induced migration in rMSCs through the regulation of Wnt2 and its Wnt2 signaling pathway. Taken together, our results demonstrate that lncRNA-TINCR functions as a competitive endogenous RNA (ceRNA) to regulate the migration of rMSCs by sponging miR-761 which modulates the role of Wnt2. These findings provide evidence that lncRNA-TINCR has a chance to serve as a potential target for enhancing MSC homing through the miR-761/Wnt2 signaling pathway.
Collapse
|
210
|
Shammaa R, El-Kadiry AEH, Abusarah J, Rafei M. Mesenchymal Stem Cells Beyond Regenerative Medicine. Front Cell Dev Biol 2020; 8:72. [PMID: 32133358 PMCID: PMC7040370 DOI: 10.3389/fcell.2020.00072] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are competent suitors of cellular therapy due to their therapeutic impact on tissue degeneration and immune-based pathologies. Additionally, their homing and immunomodulatory properties can be exploited in cancer malignancies to transport pharmacological entities, produce anti-neoplastic agents, or induce anti-tumor immunity. Herein, we create a portfolio for MSC properties, showcasing their distinct multiple therapeutic utilities and successes/challenges thereof in both animal studies and clinical trials. We further highlight the promising potential of MSCs not only in cancer management but also in instigating tumor-specific immunity - i.e., cancer vaccination. Finally, we reflect on the possible reasons impeding the clinical advancement of MSC-based cancer vaccines to assist in contriving novel methodologies from which a therapeutic milestone might emanate.
Collapse
Affiliation(s)
- Riam Shammaa
- Canadian Centre for Regenerative Therapy, Toronto, ON, Canada.,IntelliStem Technologies Inc., Toronto, ON, Canada.,Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Abed El-Hakim El-Kadiry
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Jamilah Abusarah
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada.,Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
211
|
Eljarrah A, Gergues M, Pobiarzyn PW, Sandiford OA, Rameshwar P. Therapeutic Potential of Mesenchymal Stem Cells in Immune-Mediated Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:93-108. [PMID: 31898783 DOI: 10.1007/978-3-030-31206-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can self-renew and differentiate into cells of all germ layers. MSCs can be easily attracted to the site of tissue insult with high levels of inflammatory mediators. The general ability of MSCs to migrate at the sites of tissue injury suggested an innate ability for these cells to be involved in baseline tissue repair. The bone marrow is one of the primary sources of MSCs, though they can be ubiquitous. An attractive property of MSCs for clinical application is their ability to cross allogeneic barrier. However, alone, MSCs are not immune suppressive cells. Rather, they can be licensed by the tissue microenvironment to become immune suppressor cells. Immune suppressor functions of MSCs include those that blunt cytotoxicity of natural killer cells, suppression of T-cell proliferation, and "veto" function. MSCs, as third-party cells, suppress the immune response that generally recapitulates graft-versus-host disease (GvHD) responses. Based on the plastic functions of MSCs, these cells have dominated the field of cell-based therapies, such as anti-inflammatory and drug delivery. Here, we focus on the potential use of MSC for immunological disorders such as Crohn's disease and GvHD.
Collapse
Affiliation(s)
- Adam Eljarrah
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Marina Gergues
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Piotr W Pobiarzyn
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Oleta A Sandiford
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ, USA.
| |
Collapse
|
212
|
Cao W, Liu B, Xia F, Duan M, Hong Y, Niu J, Wang L, Liu Y, Li C, Cui D. MnO 2@Ce6-loaded mesenchymal stem cells as an "oxygen-laden guided-missile" for the enhanced photodynamic therapy on lung cancer. NANOSCALE 2020; 12:3090-3102. [PMID: 31965129 DOI: 10.1039/c9nr07947e] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The critical issue in nanoscale medicine delivery systems is the targeted efficiency to guarantee the maximum accumulation of nanodrugs in tumors to exert better therapeutic action. In this study, we adopted an active and potent strategy based on mesenchymal stem cells (MSCs) certified with excellent tumor-tropism ability to load and ship MnO2@Ce6 nanoparticles into a tumor site. Notably, under the premise of the negligible cellular toxicity of MnO2@Ce6 on MSCs, its considerable uptake by MSCs enabled this nanoplatform (MnO2@Ce6-MSCs) to distribute increasingly inside the tumor. Briefly, a Ce6 photosensitizer was bound to MnO2 nanospheres by physical adsorption, improving its own stability in blood circulation. Furthermore, the delivered MnO2@Ce6 could modulate the tumor microenvironment (TME) by high sensitivity to excess hydrogen protons (H+) and H2O2. Thus, O2 generated by these reactions served as an abundant source for 1O2 conversion under a 633 nm laser exposure, which overcame the crucial bottleneck of the unfavorable hypoxia condition in TME for photodynamic therapy (PDT). In addition, MnO2 decomposed into Mn2+, which was represented by high T1 relaxivity in magnetic resonance imaging (MRI). The Mn2+ was finally removed rapidly from the body by liver metabolism and kidney filtration. These results endowed the original nanoplatform with striking potential for MSC-guided, Ce6-converted, MRI-monitored PDT for further innovation of a clinical cancer diagnosis-treatment agent.
Collapse
Affiliation(s)
- Wen Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Jimenez-Puerta GJ, Marchal JA, López-Ruiz E, Gálvez-Martín P. Role of Mesenchymal Stromal Cells as Therapeutic Agents: Potential Mechanisms of Action and Implications in Their Clinical Use. J Clin Med 2020; 9:jcm9020445. [PMID: 32041213 PMCID: PMC7074225 DOI: 10.3390/jcm9020445] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
Due to the great therapeutic interest that involves the translation of mesenchymal stromal cells (MSCs) into clinical practice, they have been widely studied as innovative drugs, in order to treat multiple pathologies. MSC-based cell therapy involves the administration of MSCs either locally or systemically into the receptor body where they can traffic and migrate towards the affected tissue and participate in the process of healing. The therapeutic effects of MSCs compromise of different mechanisms such as the functional integration of differentiated MSCs into diseased host tissue after transplantation, their paracrine support, and their impact on the regulation of both the innate and the acquired immune system. Here, we establish and provide recent advances about the principal mechanisms of action through which MSCs can perform their activity and effect as a therapeutic tool. The purpose of this review is to examine and discuss the MSCs capacity of migration, their paracrine effect, as well as MSC-mediated modifications on immune cell responses.
Collapse
Affiliation(s)
- Gonzalo José Jimenez-Puerta
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, 18016 Granada, Spain; (G.J.J.-P.); (J.A.M.)
| | - Juan Antonio Marchal
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, 18016 Granada, Spain; (G.J.J.-P.); (J.A.M.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Elena López-Ruiz
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, 18016 Granada, Spain; (G.J.J.-P.); (J.A.M.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Health Sciences, University of Jaén, 23071 Jaén, Spain
- Correspondence: (E.L.-R.); or (P.G.-M.)
| | - Patricia Gálvez-Martín
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 18016 Granada, Spain
- R&D Human Health, Bioibérica S.A.U., 08029 Barcelona, Spain
- Correspondence: (E.L.-R.); or (P.G.-M.)
| |
Collapse
|
214
|
Catharmus tinctorius volatile oil promote the migration of mesenchymal stem cells via ROCK2/Myosin light chain signaling. Chin J Nat Med 2020; 17:506-516. [PMID: 31514982 DOI: 10.1016/s1875-5364(19)30072-x] [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: 05/25/2019] [Indexed: 12/24/2022]
Abstract
MSC transplantation has been explored as a new clinical approach to stem cell-based therapies for bone diseases in regenerative medicine due to their osteogenic capability. However, only a small population of implanted MSC could successfully reach the injured areas. Therefore, enhancing MSC migration could be a beneficial strategy to improve the therapeutic potential of cell transplantation. Catharmus tinctorius volatile oil (CTVO) was found to facilitate MSC migration. Further exploration of the underlying molecular mechanism participating in the pro-migratory ability may provide a novel strategy to improve MSC transplantation efficacy. This study indicated that CTVO promotes MSC migration through enhancing ROCK2 mRNA and protein expressions. MSC migration induced by CTVO was blunted by ROCK2 inhibitor, which also decreased myosin light chain (MLC) phosphorylation. Meanwhile, the siRNA for ROCK2 inhibited the effect of CTVO on MSC migration ability and attenuated MLC phosphorylation, suggesting that CTVO may promote BMSC migration via the ROCK2/MLC signaling. Taken together, this study indicates that C. tinctorius volatile oil could enhance MSC migration via ROCK2/MLC signaling in vitro. C. tinctorius volatile oil-targeted therapy could be a beneficial strategy to improve the therapeutic potential of cell transplantation for bone diseases in regenerative medicine.
Collapse
|
215
|
Manferdini C, Paolella F, Gabusi E, Cattini L, Rojewski M, Schrezenmeier H, Addimanda O, Meliconi R, Lisignoli G. Osteoarthritic Milieu Affects Adipose-Derived Mesenchymal Stromal Cells. J Orthop Res 2020; 38:336-347. [PMID: 31424111 PMCID: PMC7003792 DOI: 10.1002/jor.24446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/07/2019] [Indexed: 02/04/2023]
Abstract
The objective of this study was to define the effects of osteoarthritic (OA) milieu on good manufactured practice-adipose-derived mesenchymal stromal cells (GMP-ASC) that are commonly utilized in cell therapies. Two different OA milieu: OA synovial fluid (SF) and OA-conditioned medium (CM) from synoviocytes were used to treat GMP-ASC both in normoxia or hypoxia. GMP-ASC were tested for cell migration, proliferation, cytokine receptors expression (CXCR1, CXCR2, CXCR3, CXCR4, CXCR7, CCR1, CCR2, CCR3, CCR5, IL6R), and cytokines (CXCL8/IL8, CXCL10/IP10, CXCL12/SDF-1, CCL2/MCP1, CCL3/MIP1α, CCL4/MIP1β, CCL5/RANTES, IL6) release. Healthy SF was used as controls. We demonstrated that GMP-ASC show an increase in proliferation, migration, and modulation of CXCR1, CXCR3, CCR1, and CCR5 receptors in hypoxic condition. Moreover, GMP-ASC migration increased 15-fold when treated either with OA-SF or OA-CM compared with healthy SF both in normoxia and hypoxia. GMP-ASC treated in both OA milieu showed an increase in CXCR3, CCR3, and IL6R and a decrease in CCR1 and CCR2 receptors. In OA-SF, we detected higher amount of CXCL10/IP10 than in OA-CM, while CCL2/MCP1 and CCL4/MIP1β were higher in OA-CM compared with OA-SF. CXCL10/IP10 was the only chemokine of the OA milieu, which was down-modulated after treatment with GMP-ASC. In conclusion, we demonstrated specific effects of OA milieu on both GMP-ASC proliferation, migration, and cytokine receptor expression that were strictly dependent on the inflammatory and hypoxic environment. The use of characterized OA milieu is crucial to define the therapeutic effect of GMP-ASC and indicates that CXCL10/IP10-CXCR3 axis is partially involved in the GMP-ASC effect on synovial macrophages. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 38:336-347, 2020.
Collapse
Affiliation(s)
- Cristina Manferdini
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Francesca Paolella
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Elena Gabusi
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Luca Cattini
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Markus Rojewski
- Institut für TransfusionsmedizinUniversität UlmUlmGermany,Institut für Klinische Transfusionsmedizin und ImmungenetikDRK‐Blutspendedienst Baden‐Württemberg–Hessen & UniversitätsklinikumUlmGermany
| | - Hubert Schrezenmeier
- Institut für TransfusionsmedizinUniversität UlmUlmGermany,Institut für Klinische Transfusionsmedizin und ImmungenetikDRK‐Blutspendedienst Baden‐Württemberg–Hessen & UniversitätsklinikumUlmGermany
| | - Olga Addimanda
- IRCCS Istituto Ortopedico RizzoliSSD Medicina e ReumatologiaBolognaItaly
| | - Riccardo Meliconi
- IRCCS Istituto Ortopedico RizzoliSSD Medicina e ReumatologiaBolognaItaly,Dipartimento di Scienze Biomediche e neuromotorieUniversità degli studi di BolognaBolognaItaly
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| |
Collapse
|
216
|
Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med 2020; 18:42. [PMID: 32000804 PMCID: PMC6993499 DOI: 10.1186/s12967-020-02234-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have attracted intense interest due to their powerful intrinsic properties of self-regeneration, immunomodulation and multi-potency, as well as being readily available and easy to isolate and culture. Notwithstanding, MSC based therapy suffers reduced efficacy due to several challenges which include unfavorable microenvironmental factors in vitro and in vivo. Body In the quest to circumvent these challenges, several modification techniques have been applied to the naïve MSC to improve its inherent therapeutic properties. These modification approaches can be broadly divided into two groups to include genetic modification and preconditioning modification (using drugs, growth factors and other molecules). This field has witnessed great progress and continues to gather interest and novelty. We review these innovative approaches in not only maintaining, but also enhancing the inherent biological activities and therapeutics of MSCs with respect to migration, homing to target site, adhesion, survival and reduced premature senescence. We discuss the application of the improved modified MSC in some selected human diseases. Possible ways of yet better enhancing the therapeutic outcome and overcoming challenges of MSC modification in the future are also elaborated. Conclusion The importance of prosurvival and promigratory abilities of MSCs in their therapeutic applications can never be overemphasized. These abilities are maintained and even further enhanced via MSC modifications against the inhospitable microenvironment during culture and transplantation. This is a turning point in MSC-based therapy with promising preclinical studies and higher future prospect.
Collapse
|
217
|
Kamal MM, Kassem DH. Therapeutic Potential of Wharton's Jelly Mesenchymal Stem Cells for Diabetes: Achievements and Challenges. Front Cell Dev Biol 2020; 8:16. [PMID: 32064260 PMCID: PMC7000356 DOI: 10.3389/fcell.2020.00016] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is an alarming metabolic disease in which insulin secreting β-cells are damaged to various extent. Unfortunately, although currently available treatments help to manage the disease, however, patients usually develop complications, as well as decreased life quality and increased mortality. Thus, efficient therapeutic interventions to treat diabetes are urgently warranted. During the past years, mesenchymal stem cells (MSCs) have made their mark as a potential weapon in various regenerative medicine applications. The main fascination about MSCs lies in their potential to exert reparative effects on an amazingly wide spectrum of tissue injury. This is further reinforced by their ease of isolation and large ex vivo expansion capacity, as well as demonstrated multipotency and immunomodulatory activities. Among all the sources of MSCs, those isolated from umbilical cord-Wharton's jelly (WJ-MSCs), have been proved to provide a great source of MSCs. WJ-MSCs do not impose any ethical concerns as those which exist regarding ESCs, and represent a readily available non-invasive source, and hence suggested to become the new gold standard for MSC-based therapies. In the current review, we shall overview achievements, as well as challenges/hurdles which are standing in the way to utilize WJ-MSCs as a novel efficient therapeutic modality for DM.
Collapse
Affiliation(s)
- Mohamed M. Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- The Center for Drug Research and Development, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dina H. Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
218
|
Mousawi F, Peng H, Li J, Ponnambalam S, Roger S, Zhao H, Yang X, Jiang LH. Chemical activation of the Piezo1 channel drives mesenchymal stem cell migration via inducing ATP release and activation of P2 receptor purinergic signaling. Stem Cells 2020; 38:410-421. [PMID: 31746084 PMCID: PMC7064961 DOI: 10.1002/stem.3114] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 08/02/2019] [Accepted: 09/01/2019] [Indexed: 12/12/2022]
Abstract
In this study, we examined the Ca2+‐permeable Piezo1 channel, a newly identified mechanosensing ion channel, in human dental pulp‐derived mesenchymal stem cells (MSCs) and hypothesized that activation of the Piezo1 channel regulates MSC migration via inducing ATP release and activation of the P2 receptor purinergic signaling. The Piezo1 mRNA and protein were readily detected in hDP‐MSCs from multiple donors and, consistently, brief exposure to Yoda1, the Piezo1 channel‐specific activator, elevated intracellular Ca2+ concentration. Yoda1‐induced Ca2+ response was inhibited by ruthenium red or GsMTx4, two Piezo1 channel inhibitors, and also by Piezo1‐specific siRNA. Brief exposure to Yoda1 also induced ATP release. Persistent exposure to Yoda1 stimulated MSC migration, which was suppressed by Piezo1‐specific siRNA, and also prevented by apyrase, an ATP scavenger, or PPADS, a P2 generic antagonist. Furthermore, stimulation of MSC migration induced by Yoda1 as well as ATP was suppressed by PF431396, a PYK2 kinase inhibitor, or U0126, an inhibitor of the mitogen‐activated protein kinase MEK/ERK signaling pathway. Collectively, these results suggest that activation of the Piezo1 channel stimulates MSC migration via inducing ATP release and subsequent activation of the P2 receptor purinergic signaling and downstream PYK2 and MEK/ERK signaling pathways, thus revealing novel insights into the molecular and signaling mechanisms regulating MSC migration. Such findings provide useful information for evolving a full understanding of MSC migration and homing and developing strategies to improve MSC‐based translational applications.
Collapse
Affiliation(s)
- Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Hongsen Peng
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Jing Li
- Lingnan Medical Research Centre, School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Sreenivasan Ponnambalam
- School of Molecular and Cell Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Hucheng Zhao
- Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Xuebin Yang
- Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| |
Collapse
|
219
|
Thyparambil NJ, Gutgesell LC, Hurley CC, Flowers LE, Day DE, Semon JA. Adult stem cell response to doped bioactive borate glass. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:13. [PMID: 31965357 DOI: 10.1007/s10856-019-6353-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/21/2019] [Indexed: 06/10/2023]
Abstract
Bioactive glasses have transformed healthcare due to their versatility. Bioactive borate glass, in particular, has shown remarkable healing properties for both hard and soft tissues. Incorporating dopants into the composition of bioactive glass helps to control mechanical properties, and it increases their usefulness for clinical applications. Using a bioactive borate glass, 13-93B3 (B3), we investigated eleven dopants on the viability and migration potential of adipose stem cells (ASCs), a therapeutic source of cells used in tissue engineering and cell therapy. Our results show that under standard cell culture conditions, only Cu-doped B3 decreased cell viability, while only Y-doped B3 attracted ASCs as it dissolved in cell culture media. Using a transwell invasion assay, priming ASCs with Co, Fe, Ga, I, Sr, or Zn-doped B3 increased their homing capacity. Because there is widespread interest in optimizing and enhancing the homing efficiency of ASCs and other therapeutic cells, we then tested if priming bone marrow mesenchymal stem cells (BMSCs) with dopants also increased their homing capacity. In the case of BMSCs, there was a significant increase in invasion when cells were primed with any of the doped-B3 glasses. This work shows that incorporating dopants into borate glasses can provide a platform for a safe and efficient method that stimulates endogenous cells and healing mechanisms.
Collapse
Affiliation(s)
- Nathan J Thyparambil
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lisa C Gutgesell
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Cassandra C Hurley
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lauren E Flowers
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Delbert E Day
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA.
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA.
| |
Collapse
|
220
|
Zhou L, Yau A, Yu H, Kuhn L, Guo W, Chen Y. Self-assembled biomimetic Nano-Matrix for stem cell anchorage. J Biomed Mater Res A 2020; 108:984-991. [PMID: 31904174 DOI: 10.1002/jbm.a.36875] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/27/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been widely applied in biomedicine due to their ability to differentiate into many different cell types and their ability to synthesize a broad spectrum of growth factors and cytokines that directly and indirectly influence other cells in their vicinity. To guide MSC infiltration to a bone fracture site, we developed a novel self-assembled Nano-Matrix which can be used as an injectable scaffold to repair bone fractures. The Nano-Matrix is formed by Janus base nanotubes (JBNTs) and fibronectin (FN). JBNTs are nucleobase-derived nanotubes mimicking collagen fibers, and FN is one of the cell adhesive glycoproteins which is responsible for cell-extracellular matrix interactions and guides stem cell migration and differentiation to desired cells types. Here, we demonstrated the successful fabrication and characterization of the JBNT/FN Nano-Matrix as well as its excellent bioactivity that encouraged human MSC migration and adhesion. This work lays a solid foundation for using the Nano-Matrix as an injectable approach to improve MSC retention and function during bone fracture healing.
Collapse
Affiliation(s)
- Libo Zhou
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Anne Yau
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut.,Brown University Medical School, Providence, Rhode Island
| | - Hongchuan Yu
- Brown University Medical School, Providence, Rhode Island
| | - Liisa Kuhn
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut.,Department of Biomedical Engineering, University of Connecticut, Farmington, Connecticut
| | - Wei Guo
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut.,Brown University Medical School, Providence, Rhode Island
| |
Collapse
|
221
|
Ali AAA, Shahror RA, Chen KY. Efficient Labeling Of Mesenchymal Stem Cells For High Sensitivity Long-Term MRI Monitoring In Live Mice Brains. Int J Nanomedicine 2020; 15:97-114. [PMID: 32021167 PMCID: PMC6955624 DOI: 10.2147/ijn.s211205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/08/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Regenerative medicine field is still lagging due to the lack of adequate knowledge regarding the homing of therapeutic cells towards disease sites, tracking of cells during treatment, and monitoring the biodistribution and fate of cells. Such necessities require labeling of cells with imaging agents that do not alter their biological characteristics, and development of suitable non-invasive imaging modalities. PURPOSE We aimed to develop, characterize, and standardize a facile labeling strategy for engineered mesenchymal stem cells without altering their viability, secretion of FGF21 protein (neuroprotective), and differentiation capabilities for non-invasive longitudinal MRI monitoring in live mice brains with high sensitivity. METHODS We compared the labeling efficiency of different commercial iron oxide nanoparticles towards our stem cells and determined the optimum labeling conditions using prussian blue staining, confocal microscopy, transmission electron microscopy, and flow cytometry. To investigate any change in biological characteristics of labeled cells, we tested their viability by WST-1 assay, expression of FGF21 by Western blot, and adipogenic and osteogenic differentiation capabilities. MRI contrast-enhancing properties of labeled cells were investigated in vitro using cell-agarose phantoms and in mice brains transplanted with the therapeutic stem cells. RESULTS We determined the nanoparticles that showed best labeling efficiency and least extracellular aggregation. We further optimized their labeling conditions (nanoparticles concentration and media supplementation) to achieve high cellular uptake and minimal extracellular aggregation of nanoparticles. Cell viability, expression of FGF21 protein, and differentiation capabilities were not impeded by nanoparticles labeling. Low number of labeled cells produced strong MRI signal decay in phantoms and in live mice brains which were visible for 4 weeks post transplantation. CONCLUSION We established a standardized magnetic nanoparticle labeling platform for stem cells that were monitored longitudinally with high sensitivity in mice brains using MRI for regenerative medicine applications.
Collapse
Affiliation(s)
- Ahmed Atef Ahmed Ali
- TMU Neuroscience Research Center – NeuroImage, College of Medicine, Taipei Medical University, Taipei110, Taiwan,Correspondence: Ahmed Atef Ahmed Ali Taipei Medical University, No. 250, Wuxing Street, Xinyi District, Taipei110, Taiwan ROCTel +886-2-2736-1661 ext 3215 Email
| | - Rami Ahmad Shahror
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei110, Taiwan,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei110, Taiwan
| |
Collapse
|
222
|
Rong X, Zhang G, Yang Y, Gao C, Chu W, Sun H, Wang Y, Li C. Transplanted Antler Stem Cells Stimulated Regenerative Healing of Radiation-induced Cutaneous Wounds in Rats. Cell Transplant 2020; 29:963689720951549. [PMID: 32907381 PMCID: PMC7784515 DOI: 10.1177/0963689720951549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/18/2020] [Accepted: 07/29/2020] [Indexed: 12/29/2022] Open
Abstract
Radiation-induced cutaneous injury is the main side effect of radiotherapy. The injury is difficult to cure and the pathogenesis is complex. Mesenchymal stem cells (MSCs) serve as a promising candidate for cell-based therapy for the treatment of cutaneous wounds. The aim of the present study was to investigate whether antler stem cells (AnSCs) have better therapeutic effects on radiation-induced cutaneous injury than currently available ones. In this study, a rat model of cutaneous wound injury from Sr-90 radiation was used. AnSCs (1 × 106/500 μl) were injected through the tail vein on the first day of irradiation. Our results showed that compared to the control group, AnSC-treated rats exhibited a delayed onset (14 days versus 7 days), shorter recovery time (51 days versus 84 days), faster healing rate (100% versus 70% on day 71), and higher healing quality with more cutaneous appendages regenerated (21:10:7/per given area compared to those of rat and human MSCs, respectively). More importantly, AnSCs promoted much higher quality of healing compared to other types of stem cells, with negligible scar formation. AnSC lineage tracing results showed that the injected-dye-stained AnSCs were substantially engrafted in the wound healing tissue, indicating that the therapeutic effects of AnSCs on wound healing at least partially through direct participation in the wound healing. Expression profiling of the wound-healing-related genes in the healing tissue of AnSC group more resembled a fetal wound healing. Revealing the mechanism underlying this higher quality of wound healing by using AnSC treatment would help to devise more effective cell-based therapeutics for radiation-induced wound healing in clinics.
Collapse
Affiliation(s)
- Xiaoli Rong
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Yanyan Yang
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Chenmao Gao
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Wenhui Chu
- School of Life Science, Taizhou University, Taizhou, China
| | - Hongmei Sun
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, China
| | - Yimin Wang
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| |
Collapse
|
223
|
Han J, Liu Y, Liu H, Li Y. Genetically modified mesenchymal stem cell therapy for acute respiratory distress syndrome. Stem Cell Res Ther 2019; 10:386. [PMID: 31843004 PMCID: PMC6915956 DOI: 10.1186/s13287-019-1518-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating hypoxemic respiratory failure, characterized by disruption of the alveolar-capillary membrane barrier. Current management for ARDS remains supportive, including lung-protective ventilation and a conservative fluid strategy. Mesenchymal stem cells (MSCs) have emerged as a potentially attractive candidate for the management of ARDS through facilitating lung tissue regeneration and repair by releasing paracrine soluble factors. Over the last decade, a variety of strategies have emerged to optimize MSC-based therapy. Among these, the strategy using genetically modified MSCs has received increased attention recently due to its distinct advantage, in conferring incremental migratory capacity and, enhancing the anti-inflammatory, immunomodulatory, angiogenic, and antifibrotic effects of these cells in numerous preclinical ARDS models, which may in turn provide additional benefits in the management of ARDS. Here, we provide an overview of recent studies testing the efficacy of genetically modified MSCs using preclinical models of ARDS.
Collapse
Affiliation(s)
- Jibin Han
- Department of Critical Care Medicine, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China
| | - Yuxiang Liu
- Shanxi Medical University, No.56, Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Hong Liu
- Department of Critical Care Medicine, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China.
| | - Yuanyuan Li
- Department of Critical Care Medicine, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
224
|
Zhou L, Liu S, Wang Z, Yao J, Cao W, Chen S, Xie W, Feng S, Xu Y, Cheng T, Han M, Feng S. Bone Marrow-Derived Mesenchymal Stem Cells Modified with Akt1 Ameliorates Acute Liver GVHD. Biol Proced Online 2019; 21:24. [PMID: 31889917 PMCID: PMC6913026 DOI: 10.1186/s12575-019-0112-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
Background Liver injury associated with acute graft-versus-host disease (aGVHD) is a frequent and severe complication of hematopoietic stem cell transplantation and remains a major cause of transplant-related mortality. Bone marrow-derived mesenchymal stem cells (BM-MSCs) has been proposed as a potential therapeutic approach for aGVHD. However, the therapeutic effects are not always achieved. In this study, we genetically engineered C57BL/6 mouse BM-MSCs with AKT1 gene and tested whether AKT1-MSCs was superior to control MSCs (Null-MSCs) for cell therapy of liver aGVHD. Results In vitro apoptosis analyses showed that, under both routine culture condition and high concentration interferon-γ (IFN-γ) (100ng/mL) stimulation condition, AKT1-MSCs had a survival (anti-apoptotic) advantage compared to Null-MSCs. In vivo imaging showed that AKT1-MSCs had better homing capacity and longer persistence in injured liver compared to Null-MSCs. Most importantly, AKT1-MSCs demonstrated an enhanced immunomodulatory function by releasing more immunosuppressive cytokines, such as IL-10. Adoptive transfer of AKT1-MSCs mitigated the histopathological abnormalities of concanavalin A(ConA)-induced liver injury along with significantly lowered serum levels of ALT and AST. The attenuation of liver injury correlated with the decrease of TNF-α and IFN-γ both in liver tissue and in the serum. Conclusions In summary, BM-MSCs genetically modified with AKT1 has a survival advantage and an enhanced immunomodulatory function both in vitro and in vivo and thus demonstrates the therapeutic potential for prevention and amelioration of liver GVHD and other immunity-associated liver injuries.
Collapse
Affiliation(s)
- Lukun Zhou
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Shuang Liu
- 2Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, Guangdong Province China
| | - Zhao Wang
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Jianfeng Yao
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Wenbin Cao
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Shulian Chen
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Wenjun Xie
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Shuqing Feng
- 3North China University of Science and Technology Affiliated Hospital, 73, Construction South Road, Lubei District, Tangshan, Hebei Province China
| | - Yuanfu Xu
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Tao Cheng
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Mingzhe Han
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| | - Sizhou Feng
- 1State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, China
| |
Collapse
|
225
|
Rezaei N, Sardarzadeh T, Sisakhtnezhad S. Thymoquinone promotes mouse mesenchymal stem cells migration in vitro and induces their immunogenicity in vivo. Toxicol Appl Pharmacol 2019; 387:114851. [PMID: 31812774 DOI: 10.1016/j.taap.2019.114851] [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] [Received: 06/21/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells (MSCs) have unique potentials, including migration and immunomodulation. Identification of the factors that enhance these activities can improve clinical applications of MSCs. This study aimed to investigate total antioxidant capacity (TAC) and migration potential of mouse MSCs exposed to thymoquinone (TQ) in vitro, and to examine the effect of TQ-treated MSCs on the expression of mouse immune cell markers. The results of total antioxidant capacity and wound healing assays showed that TQ increased the rate of MSCs TAC and migration in a dose- and time-dependent manner. The maximum TAC and migration were detected at 600 and 250 ng/ml of TQ, respectively. Functionally, the real-time PCR data analysis indicated that TQ induced c-Met and Cxcr4 expression and therefore, there may be a correlation between upregulation of these genes and increased MSCs migration. TQ also enhanced the up and down regulating impact of MSCs on Rorγt and Plzf expression and the expression of Tcf4 in mouse immune cells, respectively. Overall, this study declares that TQ increases the TAC of MSCs. It also proposes that TQ may, through activation of c-MET and CXCR4 signalling pathways, promote MSCs migration. TQ may also augment MSCs immunogenicity through its influence on the expression of genes involved in commitment of mouse immune system cells in vivo.
Collapse
Affiliation(s)
- Niloufar Rezaei
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Tayebeh Sardarzadeh
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | | |
Collapse
|
226
|
Intra-Bone Marrow Administration of Mesenchymal Stem/Stromal Cells Is a Promising Approach for Treating Osteoporosis. Stem Cells Int 2019; 2019:4214281. [PMID: 31781240 PMCID: PMC6875206 DOI: 10.1155/2019/4214281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/13/2019] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known to be useful for treating local bone diseases. However, it is not known if MSCs are effective for treating systemic bone diseases, as the risk for mortality following intravenous MSC administration has hindered research progress. In this study, we compared the safety and efficacy of intra-bone marrow and intravenous administration of MSCs for the treatment of ovariectomy- (OVX-) induced osteoporosis. Cells capable of forming bone were isolated from the murine compact bones and expanded in culture. Relatively pure MSCs possessing increased potential for cell proliferation, osteogenic differentiation, and inhibition of osteoclastogenesis were obtained by magnetic-activated cell sorting with the anti-Sca-1 antibody. Sca-1-sorted MSCs were administered to OVX mice, which were sacrificed 1 month later. We observed that 22% of the mice died after intravenous administration, whereas none of the mice died after intra-bone marrow administration. With respect to efficacy, intravenous administration improved bone mineral density (BMD) by increasing bone mineral content without affecting bone thickness, whereas intra-bone marrow administration improved BMD by increasing both bone mineral content and bone thickness. These results indicate that intra-bone marrow administration of pure MSCs is a safer and more effective approach for treating osteoporosis.
Collapse
|
227
|
Sinjari B, Diomede F, Khavinson V, Mironova E, Linkova N, Trofimova S, Trubiani O, Caputi S. Short Peptides Protect Oral Stem Cells from Ageing. Stem Cell Rev Rep 2019; 16:159-166. [DOI: 10.1007/s12015-019-09921-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
228
|
García-Sánchez D, Fernández D, Rodríguez-Rey JC, Pérez-Campo FM. Enhancing survival, engraftment, and osteogenic potential of mesenchymal stem cells. World J Stem Cells 2019; 11:748-763. [PMID: 31692976 PMCID: PMC6828596 DOI: 10.4252/wjsc.v11.i10.748] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/15/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for bone regeneration therapies due to their plasticity and easiness of sourcing. MSC-based treatments are generally considered a safe procedure, however, the long-term results obtained up to now are far from satisfactory. The main causes of these therapeutic limitations are inefficient homing, engraftment, and osteogenic differentiation. Many studies have proposed modifications to improve MSC engraftment and osteogenic differentiation of the transplanted cells. Several strategies are aimed to improve cell resistance to the hostile microenvironment found in the recipient tissue and increase cell survival after transplantation. These strategies could range from a simple modification of the culture conditions, known as cell-preconditioning, to the genetic modification of the cells to avoid cellular senescence. Many efforts have also been done in order to enhance the osteogenic potential of the transplanted cells and induce bone formation, mainly by the use of bioactive or biomimetic scaffolds, although alternative approaches will also be discussed. This review aims to summarize several of the most recent approaches, providing an up-to-date view of the main developments in MSC-based regenerative techniques.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria, Cantabria 39011, Spain
| | - Darío Fernández
- Laboratorio de Biología Celular y Molecular, Facultad de Odontología, Universidad Nacional del Nordeste, Corrientes W3400, Argentina
| | - José C Rodríguez-Rey
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria, Cantabria 39011, Spain
| | - Flor M Pérez-Campo
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria, Cantabria 39011, Spain.
| |
Collapse
|
229
|
Ma H, Duan S, Yan F, Yang H, Cao Y, Ge L, Gao R. Enhancer of zeste homolog 2 enhances the migration and chemotaxis of dental mesenchymal stem cells. J Int Med Res 2019; 48:300060519882149. [PMID: 31642363 PMCID: PMC7262854 DOI: 10.1177/0300060519882149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To investigate the function of enhancer of zeste homolog 2 (EZH2) in the migration and chemotaxis of human dental tissue-derived mesenchymal stem cells. Methods The expression of EZH2, C-X-C motif chemokine ligand 11 (CXCL11), CXCL16, and CXCR1 in stem cells from the apical papilla (SCAPs) was determined by real-time reverse transcription PCR and western blotting. The effects of EZH2 on the homing of SCAPs and the effects of EZH2-overexpressing SCAP culture supernatant on periodontal ligament stem cells (PDLSCs) were tested by scratch migration assays and transwell chemotaxis assays. Results EZH2 overexpression significantly enhanced the migration and chemotaxis of SCAPs and upregulated the expression of CXCL11, CXCL16, and CXCR1 in SCAPs. EZH2 depletion had the opposite effect, impairing the migration and chemotaxis of SCAPs and downregulating the expression of CXCL11, CXCL16, and CXCR1. The culture supernatant of EZH2-overexpressing SCAPs advanced the migration and chemotaxis of PDLSCs. Conclusions EZH2 evidently promoted the migration and chemotaxis of SCAPs by upregulating the expression of CXCL11, CXCL16, and CXCR1. Moreover, EZH2-overexpressing SCAPs enhanced the homing, migration, and chemotaxis of PDLSCs via paracrine signaling.
Collapse
Affiliation(s)
- Huarui Ma
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shaoyu Duan
- Department of Stomatology, Beijing Electric Power Hospital, Beijing, China
| | - Fei Yan
- Xiangya Stomatology Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Haoqing Yang
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Yangyang Cao
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Lihua Ge
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Runtao Gao
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
230
|
Qiao Y, Xu Z, Yu Y, Hou S, Geng J, Xiao T, Liang Y, Dong Q, Mei Y, Wang B, Qiao H, Dai J, Suo G. Single cell derived spheres of umbilical cord mesenchymal stem cells enhance cell stemness properties, survival ability and therapeutic potential on liver failure. Biomaterials 2019; 227:119573. [PMID: 31670080 DOI: 10.1016/j.biomaterials.2019.119573] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023]
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) have shown great potentials in regenerative medicine for their extensive sources, multilineage differentiation potential, low immunogenicity and self-renewal ability. However, the clinical application of UCMSCs still confronts many challenges including the requirement of large quantity of cells, low survival ability in vivo and the loss of main original characteristics due to two-dimensional (2D) culture. The traditional three-dimensional (3D)-spheroid culture can mimic in vivo conditions, but still has limitations in clinical application due to large size of spheroid against direct injection and inner cell death. Based on self-renewal tenet, we produced single cell derived sphere (SCDS) of UCMSCs through combining single cell pattern on chip with 3D culture. Compared with the 2D and traditional 3D culture, SCDS culture has many advantages to meet clinical requirements, including small size, higher abilities of survival and migration, and stronger hypoxia resistance and stemness maintenance. Furthermore, SCDS culture promotes angiogenesis in UCMSCs-xenografts and displays greater therapeutic potential on acute liver failure (ALF) in vivo. Our results suggest that SCDS culture may serve as a simple and effective strategy for UCMSCs optimization to meet clinical demand.
Collapse
Affiliation(s)
- Yong Qiao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China
| | - Zhongjuan Xu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yanzhen Yu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shulan Hou
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; School of Pharmacy, Xi'an Jiaotong University, Shaanxi, 710061, China
| | - Junsa Geng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Tongqian Xiao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Liang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China; School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qun Dong
- Department of Pathology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu, 210046, China
| | - Yan Mei
- Greepharma Inc., 211100, Nanjing, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Hong Qiao
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jianwu Dai
- State Key Laboratory of Molecular, Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Guangli Suo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu, 215123, China.
| |
Collapse
|
231
|
Subcutaneous and Visceral Adipose-Derived Mesenchymal Stem Cells: Commonality and Diversity. Cells 2019; 8:cells8101288. [PMID: 31640218 PMCID: PMC6830091 DOI: 10.3390/cells8101288] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/18/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are considered to be a useful tool for regenerative medicine, owing to their capabilities in differentiation, self-renewal, and immunomodulation. These cells have become a focus in the clinical setting due to their abundance and easy isolation. However, ASCs from different depots are not well characterized. Here, we analyzed the functional similarities and differences of subcutaneous and visceral ASCs. Subcutaneous ASCs have an extraordinarily directed mode of motility and a highly dynamic focal adhesion turnover, even though they share similar surface markers, whereas visceral ASCs move in an undirected random pattern with more stable focal adhesions. Visceral ASCs have a higher potential to differentiate into adipogenic and osteogenic cells when compared to subcutaneous ASCs. In line with these observations, visceral ASCs demonstrate a more active sonic hedgehog pathway that is linked to a high expression of cilia/differentiation related genes. Moreover, visceral ASCs secrete higher levels of inflammatory cytokines interleukin-6, interleukin-8, and tumor necrosis factor α relative to subcutaneous ASCs. These findings highlight, that both ASC subpopulations share multiple cellular features, but significantly differ in their functions. The functional diversity of ASCs depends on their origin, cellular context and surrounding microenvironment within adipose tissues. The data provide important insight into the biology of ASCs, which might be useful in choosing the adequate ASC subpopulation for regenerative therapies.
Collapse
|
232
|
Kallmeyer K, André-Lévigne D, Baquié M, Krause KH, Pepper MS, Pittet-Cuénod B, Modarressi A. Fate of systemically and locally administered adipose-derived mesenchymal stromal cells and their effect on wound healing. Stem Cells Transl Med 2019; 9:131-144. [PMID: 31613054 PMCID: PMC6954716 DOI: 10.1002/sctm.19-0091] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
There is increasing interest in the use of adipose‐derived mesenchymal stromal cells (ASCs) for wound repair. As the fate of administered cells is still poorly defined, we aimed to establish the location, survival, and effect of ASCs when administered either systemically or locally during wound repair under physiological conditions. To determine the behavior of ASCs, a rat model with wounds on the dorsal aspect of the hind paws was used and two treatment modes were assessed: ASCs administered systemically into the tail vein or locally around the wound. ASCs were transduced to express both firefly luciferase (Fluc) and green fluorescent protein to enable tracking by bioluminescence imaging and immunohistological analysis. Systemically administered ASCs were detected in the lungs 3 hours after injection with a decrease in luminescent signal at 48 hours and signal disappearance from 72 hours. No ASCs were detected in the wound. Locally administered ASCs remained strongly detectable for 7 days at the injection site and became distributed within the wound bed as early as 24 hours post injection with a significant increase observed at 72 hours. Systemically administered ASCs were filtered out in the lungs, whereas ASCs administered locally remained and survived not only at the injection site but were also detected within the wound bed. Both treatments led to enhanced wound closure. It appears that systemically administered ASCs have the potential to enhance wound repair distally from their site of entrapment in the lungs whereas locally administered ASCs enhanced wound repair as they became redistributed within the wound bed.
Collapse
Affiliation(s)
- Karlien Kallmeyer
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland.,Institute for Cellular and Molecular Medicine (ICMM), Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, South Africa
| | - Dominik André-Lévigne
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| | | | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine (ICMM), Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, South Africa.,Department of Human Genetics and Development, University of Geneva, Geneva, Switzerland
| | - Brigitte Pittet-Cuénod
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| | - Ali Modarressi
- Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
| |
Collapse
|
233
|
Dorland YL, Cornelissen AS, Kuijk C, Tol S, Hoogenboezem M, van Buul JD, Nolte MA, Voermans C, Huveneers S. Nuclear shape, protrusive behaviour and in vivo retention of human bone marrow mesenchymal stromal cells is controlled by Lamin-A/C expression. Sci Rep 2019; 9:14401. [PMID: 31591420 PMCID: PMC6779744 DOI: 10.1038/s41598-019-50955-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Culture expanded mesenchymal stromal cells (MSCs) are being extensively studied for therapeutic applications, including treatment of graft-versus-host disease, osteogenesis imperfecta and for enhancing engraftment of hematopoietic stem cells after transplantation. Thus far, clinical trials have shown that the therapeutic efficiency of MSCs is variable, which may in part be due to inefficient cell migration. Here we demonstrate that human MSCs display remarkable low migratory behaviour compared to other mesodermal-derived primary human cell types. We reveal that specifically in MSCs the nucleus is irregularly shaped and nuclear lamina are prone to wrinkling. In addition, we show that expression of Lamin A/C is relatively high in MSCs. We further demonstrate that in vitro MSC migration through confined pores is limited by their nuclei, a property that might correlate to the therapeutic inefficiency of administered MSC in vivo. Silencing expression of Lamin A/C in MSCs improves nuclear envelope morphology, promotes the protrusive activity of MSCs through confined pores and enhances their retention in the lung after intravenous administration in vivo. Our findings suggest that the intrinsic nuclear lamina properties of MSCs underlie their limited capacity to migrate, and that strategies that target the nuclear lamina might alter MSC-based cellular therapies.
Collapse
Affiliation(s)
- Yvonne L Dorland
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne S Cornelissen
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlijn Kuijk
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon Tol
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogenboezem
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martijn A Nolte
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlijn Voermans
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
234
|
Udartseva OO, Zhidkova OV, Ezdakova MI, Ogneva IV, Andreeva ER, Buravkova LB, Gollnick SO. Low-dose photodynamic therapy promotes angiogenic potential and increases immunogenicity of human mesenchymal stromal cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 199:111596. [DOI: 10.1016/j.jphotobiol.2019.111596] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/23/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
|
235
|
Gaddam S, Periasamy R, Gangaraju R. Adult Stem Cell Therapeutics in Diabetic Retinopathy. Int J Mol Sci 2019; 20:ijms20194876. [PMID: 31575089 PMCID: PMC6801872 DOI: 10.3390/ijms20194876] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetic retinopathy (DR), a complication of diabetes, is one of the leading causes of blindness in working-age adults. The pathology of the disease prevents the endogenous stem cells from participating in the natural repair of the diseased retina. Current treatments, specifically stem cell therapeutics, have shown variable efficacy in preclinical models due to the multi-faceted nature of the disease. Among the various adult stem cells, mesenchymal stem cells, especially those derived from adipose tissue and bone marrow, have been explored as a possible treatment for DR. This review summarizes the current literature around the various adult stem cell treatments for the disease and outlines the benefits and limitations of the therapeutics that are being explored in the field. The paracrine nature of adipose stem cells, in particular, has been highlighted as a potential solution to the lack of a homing and conducive environment that poses a challenge to the implantation of exogenous stem cells in the target tissue. Various methods of mesenchymal stem cell priming to adapt to a hostile retinal microenvironment have been discussed. Current clinical trials and potential safety concerns have been examined, and the future directions of stem cell therapeutics in DR have also been contemplated.
Collapse
Affiliation(s)
- Sriprachodaya Gaddam
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN 38163, USA.
| | - Ramesh Periasamy
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN 38163, USA.
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN 38163, USA.
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN 38163, USA.
| |
Collapse
|
236
|
Coppin L, Sokal E, Stéphenne X. Thrombogenic Risk Induced by Intravascular Mesenchymal Stem Cell Therapy: Current Status and Future Perspectives. Cells 2019; 8:cells8101160. [PMID: 31569696 PMCID: PMC6829440 DOI: 10.3390/cells8101160] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are currently studied and used in numerous clinical trials. Nevertheless, some concerns have been raised regarding the safety of these infusions and the thrombogenic risk they induce. MSCs express procoagulant activity (PCA) linked to the expression of tissue factor (TF) that, when in contact with blood, initiates coagulation. Some even describe a dual activation of both the coagulation and the complement pathway, called Instant Blood-Mediated Inflammatory Reaction (IBMIR), explaining the disappointing results and low engraftment rates in clinical trials. However, nowadays, different approaches to modulate the PCA of MSCs and thus control the thrombogenic risk after cell infusion are being studied. This review summarizes both in vitro and in vivo studies on the PCA of MSC of various origins. It further emphasizes the crucial role of TF linked to the PCA of MSCs. Furthermore, optimization of MSC therapy protocols using different methods to control the PCA of MSCs are described.
Collapse
Affiliation(s)
- Louise Coppin
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Etienne Sokal
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Xavier Stéphenne
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
237
|
Guo S, Perets N, Betzer O, Ben-Shaul S, Sheinin A, Michaelevski I, Popovtzer R, Offen D, Levenberg S. Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury. ACS NANO 2019; 13:10015-10028. [PMID: 31454225 DOI: 10.1021/acsnano.9b01892] [Citation(s) in RCA: 290] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Individuals with spinal cord injury (SCI) usually suffer from permanent neurological deficits, while spontaneous recovery and therapeutic efficacy are limited. Here, we demonstrate that when given intranasally, exosomes derived from mesenchymal stem cells (MSC-Exo) could pass the blood brain barrier and migrate to the injured spinal cord area. Furthermore, MSC-Exo loaded with phosphatase and tensin homolog small interfering RNA (ExoPTEN) could attenuate the expression of PTEN in the injured spinal cord region following intranasal administrations. In addition, the loaded MSC-Exo considerably enhanced axonal growth and neovascularization, while reducing microgliosis and astrogliosis. The intranasal ExoPTEN therapy could also partly improve structural and electrophysiological function and, most importantly, significantly elicited functional recovery in rats with complete SCI. The results imply that intranasal ExoPTEN may be used clinically to promote recovery for SCI individuals.
Collapse
Affiliation(s)
- Shaowei Guo
- Department of Biomedical Engineering , Technion-Israel Institute of Technology , Haifa 3200003 , Israel
- The First Affiliated Hospital , Shantou University Medical College , Shantou 515041 , China
| | | | - Oshra Betzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Shahar Ben-Shaul
- Department of Biomedical Engineering , Technion-Israel Institute of Technology , Haifa 3200003 , Israel
| | | | - Izhak Michaelevski
- Department of Molecular Biology , Ariel University , Ariel 40700 , Israel
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering , Technion-Israel Institute of Technology , Haifa 3200003 , Israel
| |
Collapse
|
238
|
Kim OH, Yoon OJ, Lee HJ. Silk fibroin scaffolds potentiate immunomodulatory function of human mesenchymal stromal cells. Biochem Biophys Res Commun 2019; 519:323-329. [PMID: 31506179 DOI: 10.1016/j.bbrc.2019.09.006] [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] [Received: 08/09/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022]
Abstract
Although mesenchymal stromal cells (MSCs) show great potential for use in regenerative medicine, their therapeutic efficacy remains limited because of their low adaptation efficiency and viability observed in clinical trials. To potentiate the adaptation and survival efficiency of MSCs after administration in vivo, silk fibroin nanofibers (SFNs) were applied as a scaffold. SFNs are biocompatible, biodegradable polymers with tunable architectures and mechanical properties. Treatment with interferon (IFN)-γ for 18 h increased the expression of immunomodulatory functional cytokines, IDO and COX2 in MSCs. Further, the MSCs grown on SFN sheets showed enhanced IDO1 and COX2 expression following IFN-γ treatment. MSCs showed significantly greater migratory ability on SFN sheets than on glass surfaces or PLGA control sheets. Though IFN-γ treatment slightly reduced the migration ability of MSCs cultured on glass or poly(lactic-co-glycolic acid) (PLGA) nanofiber sheets, it did not alter MSC motility on SFN sheets. Furthermore, MSCs cultured on SFN sheets dramatically suppressed TNF-α secretion from lipopolysaccharide-activated murine splenocytes, suggesting that the immunomodulatory function of MSCs was enhanced by the SFN sheets. Taken together, these data demonstrate that SFN sheets potentiate the reparative and regenerative properties of MSCs.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Ok Ja Yoon
- Da Vinci College of General Education, Chung-Ang University, Seoul, 06974, South Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
239
|
Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 2019; 76:3323-3348. [PMID: 31055643 PMCID: PMC11105258 DOI: 10.1007/s00018-019-03125-1] [Citation(s) in RCA: 357] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are isolated from multiple biological tissues-adult bone marrow and adipose tissues and neonatal tissues such as umbilical cord and placenta. In vitro, MSCs show biological features of extensive proliferation ability and multipotency. Moreover, MSCs have trophic, homing/migration and immunosuppression functions that have been demonstrated both in vitro and in vivo. A number of clinical trials are using MSCs for therapeutic interventions in severe degenerative and/or inflammatory diseases, including Crohn's disease and graft-versus-host disease, alone or in combination with other drugs. MSCs are promising for therapeutic applications given the ease in obtaining them, their genetic stability, their poor immunogenicity and their curative properties for tissue repair and immunomodulation. The success of MSC therapy in degenerative and/or inflammatory diseases might depend on the robustness of the biological functions of MSCs, which should be linked to their therapeutic potency. Here, we outline the fundamental and advanced concepts of MSC biological features and underline the biological functions of MSCs in their basic and translational aspects in therapy for degenerative and/or inflammatory diseases.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Benoit Favier
- CEA, DRF-IBFJ, IDMIT, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang Occitanie, UMR 5273 CNRS, INSERM U1031, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Francois Jacob Institute, Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, IRSL, UMRS 976, Paris, France
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
240
|
Ma T, Luan S, Tao R, Lu D, Guo L, Liu J, Shu J, Zhou X, Han Y, Jia Y, Li G, Zhang H, Han W, Han Y, Li H. Targeted Migration of Human Adipose-Derived Stem Cells to Secondary Lymphoid Organs Enhances Their Immunomodulatory Effect and Prolongs the Survival of Allografted Vascularized Composites. Stem Cells 2019; 37:1581-1594. [PMID: 31414513 DOI: 10.1002/stem.3078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
The targeted delivery of therapeutic agents to secondary lymphoid organs (SLOs), which are the niches for immune initiation, provides an unprecedented opportunity for immune intolerance induction. The alloimmune rejection postvascularized composite allotransplantation (VCA) is mediated by T lymphocytes. Human adipose-derived stem cells (hASCs) possess the superiority of convenient availability and potent immunoregulatory property, but their therapeutic results in the VCA are unambiguous thus far. Chemokine receptor 7 (CCR7) can specifically guide immune cells migrating into SLOs. There, the genes of CCR7-GFP or GFP alone were introduced into hASCs by lentivirus. hASCs/CCR7 maintained the multidifferentiation and immunoregulatory abilities, but it gained the migration capacity elicited by secondary lymphoid organ chemokine (SCL) (CCR7 ligand) in vitro. Noteworthily, intravenously infused hASCs/CCR7 targetedly relocated in the T-cell aggression area in SLOs. In a rat VCA model, hASCs/GFP transfusion had a rare effect on the allografted vascularized composite. However, hASCs/CCR7 infusion potently prolonged the grafts' survival time. The ameliorated pathologic exhibition and the regulated inflammatory cytokines in the peripheral blood were also observed. The altered axis of Th1/Th2 and Tregs/Th17 in SLOs may underlie the downregulated rejection response. Moreover, the proteomic examination of splenic T lymphocytes also confirmed that hASCs/CCR7 decreased the proteins related to cytokinesis, lymphocyte proliferation, differentiation, and apoptotic process. In conclusion, our present study demonstrated that targeted migration of hASCs/CCR7 to SLOs highly intensifies their in vivo immunomodulatory effect in the VCA model for the first time. We believe this SLO-targeting strategy may improve the clinical therapeutic efficacy of hASC for allogeneic and autogenic immune disease. Stem Cells 2019;37:1581-1594.
Collapse
Affiliation(s)
- Tian Ma
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.,Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - ShaoLiang Luan
- Department of Vascular Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Di Lu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, People's Republic of China
| | - LingLi Guo
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - JieJie Liu
- Department of Molecular Biology, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jun Shu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - XiangBin Zhou
- Department of Stomatology, The Third Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - YuDi Han
- Department of Burn and Plastic Surgery, The Seventh Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - YiQing Jia
- Department of Emergency, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Guo Li
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Hui Zhang
- Department of Plastic Surgery, The Second Hospital of Shanxi Medical University, Shanxi, People's Republic of China
| | - WeiDong Han
- Department of Molecular Biology, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, People's Republic of China
| |
Collapse
|
241
|
Cheng S, Nethi SK, Rathi S, Layek B, Prabha S. Engineered Mesenchymal Stem Cells for Targeting Solid Tumors: Therapeutic Potential beyond Regenerative Therapy. J Pharmacol Exp Ther 2019; 370:231-241. [PMID: 31175219 PMCID: PMC6640188 DOI: 10.1124/jpet.119.259796] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have previously demonstrated considerable promise in regenerative medicine based on their ability to proliferate and differentiate into cells of different lineages. More recently, there has been a significant interest in using MSCs as cellular vehicles for targeted cancer therapy by exploiting their tumor homing properties. Initial studies focused on using genetically modified MSCs for targeted delivery of various proapoptotic, antiangiogenic, and therapeutic proteins to a wide variety of tumors. However, their use as drug delivery vehicles has been limited by poor drug load capacity. This review discusses various strategies for the nongenetic modification of MSCs that allows their use in tumor-targeted delivery of small molecule chemotherapeutic agents. SIGNIFICANCE STATEMENT: There has been considerable interest in exploiting the tumor homing potential of MSCs to develop them as a vehicle for the targeted delivery of cytotoxic agents to tumor tissue. The inherent tumor-tropic and drug-resistant properties make MSCs ideal carriers for toxic payload. While significant progress has been made in the area of the genetic modification of MSCs, studies focused on identification of molecular mechanisms that contribute to the tumor tropism along with optimization of the engineering conditions can further improve their effectiveness as drug delivery vehicles.
Collapse
Affiliation(s)
- Shen Cheng
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Susheel Kumar Nethi
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Sneha Rathi
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Buddhadev Layek
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Swayam Prabha
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| |
Collapse
|
242
|
Kwon S, Yoo KH, Sym SJ, Khang D. Mesenchymal stem cell therapy assisted by nanotechnology: a possible combinational treatment for brain tumor and central nerve regeneration. Int J Nanomedicine 2019; 14:5925-5942. [PMID: 31534331 PMCID: PMC6681156 DOI: 10.2147/ijn.s217923] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) intrinsically possess unique features that not only help in their migration towards the tumor-rich environment but they also secrete versatile types of secretomes to induce nerve regeneration and analgesic effects at inflammatory sites. As a matter of course, engineering MSCs to enhance their intrinsic abilities is growing in interest in the oncology and regenerative field. However, the concern of possible tumorigenesis of genetically modified MSCs prompted the development of non-viral transfected MSCs armed with nanotechnology for more effective cancer and regenerative treatment. Despite the fact that a large number of successful studies have expanded our current knowledge in tumor-specific targeting, targeting damaged brain site remains enigmatic due to the presence of a blood–brain barrier (BBB). A BBB is a barrier that separates blood from brain, but MSCs with intrinsic features of transmigration across the BBB can efficiently deliver desired drugs to target sites. Importantly, MSCs, when mediated by nanoparticles, can further enhance tumor tropism and can regenerate the damaged neurons in the central nervous system through the promotion of axon growth. This review highlights the homing and nerve regenerative abilities of MSCs in order to provide a better understanding of potential cell therapeutic applications of non-genetically engineered MSCs with the aid of nanotechnology.
Collapse
Affiliation(s)
- Song Kwon
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Kwai Han Yoo
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Sun Jin Sym
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea.,Department of Gachon Advanced Institute for Health Science & Technology (Gaihst), Gachon University, Incheon 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
243
|
Chiu SP, Lee YW, Wu LY, Tung TH, Gomez S, Lo CM, Wang JY. Application of ECIS to Assess FCCP-Induced Changes of MSC Micromotion and Wound Healing Migration. SENSORS 2019; 19:s19143210. [PMID: 31330904 PMCID: PMC6679573 DOI: 10.3390/s19143210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Electric cell-substrate impedance sensing (ECIS) is an emerging technique for sensitively monitoring morphological changes of adherent cells in tissue culture. In this study, human mesenchymal stem cells (hMSCs) were exposed to different concentrations of carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) for 20 h and their subsequent concentration-dependent responses in micromotion and wound healing migration were measured by ECIS. FCCP disrupts ATP synthesis and results in a decrease in cell migration rates. To detect the change of cell micromotion in response to FCCP challenge, time-series resistances of cell-covered electrodes were monitored and the values of variance were calculated to verify the difference. While Seahorse XF-24 extracellular flux analyzer can detect the effect of FCCP at 3 μM concentration, the variance calculation of the time-series resistances measured at 4 kHz can detect the effect of FCCP at concentrations as low as 1 μM. For wound healing migration, the recovery resistance curves were fitted by sigmoid curve and the hill slope showed a concentration-dependent decline from 0.3 μM to 3 μM, indicating a decrease in cell migration rate. Moreover, dose dependent incline of the inflection points from 0.3 μM to 3 μM FCCP implied the increase of the half time for wound recovery migration. Together, our results demonstrate that partial uncoupling of mitochondrial oxidative phosphorylation reduces micromotion and wound healing migration of hMSCs. The ECIS method used in this study offers a simple and sensitive approach to investigate stem cell migration and its regulation by mitochondrial dynamics.
Collapse
Affiliation(s)
- Sheng-Po Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yu-Wei Lee
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Ling-Yi Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tse-Hua Tung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Sofia Gomez
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
244
|
Shojaee A, Parham A, Ejeian F, Nasr Esfahani MH. Equine adipose mesenchymal stem cells (eq-ASCs) appear to have higher potential for migration and musculoskeletal differentiation. Res Vet Sci 2019; 125:235-243. [PMID: 31310927 DOI: 10.1016/j.rvsc.2019.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 05/14/2019] [Accepted: 06/30/2019] [Indexed: 12/28/2022]
Abstract
Equine adipose-derived mesenchymal stem cells (eq-ASCs) possess excellent regeneration potential especially for treatment of musculoskeletal disorders. Besides their common characteristics, MSCs harvested from different species reveal some species-specific and donor-dependent behaviors. Hence, the molecular analysis of MSCs may shed more light on their future clinical application of these cells. This study aimed to investigate some behavioral aspects of eq-ASCs in vitro which may influence the efficacy of stem cell therapy. For this purpose, MSCs of a donor horse were isolated, characterized and expanded under normal culture conditions. During continuous culture condition, eq-ASCs were started to formed aggregated structures that was accompanied with the up-regulation of migratory related genes including transforming growth factor beta 1 (TGFB1) and its receptor 3 (TGFBR3), and snail family transcriptional repressor 1 (SNAI1), E-cadherin (CDH1) and β-catenin (CTNNB1). Moreover, the expression of a musculoskeletal progenitor marker, scleraxis bHLH transcription factor (SCX), was also increased after 3 days. In order to clarify the impact of TGFB signaling pathway on cultured cells, gain- and loss-of-function treatment by TGFB3 and SB431542 (TGFB inhibitor) were performed, respectively. We found that TGFB3 treatment exaggerated the aggregate formation effects, in some extend via induction of cytoskeletal actin rearrangement, while inhibition of TGFB signaling pathway by SB431542 reversed this phenomenon. Overall, our findings support the fact that eq-ASCs have an inherent capacity for migration, which was enhanced by TGFB3 treatment and, this ability may play crucial role in cell motility and wound healing of transplanted cells.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abbas Parham
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell Biology and Alternative Regenerative Medicine Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Fatemeh Ejeian
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
245
|
Ahn YJ, Kong TH, Choi JS, Yun WS, Key J, Seo YJ. Strategies to enhance efficacy of SPION-labeled stem cell homing by magnetic attraction: a systemic review with meta-analysis. Int J Nanomedicine 2019; 14:4849-4866. [PMID: 31308662 PMCID: PMC6613362 DOI: 10.2147/ijn.s204910] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Stem cells possess a promising potential in the clinical field. The application and effective delivery of stem cells to the desired target organ or site of injury plays an important role. This review describes strategies on understanding the effective delivery of stem cells labeled with superparamagnetic iron oxide nanoparticles (SPION) using an external magnet to enhance stem cell migration in vivo and in vitro. Fourteen total publications among 174 articles were selected. Stem cell type, SPION characteristics, labeling time, and magnetic force in vivo are considered important factors affecting the effective delivery of stem cells to the homing site. Most papers reported that the efficiency was increased when magnet is applied compared to those without. Ten studies analyzed the homing competency of SPION-labeled MSCs in vitro by observing the migration of the cell toward the external magnet. In cell-based experiments, the mechanism of magnetic attraction, the kind of nanoparticles, and various stem cells were studied well. Meta-analysis has shown the mean size of nanoparticles and degree of recovery or regeneration of damaged target organs upon in vivo studies. This strategy may provide a guideline for designing studies involving stem cell homing and further expand stem cell.
Collapse
Affiliation(s)
- Ye Ji Ahn
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Tae Hoon Kong
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Jin Sil Choi
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Wan Su Yun
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Jaehong Key
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Young Joon Seo
- Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| |
Collapse
|
246
|
Conley SM, Shook JE, Zhu XY, Eirin A, Jordan KL, Woollard JR, Isik B, Hickson LJ, Puranik AS, Lerman LO. Metabolic Syndrome Induces Release of Smaller Extracellular Vesicles from Porcine Mesenchymal Stem Cells. Cell Transplant 2019; 28:1271-1278. [PMID: 31250656 PMCID: PMC6767891 DOI: 10.1177/0963689719860840] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) belong to the endogenous cellular reparative
system, and can be used exogenously in cell-based therapy. MSCs release extracellular
vesicles (EVs), including exosomes and microvesicles, which mediate some of their
therapeutic activity through intercellular communication. We have previously demonstrated
that metabolic syndrome (MetS) modifies the cargo packed within swine EV, but whether it
influences their phenotypical characteristics remains unclear. This study tested the
hypothesis that MetS shifts the size distribution of MSC-derived EVs. Adipose
tissue-derived MSC-EV subpopulations from Lean (n = 6) and MetS
(n = 6) pigs were characterized for number and size using
nanoparticle-tracking analysis, flow cytometry, and transmission electron microscopy.
Expression of exosomal genes was determined using next-generation RNA-sequencing
(RNA-seq). The number of EV released from Lean and MetS pig MSCs was similar, yet
MetS-MSCs yielded a higher proportion of small-size EVs (202.4 ± 17.7 nm vs. 280.3 ± 15.1
nm), consistent with exosomes. RNA-seq showed that their EVs were enriched with exosomal
markers. Lysosomal activity remained unaltered in MetS-MSCs. Therefore, MetS alters the
size distribution of MSC-derived EVs in favor of exosome release. These observations may
reflect MSC injury and membrane recycling in MetS or increased expulsion of waste
products, and may have important implications for development of adequate cell-based
treatments.
Collapse
Affiliation(s)
- Sabena M Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - John E Shook
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA.,Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Busra Isik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Amrutesh S Puranik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA.,Division of Rheumatology, Colton Center for Autoimmunity, New York University Langone Medical Center, New York, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| |
Collapse
|
247
|
Shojaee A, Parham A. Strategies of tenogenic differentiation of equine stem cells for tendon repair: current status and challenges. Stem Cell Res Ther 2019; 10:181. [PMID: 31215490 PMCID: PMC6582602 DOI: 10.1186/s13287-019-1291-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tendon injuries, as one of the most common orthopedic disorders, are the major cause of early retirement or wastage among sport horses which mainly affect the superficial digital flexor tendon (SDFT). Tendon repair is a slow process, and tendon tissue is often replaced by scar tissue. The current treatment options are often followed by an incomplete recovery that increases the susceptibility to re-injury. Recently, cell therapy has been used in veterinary medicine to treat tendon injuries, although the risk of ectopic bone formation after cell injection is possible in some cases. In vitro tenogenic induction may overcome the mentioned risk in clinical application. Moreover, a better understanding of treatment strategies for musculoskeletal injuries in horse may have future applications for human and vice versa. This comprehensive review outlines the current strategies of stem cell therapy in equine tendon injury and in vitro tenogenic induction of equine stem cell.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abbas Parham
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran. .,Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
248
|
Timaner M, Tsai KK, Shaked Y. The multifaceted role of mesenchymal stem cells in cancer. Semin Cancer Biol 2019; 60:225-237. [PMID: 31212021 DOI: 10.1016/j.semcancer.2019.06.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells derived from the mesoderm that give rise to several mesenchymal lineages, including osteoblasts, adipocytes, chondrocytes and myocytes. Their potent ability to home to tumors coupled with their differentiation potential and immunosuppressive function positions MSCs as key regulators of tumor fate. Here we review the existing knowledge on the involvement of MSCs in multiple tumor-promoting processes, including angiogenesis, epithelial-mesenchymal transition, metastasis, immunosuppression and therapy resistance. We also discuss the clinical potential of MSC-based therapy for cancer.
Collapse
Affiliation(s)
- Michael Timaner
- Technion-Integerated Cancer Center, Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Kelvin K Tsai
- Laboratory of Advanced Molecular Therapeutics, and Division of Gastroenterology, Wan Fang Hospital, and Graduate Institutes of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei Taiwan; National Institute of Cancer Research, National Health Research Institutes, Taiwan
| | - Yuval Shaked
- Technion-Integerated Cancer Center, Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
249
|
Perets N, Betzer O, Shapira R, Brenstein S, Angel A, Sadan T, Ashery U, Popovtzer R, Offen D. Golden Exosomes Selectively Target Brain Pathologies in Neurodegenerative and Neurodevelopmental Disorders. NANO LETTERS 2019; 19:3422-3431. [PMID: 30761901 DOI: 10.1021/acs.nanolett.8b04148] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Exosomes, nanovesicles that are secreted by different cell types, enable intercellular communication at local or distant sites. Alhough they have been found to cross the blood brain barrier, their migration and homing abilities within the brain remain unstudied. We have recently developed a method for longitudinal and quantitative in vivo neuroimaging of exosomes based on the superior visualization abilities of classical X-ray computed tomography (CT), combined with gold nanoparticles as labeling agents. Here, we used this technique to track the migration and homing patterns of intranasally administrated exosomes derived from bone marrow mesenchymal stem cells (MSC-exo) in different brain pathologies, including stroke, autism, Parkinson's disease, and Alzheimer's disease. We found that MSC-exo specifically targeted and accumulated in pathologically relevant murine models brains regions up to 96 h post administration, while in healthy controls they showed a diffuse migration pattern and clearance by 24 h. The neuro-inflammatory signal in pathological brains was highly correlated with MSC-exo accumulation, suggesting that the homing mechanism is inflammatory-driven. In addition, MSC-exo were selectively uptaken by neuronal cells, but not glial cells, in the pathological regions. Taken together, these findings can significantly promote the application of exosomes for therapy and targeted drug delivery in various brain pathologies.
Collapse
Affiliation(s)
- Nisim Perets
- Sagol School of Neuroscience , Tel Aviv University , Tel Aviv 6997801 , Israel
- Sacklar School of Medicine, Department of Human Genetics and Biochemistry , Tel Aviv University , Tel Aviv 6997801 , Israel
| | - Oshra Betzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Ronit Shapira
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty , Tel Aviv University , Tel Aviv 6997801 , Israel
| | - Shmuel Brenstein
- Sagol School of Neuroscience , Tel Aviv University , Tel Aviv 6997801 , Israel
| | - Ariel Angel
- Sagol School of Neuroscience , Tel Aviv University , Tel Aviv 6997801 , Israel
| | - Tamar Sadan
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Uri Ashery
- Sagol School of Neuroscience , Tel Aviv University , Tel Aviv 6997801 , Israel
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty , Tel Aviv University , Tel Aviv 6997801 , Israel
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat Gan 5290002 , Israel
| | - Daniel Offen
- Sagol School of Neuroscience , Tel Aviv University , Tel Aviv 6997801 , Israel
- Sacklar School of Medicine, Department of Human Genetics and Biochemistry , Tel Aviv University , Tel Aviv 6997801 , Israel
| |
Collapse
|
250
|
Current Trends and Future Perspective of Mesenchymal Stem Cells and Exosomes in Corneal Diseases. Int J Mol Sci 2019; 20:ijms20122853. [PMID: 31212734 PMCID: PMC6627168 DOI: 10.3390/ijms20122853] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/01/2019] [Accepted: 06/10/2019] [Indexed: 02/07/2023] Open
Abstract
The corneal functions (transparency, refractivity and mechanical strength) deteriorate in many corneal diseases but can be restored after corneal transplantation (penetrating and lamellar keratoplasties). However, the global shortage of transplantable donor corneas remains significant and patients are subject to life-long risk of immune response and graft rejection. Various studies have shown the differentiation of multipotent mesenchymal stem cells (MSCs) into various corneal cell types. With the unique properties of immunomodulation, anti-angiogenesis and anti-inflammation, they offer the advantages in corneal reconstruction. These effects are widely mediated by MSC differentiation and paracrine signaling via exosomes. Besides the cell-free nature of exosomes in circumventing the problems of cell-fate control and tumorigenesis, the vesicle content can be genetically modified for optimal therapeutic affinity. The pharmacology and toxicology, xeno-free processing with sustained delivery, scale-up production in compliant to Good Manufacturing Practice regulations, and cost-effectiveness are the current foci of research. Routes of administration via injection, topical and/or engineered bioscaffolds are also explored for its applicability in treating corneal diseases.
Collapse
|