401
|
Barnes CH, Maas CS. Autologous Fat Harvest and Preparation for Optimal Predictable Outcomes. Facial Plast Surg Clin North Am 2019; 27:419-423. [PMID: 31280857 DOI: 10.1016/j.fsc.2019.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Best practices in fat transfer to the face focus on tissue harvest and processing techniques. This article discusses the role of adipose-derived mesenchymal stem cells (MSCs) in mitigating tissue loss in grafting. Discrepancies among common practice and recent study results have propagated uncertainty with long-term results. Fortunately, recent increases in the understanding of these MSCs are leading providers to identify statistically more favorable tissue donor sites, harvest technique, and preparation methods to increase their concentration in transferred tissue. Future studies are needed to support or confound the long-term effects of MSC transfer on facial fat grafting.
Collapse
Affiliation(s)
- Christian H Barnes
- Facial Plastic and Reconstructive Surgery, The Maas Clinic, 2400 Clay Street, San Francisco, CA 94115, USA
| | - Corey S Maas
- Facial Plastic and Reconstructive Surgery, The Maas Clinic, 2400 Clay Street, San Francisco, CA 94115, USA.
| |
Collapse
|
402
|
Promoting Osteogenic Differentiation of Human Adipose-Derived Stem Cells by Altering the Expression of Exosomal miRNA. Stem Cells Int 2019; 2019:1351860. [PMID: 31354836 PMCID: PMC6636464 DOI: 10.1155/2019/1351860] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Human adipose-derived stem cells (ADSCs) can release exosomes; however, their specific functions remain elusive. In this study, we verified that exosomes derived from osteogenically differentiated ADSCs can promote osteogenic differentiation of ADSCs. Furthermore, in order to investigate the importance of exosomal microRNAs (miRNAs) in osteogenic differentiation of ADSCs, we used microarray assays to analyze the expression profiles of exosomal miRNAs derived from undifferentiated as well as osteogenically differentiated ADSCs; 201 miRNAs were upregulated and 33 miRNAs were downregulated between the two types of exosomes. Additionally, bioinformatic analyses, which included gene ontology analyses, pathway analysis, and miRNA-mRNA-network investigations, were performed. The results of these analyses revealed that the differentially expressed exosomal miRNAs participate in multiple biological processes, such as gene expression, synthesis of biomolecules, cell development, differentiation, and signal transduction, among others. Moreover, we found that these differentially expressed exosomal miRNAs connect osteogenic differentiation to processes such as axon guidance, MAPK signaling, and Wnt signaling. To the best of our knowledge, this is the first study to identify and characterize exosomal miRNAs derived from osteogenically differentiated ADSCs. This study confirms that alterations in the expression of exosomal miRNAs can promote osteogenic differentiation of ADSCs, which also provides the foundation for further research on the regulatory functions of exosomal miRNAs in the context of ADSC osteogenesis.
Collapse
|
403
|
Ronchi G, Morano M, Fregnan F, Pugliese P, Crosio A, Tos P, Geuna S, Haastert-Talini K, Gambarotta G. The Median Nerve Injury Model in Pre-clinical Research - A Critical Review on Benefits and Limitations. Front Cell Neurosci 2019; 13:288. [PMID: 31316355 PMCID: PMC6609919 DOI: 10.3389/fncel.2019.00288] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
The successful introduction of innovative treatment strategies into clinical practise strongly depends on the availability of effective experimental models and their reliable pre-clinical assessment. Considering pre-clinical research for peripheral nerve repair and reconstruction, the far most used nerve regeneration model in the last decades is the sciatic nerve injury and repair model. More recently, the use of the median nerve injury and repair model has gained increasing attention due to some significant advantages it provides compared to sciatic nerve injury. Outstanding advantages are the availability of reliable behavioural tests for assessing posttraumatic voluntary motor recovery and a much lower impact on the animal wellbeing. In this article, the potential application of the median nerve injury and repair model in pre-clinical research is reviewed. In addition, we provide a synthetic overview of a variety of methods that can be applied in this model for nerve regeneration assessment. This article is aimed at helping researchers in adequately adopting this in vivo model for pre-clinical evaluation of peripheral nerve reconstruction as well as for interpreting the results in a translational perspective.
Collapse
Affiliation(s)
- Giulia Ronchi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi Foundation (NICO), University of Turin, Turin, Italy
| | - Michela Morano
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi Foundation (NICO), University of Turin, Turin, Italy
| | - Federica Fregnan
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi Foundation (NICO), University of Turin, Turin, Italy
| | - Pierfrancesco Pugliese
- Dipartimento di Chirurgia Generale e Specialistica, Azienda Ospedaliera Universitaria, Ancona, Italy
| | - Alessandro Crosio
- UO Microchirurgia e Chirurgia della Mano, Ospedale Gaetano Pini, Milan, Italy
| | - Pierluigi Tos
- UO Microchirurgia e Chirurgia della Mano, Ospedale Gaetano Pini, Milan, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi Foundation (NICO), University of Turin, Turin, Italy
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hanover, Germany.,Center for Systems Neuroscience (ZSN) Hannover, Hanover, Germany
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
404
|
Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci 2019; 20:ijms20123105. [PMID: 31242644 PMCID: PMC6627452 DOI: 10.3390/ijms20123105] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic musculoskeletal (MSK) pain is one of the most common medical complaints worldwide and musculoskeletal injuries have an enormous social and economical impact. Current pharmacological and surgical treatments aim to relief pain and restore function; however, unsatiscactory outcomes are commonly reported. In order to find an accurate treatment to such pathologies, over the last years, there has been a significantly increasing interest in cellular therapies, such as adipose-derived mesenchymal stem cells (AMSCs). These cells represent a relatively new strategy in regenerative medicine, with many potential applications, especially regarding MSK disorders, and preclinical and clinical studies have demonstrated their efficacy in muscle, tendon, bone and cartilage regeneration. Nevertheless, several worries about their safety and side effects at long-term remain unsolved. This article aims to review the current state of AMSCs therapy in the treatment of several MSK diseases and their clinical applications in veterinary and human medicine.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
405
|
Costa-Almeida R, Calejo I, Gomes ME. Mesenchymal Stem Cells Empowering Tendon Regenerative Therapies. Int J Mol Sci 2019; 20:E3002. [PMID: 31248196 PMCID: PMC6627139 DOI: 10.3390/ijms20123002] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/19/2022] Open
Abstract
Tendon tissues have limited healing capacity. The incidence of tendon injuries and the unsatisfactory functional outcomes of tendon repair are driving the search for alternative therapeutic approaches envisioning tendon regeneration. Cellular therapies aim at delivering adequate, regeneration-competent cell types to the injured tendon and toward ultimately promoting its reconstruction and recovery of functionality. Mesenchymal stem cells (MSCs) either obtained from tendons or from non-tendon sources, like bone marrow (BM-MSCs) or adipose tissue (ASCs), have been receiving increasing attention over the years toward enhancing tendon healing. Evidences from in vitro and in vivo studies suggest MSCs can contribute to accelerate and improve the quality of tendon healing. Nonetheless, the exact mechanisms underlying these repair events are yet to be fully elucidated. This review provides an overview of the main challenges in the field of cell-based regenerative therapies, discussing the role of MSCs in boosting tendon regeneration, particularly through their capacity to enhance the tenogenic properties of tendon resident cells.
Collapse
Affiliation(s)
- Raquel Costa-Almeida
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
| | - Isabel Calejo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
406
|
Efficacy of Mesenchymal Stem/Stromal Cell Infusion in Septic Patients Is a Puzzle Worthy of Attention. Crit Care Med 2019; 47:1001-1002. [PMID: 31205082 DOI: 10.1097/ccm.0000000000003783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
407
|
Bourebaba L, Bedjou F, Röcken M, Marycz K. Nortropane alkaloids as pharmacological chaperones in the rescue of equine adipose-derived mesenchymal stromal stem cells affected by metabolic syndrome through mitochondrial potentiation, endoplasmic reticulum stress mitigation and insulin resistance alleviation. Stem Cell Res Ther 2019; 10:178. [PMID: 31215461 PMCID: PMC6582509 DOI: 10.1186/s13287-019-1292-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/20/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Equine metabolic syndrome (EMS) refers to a cluster of associated abnormalities and metabolic disorders, including insulin resistance and adiposity. The numerous biological properties of mesenchymal stem cells (MSCs), including self-renewal and multipotency, have been the subject of many in-depth studies, for the management of EMS; however, it has been shown that this cell type may be affected by the condition, impairing thus seriously their therapeutic potential. Therefore, an attempt to rescue EMS adipose-derived stem cells (ASCs) with calystegines (polyhydroxylated alkaloids) that are endowed with strong antioxidant and antidiabetic abilities was performed. METHODS ASCs isolated from EMS horses were subsequently treated with various concentrations of total calystegines. Different parameters were then assessed using flow cytometry, confocal as well as SE microscopy, and RT-qPCR. RESULTS Our results clearly demonstrated that calystegines could improve EqASC viability and proliferation and significantly reduce apoptosis, via improvement of mitochondrial potentiation and functionality, regulation of pro- and anti-apoptotic pathways, and suppression of ER stress. Furthermore, nortropanes positively upregulated GLUT4 and IRS transcripts, indicating a possible sensitizing or mimetic effect to insulin. Most interesting finding in this investigation lies in the modulatory effect of autophagy, a process that allows the maintenance of cellular homeostasis; calystegines acted as pharmacological chaperones to promote cell survival. CONCLUSION Obtained data open new perspectives in the development of new drugs, which may improve the metabolic dynamics of cells challenged by MS.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| | - Fatiha Bedjou
- Laboratoire de Biotechnologies végétales et d'Ethnobotanique, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000, Bejaia, Algeria
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
408
|
Li Z, Yan G, Diao Q, Yu F, Li X, Sheng X, Liu Y, Dai Y, Zhou H, Zhen X, Hu Y, Péault B, Ding L, Sun H, Li H. Transplantation of human endometrial perivascular cells with elevated CYR61 expression induces angiogenesis and promotes repair of a full-thickness uterine injury in rat. Stem Cell Res Ther 2019; 10:179. [PMID: 31215503 PMCID: PMC6582612 DOI: 10.1186/s13287-019-1272-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/25/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Disruptions of angiogenesis can have a significant effect on the healing of uterine scars. Human endometrial perivascular cells (CD146+PDGFRβ+) function as stem cells in the endometrium. Cysteine-rich angiogenic inducer 61 (CYR61) plays an important role in vascular development. The purpose of this study was to observe the effects of the transplantation of human endometrial perivascular cells (En-PSCs) overexpressing CYR61 on structural and functional regeneration in rat models of partial full-thickness uterine excision. METHODS We first sorted human En-PSCs from endometrial single-cell suspensions by flow cytometry. Human En-PSCs expressing low or high levels of CYR61 were then generated via transfection with a CYR61-specific small interfering ribonucleic acid (si-CYR61) construct or overexpression plasmid. To establish a rat model of uterine injury, a subset of uterine wall was then resected from each uterine horn in experimental animals. Female rats were randomly assigned to five groups, including a sham-operated group and four repair groups that received either PBS loaded on a collagen scaffold (collagen/PBS), En-PSCs loaded on a collagen scaffold (collagen/En-PSCs), En-PSCs with low CYR61 expression loaded on a collagen scaffold (collagen/si-CYR61 En-PSCs), and En-PSCs overexpressing CYR61 loaded on a collagen scaffold (collagen/ov-CYR61 En-PSCs). These indicated constructs were sutured in the injured uterine area to replace the excised segment. On days 30 and 90 after transplantation, a subset of rats in each group was sacrificed, and uterine tissue was recovered and serially sectioned. Hematoxylin and eosin staining and immunohistochemical staining were then performed. Finally, the remaining rats of each group were mated with fertile male rats on day 90 for a 2-week period. RESULTS Sorted En-PSCs expressed all recognized markers of mesenchymal stem cells (MSCs), including CD10, CD13, CD44, CD73, CD90, and CD105, and exhibited differentiation potential toward adipocytes, osteoblasts, and neuron-like cells. Compared with En-PSCs and En-PSCs with low CYR61 expression, En-PSCs with elevated CYR61 expression enhanced angiogenesis by in vitro co-culture assays. At day 90 after transplantation, blood vessel density in the collagen/ov-CYR61 En-PSCs group (11.667 ± 1.287) was greater than that in the collagen/En-PSCs group (7.167 ± 0.672) (P < 0.05) and the collagen/si-CYR61 En-PSCs group (3.750 ± 0.906) (P < 0.0001). Pregnancy rates differed among groups, from 40% in the collagen/PBS group to 80% in the collagen/En-PSCs group, 12.5% in the collagen/si-CYR61 En-PSCs group, and 80% in the collagen/ov-CYR61 En-PSCs group. In addition, four embryos were evident in the injured uterine horns of the collagen/ov-CYR61 En-PSCs group, while no embryos were identified in the injured uterine horns of the collagen/PBS group. CONCLUSIONS The results showed that CYR61 plays an important role in angiogenesis. Collagen/ov-CYR61 En-PSCs promoted endometrial and myometrial regeneration and induced neovascular regeneration in injured rat uteri. The pregnancy rate of rats treated with transplantation of collagen/En-PSCs or collagen/ov-CYR61 En-PSCs was improved. Moreover, the number of embryos implantation on the injured area in uterus was increased after transplantation of collagen/ov-CYR61 En-PSCs.
Collapse
Affiliation(s)
- Zhongxun Li
- Department of Histology and Embryology of Shanxi Medical University, Taiyuan, 030001 China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Guijun Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Qiang Diao
- Department of Medical Imaging, Jinling Hospital, Nanjing University Medical School, Nanjing, 210002 China
| | - Fei Yu
- Center for Experimental Animal, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xin’an Li
- Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xiaoqiang Sheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Yong Liu
- Department of Experimental Medicine, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Yimin Dai
- Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Huaijun Zhou
- Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xin Zhen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Yali Hu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Bruno Péault
- UKMRC Center for Regenerative Medicine and Center for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Lijun Ding
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
- UKMRC Center for Regenerative Medicine and Center for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
- Clinical Center for Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Haixiang Sun
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
- Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Hairong Li
- Department of Histology and Embryology of Shanxi Medical University, Taiyuan, 030001 China
| |
Collapse
|
409
|
Yuan QL, Zhang YG, Chen Q. Mesenchymal Stem Cell (MSC)-Derived Extracellular Vesicles: Potential Therapeutics as MSC Trophic Mediators in Regenerative Medicine. Anat Rec (Hoboken) 2019; 303:1735-1742. [PMID: 31168963 DOI: 10.1002/ar.24186] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/29/2018] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are pluripotent progenitor cells with the capabilities of self-renewing, differentiating into multiple lineages, and achieving trophic effects during tissue repair. MSCs can secrete extracellular vesicles (EVs) including exosomes and microvesicles, which mediate their trophic effects on other cells. Carrying a variety of intracellular molecules of MSCs including lipids, proteins, RNA (mRNA and noncoding RNA), and DNA, EVs deliver them into other cells to regulate tissue regeneration process. The therapeutic effects of MSC-derived EVs have been observed in a number of animal disease models. In this review, we focus on the current state and future directions of MSC-derived EVs in regenerative medicine. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qi-Ling Yuan
- Department of Orthopaedics, Bone and Joint Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yin-Gang Zhang
- Department of Orthopaedics, Bone and Joint Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qian Chen
- Department of Orthopaedics, Brown University, Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
410
|
Sauerova P, Suchy T, Supova M, Bartos M, Klima J, Juhasova J, Juhas S, Kubikova T, Tonar Z, Sedlacek R, Piola M, Fiore GB, Soncini M, Hubalek Kalbacova M. Positive impact of dynamic seeding of mesenchymal stem cells on bone-like biodegradable scaffolds with increased content of calcium phosphate nanoparticles. Mol Biol Rep 2019; 46:4483-4500. [DOI: 10.1007/s11033-019-04903-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/30/2019] [Indexed: 12/01/2022]
|
411
|
Szychlinska MA, D'Amora U, Ravalli S, Ambrosio L, Di Rosa M, Musumeci G. Functional Biomolecule Delivery Systems and Bioengineering in Cartilage Regeneration. Curr Pharm Biotechnol 2019; 20:32-46. [PMID: 30727886 DOI: 10.2174/1389201020666190206202048] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
Osteoarthritis (OA) is a common degenerative disease which involves articular cartilage, and leads to total joint disability in the advanced stages. Due to its avascular and aneural nature, damaged cartilage cannot regenerate itself. Stem cell therapy and tissue engineering represent a promising route in OA therapy, in which cooperation of mesenchymal stem cells (MSCs) and three-dimensional (3D) scaffolds contribute to cartilage regeneration. However, this approach still presents some limits such as poor mechanical properties of the engineered cartilage. The natural dynamic environment of the tissue repair process involves a collaboration of several signals expressed in the biological system in response to injury. For this reason, tissue engineering involving exogenous "influencers" such as mechanostimulation and functional biomolecule delivery systems (BDS), represent a promising innovative approach to improve the regeneration process. BDS provide a controlled release of biomolecules able to interact between them and with the injured tissue. Nano-dimensional BDS is the future hope for the design of personalized scaffolds, able to overcome the delivery problems. MSC-derived extracellular vesicles (EVs) represent an attractive alternative to BDS, due to their innate targeting abilities, immunomodulatory potential and biocompatibility. Future advances in cartilage regeneration should focus on multidisciplinary strategies such as modular assembly strategies, EVs, nanotechnology, 3D biomaterials, BDS, mechanobiology aimed at constructing the functional scaffolds for actively targeted biomolecule delivery. The aim of this review is to run through the different approaches adopted for cartilage regeneration, with a special focus on biomaterials, BDS and EVs explored in terms of their delivery potential, healing capabilities and mechanical features.
Collapse
Affiliation(s)
- Marta A Szychlinska
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia no. 87, Catania, Italy
| | - Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy, 54, Mostra d'Oltremare Pad. 20, 80125, Naples, Italy
| | - Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia no. 87, Catania, Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy, 54, Mostra d'Oltremare Pad. 20, 80125, Naples, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia no. 87, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia no. 87, Catania, Italy
| |
Collapse
|
412
|
Soria-Juan B, Escacena N, Capilla-González V, Aguilera Y, Llanos L, Tejedo JR, Bedoya FJ, Juan V, De la Cuesta A, Ruiz-Salmerón R, Andreu E, Grochowicz L, Prósper F, Sánchez-Guijo F, Lozano FS, Miralles M, Del Río-Solá L, Castellanos G, Moraleda JM, Sackstein R, García-Arranz M, García-Olmo D, Martín F, Hmadcha A, Soria B. Cost-Effective, Safe, and Personalized Cell Therapy for Critical Limb Ischemia in Type 2 Diabetes Mellitus. Front Immunol 2019; 10:1151. [PMID: 31231366 PMCID: PMC6558400 DOI: 10.3389/fimmu.2019.01151] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/07/2019] [Indexed: 12/26/2022] Open
Abstract
Cell therapy is a progressively growing field that is rapidly moving from preclinical model development to clinical application. Outcomes obtained from clinical trials reveal the therapeutic potential of stem cell-based therapy to deal with unmet medical treatment needs for several disorders with no therapeutic options. Among adult stem cells, mesenchymal stem cells (MSCs) are the leading cell type used in advanced therapies for the treatment of autoimmune, inflammatory and vascular diseases. To date, the safety and feasibility of autologous MSC-based therapy has been established; however, their indiscriminate use has resulted in mixed outcomes in preclinical and clinical studies. While MSCs derived from diverse tissues share common properties depending on the type of clinical application, they markedly differ within clinical trials in terms of efficacy, resulting in many unanswered questions regarding the application of MSCs. Additionally, our experience in clinical trials related to critical limb ischemia pathology (CLI) shows that the therapeutic efficacy of these cells in different animal models has only been partially reproduced in humans through clinical trials. Therefore, it is crucial to develop new research to identify pitfalls, to optimize procedures and to clarify the repair mechanisms used by these cells, as well as to be able to offer a next generation of stem cell that can be routinely used in a cost-effective and safe manner in stem cell-based therapies targeting CLI.
Collapse
Affiliation(s)
| | - Natalia Escacena
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Vivian Capilla-González
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Yolanda Aguilera
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Lucía Llanos
- Fundación Jiménez Díaz Health Research Institute, Madrid, Spain
| | - Juan R Tejedo
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Francisco J Bedoya
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | | | - Antonio De la Cuesta
- Unidad de Isquemia Crónica de Miembros Inferiores, Hospital Victoria Eugenia de la Cruz Roja, Sevilla, Spain
| | | | | | | | | | | | | | - Manuel Miralles
- Department of Surgery, University of Valencia, Valencia, Spain
| | | | - Gregorio Castellanos
- Servicio Hematología y Hemoterapia, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - José M Moraleda
- Servicio Hematología y Hemoterapia, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Robert Sackstein
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | | | | | - Franz Martín
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Bernat Soria
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | | |
Collapse
|
413
|
Castiglione F, Hedlund P, Weyne E, Hakim L, Montorsi F, Salonia A, Bivalacqua TJ, De Ridder D, Milenkovic U, Ralph D, Garaffa G, Muneer A, Joniau S, Albersen M. Intratunical injection of stromal vascular fraction prevents fibrosis in a rat model of Peyronie's disease. BJU Int 2019; 124:342-348. [DOI: 10.1111/bju.14570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Fabio Castiglione
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
- Institute of Urology; University College of London Hospital; London UK
- Division of Oncology/Unit of Urology; Urological Research Institute; IRCCS Ospedale San Raffaele; Milan Italy
| | - Petter Hedlund
- Department of Clinical and Experimental Pharmacology; Lund University; Lund Sweden
- Division of Drug Research; Department of Medical and Health Sciences; Linköping University; Linköping Sweden
| | - Emmanuel Weyne
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Lukman Hakim
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
- Department of Urology; Airlangga University / Dr Soetomo General Hospital; Surabaya Indonesia
| | - Francesco Montorsi
- Division of Oncology/Unit of Urology; Urological Research Institute; IRCCS Ospedale San Raffaele; Milan Italy
| | - Andrea Salonia
- Division of Oncology/Unit of Urology; Urological Research Institute; IRCCS Ospedale San Raffaele; Milan Italy
| | - Trinity J. Bivalacqua
- James Buchanan Brady Urological Institute; Department of Urology; Johns Hopkins Medical Institutions; Baltimore MD USA
| | - Dirk De Ridder
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Uros Milenkovic
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - David Ralph
- Institute of Urology; University College of London Hospital; London UK
| | - Giulio Garaffa
- Institute of Urology; University College of London Hospital; London UK
| | - Asif Muneer
- Institute of Urology; University College of London Hospital; London UK
- Division of Surgery and Interventional Science; NIHR Biomedical Research Centre; University College London Hospital; London UK
| | - Steven Joniau
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Maarten Albersen
- Laboratory for Experimental Urology; Organ Systems; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | | |
Collapse
|
414
|
Emerging Role of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Pathogenesis of Haematological Malignancies. Stem Cells Int 2019; 2019:6854080. [PMID: 31281380 PMCID: PMC6589251 DOI: 10.1155/2019/6854080] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/11/2019] [Indexed: 02/07/2023] Open
Abstract
Homoeostasis of bone marrow microenvironment depends on a precise balance between cell proliferation and death, which is supported by the cellular-extracellular matrix crosstalk. Multipotent mesenchymal stromal cells (MSC) are the key elements to provide the specialized bone marrow microenvironment by supporting, maintaining, and regulating the functions and fate of haematopoietic stem cells. Despite the great potential of MSC for cell therapy in several diseases due to their regenerative, immunomodulatory, and anti-inflammatory properties, they can also contribute to modulate tumor microenvironment. The extracellular vesicles that comprise exosomes and microvesicles are important mediators of intercellular communication due to their ability to change phenotype and physiology of different cell types. These vesicles may interact not only with neighbouring cells but also with cells from distant tissues to either maintain tissue homoeostasis or participate in disease pathogenesis. This review focuses on the current knowledge about the physiological role of MSC-extracellular vesicles, as well as their deregulation in haematological malignancies and their potential applications as biomarkers for diagnosis, progression, and treatment monitoring of such diseases.
Collapse
|
415
|
Mesenchymal stem cells immunomodulation: The road to IFN-γ licensing and the path ahead. Cytokine Growth Factor Rev 2019; 47:32-42. [DOI: 10.1016/j.cytogfr.2019.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
|
416
|
de Lima Santos A, Silva CGD, de Sá Barretto LS, Franciozi CEDS, Tamaoki MJS, de Almeida FG, Faloppa F. Biomechanical evaluation of tendon regeneration with adipose-derived stem cell. J Orthop Res 2019; 37:1281-1286. [PMID: 30474884 DOI: 10.1002/jor.24182] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/08/2018] [Indexed: 02/04/2023]
Abstract
The use of Adipose-Derived Stem Cells (ADSC) has been presented as a new alternative for tendon reconstruction. Have been admitted that ADSCs are related to better outcomes when used in tendon healing. This research was designed to apply the potential of ADSCs in tendon healing. Flexor digitorum superficialis tendon lesion was performed on both legs of eleven New Zealand rabbits and them, at the same time, treated as follows: Suture alone (Group III - Suture, n:10), suture associated with ADSC (Group IV - Suture + ADSC, n:10) or without suture (Group II - SHAN, n:2). At four weeks after the tendon surgery, the animal was euthanized, and the tendon evaluated (biomechanically and macroscopically). We used 5 additional New Zealand rabbits in the control group "Group I - Control, n:10". In the macroscopic evaluation, the group with ADSC presented a more homogeneous gross morphology compared with the group III. Biomechanical testing showed a lower ultimate tensile load, stiffness and a higher cross-sectional area in the group III and IV compared with the control group. The group with ADSC showed a greater ultimate tensile load, a larger cross-sectional area and bigger deformation at the ultimate tensile load when compared to the group without ADSC. In general terms, the use of ADSCs in tendon healing have biomechanical advantages compared to the non-use of ADSCs at 4 weeks after surgery. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1281-1286, 2019.
Collapse
Affiliation(s)
- Alex de Lima Santos
- Department of Orthopedic and Traumatology, Federal University of Sao Paulo, Brazil
| | - Camila Gonzaga da Silva
- Discipline of Urology, Department of Surgery, Federal University of São Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Flavio Faloppa
- Department of Orthopedic and Traumatology, Federal University of Sao Paulo, Brazil
| |
Collapse
|
417
|
Everts PA, Malanga GA, Paul RV, Rothenberg JB, Stephens N, Mautner KR. Assessing clinical implications and perspectives of the pathophysiological effects of erythrocytes and plasma free hemoglobin in autologous biologics for use in musculoskeletal regenerative medicine therapies. A review. Regen Ther 2019; 11:56-64. [PMID: 31193111 PMCID: PMC6517793 DOI: 10.1016/j.reth.2019.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/19/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023] Open
Abstract
Autologous biologics, defined as platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMC), are cell-based therapy treatment options in regenerative medicine practices, and have been increasingly used in orthopedics, sports medicine, and spinal disorders. These biological products are produced at point-of-care; thereby, avoiding expensive and cumbersome culturing and expansion techniques. Numerous commercial PRP and BMC systems are available but reports and knowledge of bio-cellular formulations produced by these systems are limited. This limited information hinders evaluating clinical and research outcomes and thus making conclusions about their biological effectiveness. Some of their important cellular and protein properties have not been characterized, which is critical for understanding the mechanisms of actions involved in tissue regenerative processes. The presence and role of red blood cells (RBCs) in any biologic has not been addressed extensively. Furthermore, some of the pathophysiological effects and phenomena related to RBCs have not been studied. A lack of a complete understanding of all of the biological components and their functional consequences hampers the development of clinical standards for any biological preparation. This paper aims to review the clinical implications and pathophysiological effects of RBCs in PRP and BMC; emphasizes hemolysis, eryptosis, and the release of macrophage inhibitory factor; and explains several effects on the microenvironment, such as inflammation, oxidative stress, vasoconstriction, and impaired cell metabolism. Different biological formulations optimize disease specific regenerative treatment protocols. Disintegrated RBC's release harmful components to regenerative therapy treatment vials. The effectiveness of MSC injection depends on the quality of the bone marrow aspiration procedure. PRP and BMC should contain minimal to no erythrocytes.
Collapse
Key Words
- BM-MSCs, bone marrow-mesenchymal cells
- BMA, bone marrow aspiration
- BMC, bone marrow concentrate
- Bone marrow mesenchymal cells
- Eryptosis
- HSCs, hematopoietic stem cells
- Hb, hemoglobin
- Hp, haptoglobin
- Hx, hemopexin
- Inflammation
- MIF, Macrophage migration inhibitory factor
- MNCs, mononucleated cells
- Macrophage migration inhibitor factor
- NO, nitric oxide
- OA, osteoarthritis
- Oxidative stress
- PAF, platelet activating factor
- PFH, plasma free hemoglobin
- PRP, platelet-rich plasma
- PS, phosphatidylserine
- Plasma free hemoglobin
- Platelet-rich plasma
- RBC, red blood cell
- ROS, reactive oxygen species
Collapse
Affiliation(s)
- Peter A. Everts
- Gulf Coast Biologics, Scientific and Research Department, Fort Myers, FL, USA
- Corresponding author. Gulf Coast Biologics, 6900 Daniels Pkwy, Suite #29-282, Fort Myers, FL 33912, USA.
| | - Gerard A. Malanga
- New Jersey Regenerative Institute LLC, Cedar Knolls, NJ, USA
- Department of Physical Medicine and Rehabilitation, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Rowan V. Paul
- California Pacific Orthopedics, San Francisco, CA, USA
- California Pacific Medical Center, San Francisco, CA, USA
- Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Joshua B. Rothenberg
- Boca Raton Regional Hospital, Regenerative Medicine and Orthopedics Biologic Department, Boca Raton, FL, USA
- BocaCare Orthopedics, Boca Raton, FL, USA
| | | | - Kenneth R. Mautner
- Emory University, Department of Physical Medicine & Rehabilitation, Atlanta GA, USA
- Emory University, Department of Orthopedics, Atlanta GA, USA
| |
Collapse
|
418
|
Najar M, Bouhtit F, Melki R, Afif H, Hamal A, Fahmi H, Merimi M, Lagneaux L. Mesenchymal Stromal Cell-Based Therapy: New Perspectives and Challenges. J Clin Med 2019; 8:626. [PMID: 31071990 PMCID: PMC6572531 DOI: 10.3390/jcm8050626] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cells have been the focus of intense research opening up new possibilities for the treatment of various diseases. Mesenchymal stromal cells (MSCs) are multipotent cells with relevant immunomodulatory properties and are thus considered as a promising new strategy for immune disease management. To enhance their efficiency, several issues related to both MSC biology and functions are needed to be identified and, most importantly, well clarified. The sources from which MSCs are isolated are diverse and might affect their properties. Both clinicians and scientists need to handle a phenotypic-characterized population of MSCs, particularly regarding their immunological profile. Moreover, it is now recognized that the tissue-reparative effects of MSCs are based on their immunomodulatory functions that are activated following a priming/licensing step. Thus, finding the best ways to pre-conditionate MSCs before their injection will strengthen their activity potential. Finally, soluble elements derived from MSC-secretome, including extracellular vesicles (EVs), have been proposed as a cell-free alternative tool for therapeutic medicine. Collectively, these features have to be considered and developed to ensure the efficiency and safety of MSC-based therapy. By participating to this Special Issue "Mesenchymal Stem/Stromal Cells in Immunity and Disease", your valuable contribution will certainly enrich the content and discussion related to the thematic of MSCs.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), and Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada.
- Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco.
| | - Fatima Bouhtit
- Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco.
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Bruxelles, Belgium.
| | - Rahma Melki
- Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco.
| | - Hassan Afif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), and Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada.
| | - Abdellah Hamal
- Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco.
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), and Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada.
| | - Makram Merimi
- Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco.
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Bruxelles, Belgium.
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Bruxelles, Belgium.
| |
Collapse
|
419
|
Wang J, Zhang Y, Cloud C, Duke T, Owczarski S, Mehrotra S, Adams DB, Morgan K, Gilkeson G, Wang H. Mesenchymal Stem Cells from Chronic Pancreatitis Patients Show Comparable Potency Compared to Cells from Healthy Donors. Stem Cells Transl Med 2019; 8:418-429. [PMID: 30680957 PMCID: PMC6477001 DOI: 10.1002/sctm.18-0093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are proven to be beneficial in islet transplantation, suggesting a potential therapeutic role of them in total pancreatectomy with islet autotransplantation (TP-IAT) for chronic pancreatitis (CP) patients. We investigated whether MSCs derived from CP patients are suitable for use in autologous cell therapy. MSCs from healthy donors (H-MSCs) and CP patients (CP-MSCs) were studied for phenotype, colony formation potential, multilineage differentiation ability, proliferation, senescence, secretory characters, and immunosuppressive functions. The potential protective effect of CP-MSCs was evaluated on hypoxia-induced islet cell death. Cell surface markers were similar between H-MSCs and CP-MSCs, as well as the ability of colony formation, multilineage differentiation, secretion of vascular endothelial growth factor and transforming growth factor (TGF-β), senescence, and inhibition of T cells proliferation in vitro. We found that growth differentiation factor 6 and hepatocyte growth factor (HGF) were significantly downregulated, whereas TGFβ and matrix metalloproteinase-2 were significantly upregulated in CP-MSCs compared with H-MSCs, among 84 MSC-related genes investigated in this study. MSCs from CP patients secreted less HGF, compared with the H-MSCs. A higher interferon-γ-induced indoleamine 2,3-dioxygenase expression was observed in CP-MSCs compared to H-MSCs. Moreover, CP-MSCs prevented hypoxia-induced β cell deaths to a similar extent as H-MSCs. Regardless of moderate difference in gene expression, CP-MSCs possess similar immunomodulatory and prosurvival functions to H-MSCs, and may be suitable for autologous cell therapy in CP patients undergoing TP-IAT. Stem Cells Translational Medicine 2019;8:418-429.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Yong Zhang
- College of Life ScienceQingdao Agricultural UniversityQingdaoPeople's Republic of China
| | - Colleen Cloud
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Tara Duke
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Stefanie Owczarski
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Shikhar Mehrotra
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - David B. Adams
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Katherine Morgan
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Gary Gilkeson
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Hongjun Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
420
|
Bernardini C, Bertocchi M, Zannoni A, Salaroli R, Tubon I, Dothel G, Fernandez M, Bacci ML, Calzà L, Forni M. Constitutive and LPS-stimulated secretome of porcine Vascular Wall-Mesenchymal Stem Cells exerts effects on in vitro endothelial angiogenesis. BMC Vet Res 2019; 15:123. [PMID: 31029157 PMCID: PMC6487069 DOI: 10.1186/s12917-019-1873-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
Abstract
Background MSCs secretome is under investigation as an alternative to whole-cell-based therapies, since it is enriched of bioactive molecules: growth factors, cytokines and chemokines. Taking into account the translational value of the pig model, the leading aim of the present paper was to characterize the secretome of porcine Vascular Wall–Mesenchymal Stem Cells (pVW-MSCs) and its change in presence of LPS stimulation. Moreover, considering the importance of angiogenesis in regenerative mechanisms, we analysed the effect of pVW-MSCs secretome on in vitro angiogenesis. Results Our results demonstrated that conditioned medium from unstimulated pVW-MSCs contained high levels of IL-8, GM-CSF, IFN-γ and other immunomodulatory proteins: IL-6 IL-18 IL-4 IL-2 IL-10. LPS modulates pVW-MSCs gene expression and secretome composition, in particular a significant increase of IL-6 and IL-8 was observed; conversely, the amount of GM-CSF, IFN-γ, IL-2, IL-4, IL-10 and IL-18 showed a significant transient decrease with the LPS stimulation. Conditioned medium from unstimulated pVW-MSCs induced in vitro endothelial angiogenesis, which is more evident when the conditioned medium was from LPS stimulated pVW-MSCs. Conclusions The lines of evidence here presented shed a light on possible future application of secretome derived by pVW-MSCs on research studies in translational regenerative medicine.
Collapse
Affiliation(s)
- Chiara Bernardini
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy.
| | - Martina Bertocchi
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Roberta Salaroli
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Irvin Tubon
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy.,Escuela de Enfermeria, Facultad de Ciencias Medicas, Universidad Regional Autónoma de Los Andes UNIANDES, Ambato, EC180150, Ecuador
| | - Giovanni Dothel
- Department of Medical and Surgical Sciences - DIMEC, University of Bologna, Bologna, Italy
| | - Mercedes Fernandez
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Laura Calzà
- Department of Pharmacy and Biotechnology - FaBiT, University of Bologna, Bologna, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| |
Collapse
|
421
|
Abstract
PURPOSE OF REVIEW Tissue engineering and regenerative medicine has emerged as a new scientific interdisciplinary field focusing on developing new strategies to repair or recreate tissues and organs. This review gathers findings on erectile dysfunction and, Peyronie's disease from recent preclinical and clinical studies under heading of stem-cell regenerative medicine. RECENT FINDINGS Over the last 2 years, preclinical studies on rat models demonstrated the tangible beneficial role of stem cells and stromal vascular fraction in the context of preventing fibrosis and restoring erectile function in different animal models of Erectile dysfunction and Peyronie's disease. There are not solid evidences in the clinical settings. SUMMARY Large randomized, double blind clinical trials are needed to prove the efficacy of stem-cell therapy on human patients. Owing to the lack of solid evidences, the stem-cell therapy should be only administrated in a clinical research setting.
Collapse
|
422
|
Zou J, Wang W, Kratz K, Xu X, Nie Y, Ma N, Lendlein A. Evaluation of human mesenchymal stem cell senescence, differentiation and secretion behavior cultured on polycarbonate cell culture inserts. Clin Hemorheol Microcirc 2019; 70:573-583. [PMID: 30372670 DOI: 10.3233/ch-189322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polycarbonate (PC) substrate is well suited for culturing human mesenchymal stem cells (MSCs) with high proliferation rate, low cell apoptosis rate and negligible cytotoxic effects. However, little is known about the influence of PC on MSC activity including senescence, differentiation and secretion. In this study, the PC cell culture insert was applied for human MSC culture and was compared with polystyrene (PS) and standard tissue culture plate (TCP). The results showed that MSCs were able to adhere on PC surface, exhibiting a spindle-shaped morphology. The size and distribution of focal adhesions of MSCs were similar on PC and TCP. The senescence level of MSCs on PC was comparable to that on TCP, but was significantly lower than that on PS. MSCs on PC were capable of self-renewal and differentiation into multiple cell lineages, including osteogenic and adipogenic lineages. MSCs cultured on PC secreted a higher level inflammatory cytokines and pro-angiogenic factors including FGF2 and VEGF. Conclusively, PC represents a promising cell culture material for human MSCs.
Collapse
|
423
|
Luque-Campos N, Contreras-López RA, Jose Paredes-Martínez M, Torres MJ, Bahraoui S, Wei M, Espinoza F, Djouad F, Elizondo-Vega RJ, Luz-Crawford P. Mesenchymal Stem Cells Improve Rheumatoid Arthritis Progression by Controlling Memory T Cell Response. Front Immunol 2019; 10:798. [PMID: 31040848 PMCID: PMC6477064 DOI: 10.3389/fimmu.2019.00798] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/26/2019] [Indexed: 12/14/2022] Open
Abstract
In the last years, mesenchymal stem cell (MSC)-based therapies have become an interesting therapeutic opportunity for the treatment of rheumatoid arthritis (RA) due to their capacity to potently modulate the immune response. RA is a chronic autoimmune inflammatory disorder with an incompletely understood etiology. However, it has been well described that peripheral tolerance defects and the subsequent abnormal infiltration and activation of diverse immune cells into the synovial membrane, are critical for RA development and progression. Moreover, the imbalance between the immune response of pro-inflammatory and anti-inflammatory cells, in particular between memory Th17 and memory regulatory T cells (Treg), respectively, is well admitted to be associated to RA immunopathogenesis. In this context, MSCs, which are able to alter the frequency and function of memory lymphocytes including Th17, follicular helper T (Tfh) cells and gamma delta (γδ) T cells while promoting Treg cell generation, have been proposed as a candidate of choice for RA cell therapy. Indeed, given the plasticity of memory CD4+ T cells, it is reasonable to think that MSCs will restore the balance between pro-inflammatory and anti-inflammatory memory T cells populations deregulated in RA leading to prompt their therapeutic function. In the present review, we will discuss the role of memory T cells implicated in RA pathogenesis and the beneficial effects exerted by MSCs on the phenotype and functions of these immune cells abnormally regulated in RA and how this regulation could impact RA progression.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Rafael A Contreras-López
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - María Jose Paredes-Martínez
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maria Jose Torres
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | | - Mingxing Wei
- Cellvax, SAS, Parc BIOCITECH, Romainville, France
| | | | | | - Roberto Javier Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
424
|
Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration. Acta Biomater 2019; 89:252-264. [PMID: 30878447 DOI: 10.1016/j.actbio.2019.03.021] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are potential therapeutics for the treatment of periodontal defects. It is increasingly accepted that MSCs mediate tissue repair through secretion of trophic factors, particularly exosomes. Here, we investigated the therapeutic effects of human MSC exosome-loaded collagen sponge for regeneration of surgically created periodontal intrabony defects in an immunocompetent rat model. We observed that relative to control rats, exosome-treated rats repaired the defects more efficiently with regeneration of periodontal tissues including newly-formed bone and periodontal ligament (PDL). We also observed that concomitant with this, there was increased cellular infiltration and proliferation. We therefore postulated that MSC exosomes enhanced regeneration through increased cellular mobilisation and proliferation. Using PDL cell cultures, we demonstrated that MSC exosomes could increase PDL cell migration and proliferation through CD73-mediated adenosine receptor activation of pro-survival AKT and ERK signalling. Inhibition of AKT or ERK phosphorylation suppressed PDL cell migration and proliferation. Our findings demonstrated for the first time that MSC exosomes enhance periodontal regeneration possibly by increasing PDL migration and proliferation. This study suggests that MSC exosome is a viable ready-to-use and cell-free MSC therapeutic for the treatment of periodontal defects. STATEMENT OF SIGNIFICANCE: Mesenchymal stem cell (MSC) therapies have demonstrated regenerative potential for the treatment of periodontal defects. However, translation of cellular therapies is hampered by challenges in maintaining optimal cell vitality and viability from manufacturing and storage to final delivery to patients. Although the use of MSCs for tissue repair was first predicated on their differentiation potential, the therapeutic efficacy of MSCs has increasingly been attributed to its paracrine secretion, particularly exosomes or small extracellular vesicles. In this study, MSC exosome-loaded collagen sponge enhanced periodontal regeneration in an immunocompetent rat periodontal defect model without any obvious adverse effects. These findings provide the basis for future development of MSC exosomes as a cell-free strategy for periodontal regeneration.
Collapse
|
425
|
Borkowska-Kuczkowska A, Sługocka D, Świątkowska-Flis B, Boruczkowski D. The use of mesenchymal stem cells for the treatment of progressive retinal diseases: a review. Regen Med 2019; 14:321-329. [PMID: 30977436 DOI: 10.2217/rme-2019-0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Some ocular diseases, such as dystrophies, retinal and macular degeneration, optic nerve atrophy, and Stargardt disease, are progressive and irreversible. In this review, we focus on the use of mesenchymal stem cells (MSCs) in the treatment of these diseases. In animal studies, MSC transplantation significantly delayed retinal degeneration, led to the regeneration of cone cells, and supported the survival of retinal ganglion cells and axon regeneration. In clinical practice, patients with Behcet's disease with retinal vasculitis who received MSC injections experienced a decrease in retinal vasculitis but no improvement in vision acuity. Nonetheless, there is no evidence that MSCs are carcinogenic, and they even reduce the size of tumors in vitro. Furthermore, MSCs do not trigger the immune response.
Collapse
Affiliation(s)
- Agnieszka Borkowska-Kuczkowska
- Polish Center of Cell Therapy & Immunotherapy in Częstochowa, Waly Dwernickiego 43/45, 42-202 Częstochowa, Poland.,Agamed Center of Ophthalmology, Jasnogórska 4, 42-202 Częstochowa, Poland
| | - Dominika Sługocka
- Polish Center of Cell Therapy & Immunotherapy in Częstochowa, Waly Dwernickiego 43/45, 42-202 Częstochowa, Poland
| | - Beata Świątkowska-Flis
- Polish Center of Cell Therapy & Immunotherapy in Częstochowa, Waly Dwernickiego 43/45, 42-202 Częstochowa, Poland
| | - Dariusz Boruczkowski
- Polski Bank Komórek Macierzystych SA (FamiCord Group), Jana Pawła II 29, 00-867 Warsaw, Poland
| |
Collapse
|
426
|
Selim RE, Ahmed HH, Abd-Allah SH, Sabry GM, Hassan RE, Khalil WKB, Abouhashem NS. Mesenchymal Stem Cells: a Promising Therapeutic Tool for Acute Kidney Injury. Appl Biochem Biotechnol 2019; 189:284-304. [PMID: 30976980 DOI: 10.1007/s12010-019-02995-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is a rapid loss of renal function. It has high mortality rates. Still, renal replacement therapy is considered the best solution for recovering AKI. This opens a line of thought to develop an alternative therapy for it without complications. Mesenchymal stem cells are considered a new therapy for treating kidney diseases. The aim of this work was to address the anti-apoptotic, antioxidative, and pro-angiogenic effects of adipose tissue-derived MSCs (AD-MSCs) and bone marrow-MSCs (BM-MSCs) for treating AKI. Adult male Wistar rats were assigned into nine groups (n = 10): (1) the control group; (2) the AKI group, receiving cisplatin; (3) the AKI group treated with AD-MSCs (1 × 106); (4) the AKI group treated with AD-MSCs (2 × 106); (5) the AKI group treated with AD-MSCs (4 × 106); (6) the AKI group treated with losartan; (7) the AKI group treated with BM-MSCs (1 × 106); (8) the AKI group treated with BM-MSCs (2 × 106); and (9) the AKI group treated with BM-MSCs (4 × 106). The results showed a significant rise in creatinine, urea, and cystatin C (cys C) levels and upregulation of p38 mRNA, whereas a significant decline in NAD(P)H quinone oxidoreductase 1 (NQO-1) protein and downregulation of B-cell lymphoma-2 (Bcl-2) mRNA and vascular endothelial growth factor (VEGF) mRNA were recorded in AKI. MSCs could improve renal functions manifested by decreased urea, creatinine, and cys C levels; downregulation of p38; and upregulation of Bcl-2 and VEGF. Moreover, MSC therapy could induce NQO-1 in the treated rats relative to the untreated rats. So, cell-based therapy can reduce AKI through the antioxidative, anti-apoptotic, and pro-angiogenic properties of MSCs. Therefore, the findings received in this attempt create a fertile base for the setup of cell therapy in patients with AKI.
Collapse
Affiliation(s)
- Rehab E Selim
- Hormones Department, National Research Centre, Giza, Egypt. .,Stem Cell Lab., Centre of Excellence for Advanced Science, National Research Centre, Dokki, Giza, Egypt.
| | - Hanaa H Ahmed
- Hormones Department, National Research Centre, Giza, Egypt.,Stem Cell Lab., Centre of Excellence for Advanced Science, National Research Centre, Dokki, Giza, Egypt
| | - Somia H Abd-Allah
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Gilane M Sabry
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Rasha E Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Nehal S Abouhashem
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
427
|
Abstract
INTRODUCTION Aberrant wound healing is a significant healthcare problem, posing a substantial burden on patients, their families, and the healthcare system. Existing treatment options remain only moderately effective and often fail to promote the closure of non-healing wounds in susceptible populations, such as aging and diabetic patients. Stem cell therapy has emerged as a promising treatment modality, with the potential to restore tissue to its pre-injured state. Of particular interest are mesenchymal stromal cells, which have been shown to accelerate wound healing by modulating the immune response and promoting angiogenesis. AREAS COVERED This review provides an overview of wound healing and current methods for the management of chronic wounds, as well as the current state and considerations for optimizing stem cell therapy. Considerations include stem cell types, tissue source, donor selection, cell heterogeneity, delivery methods, and genetic engineering. EXPERT OPINION A growing body of evidence has shown that delivery of stem cells, particularly mesenchymal stromal cells, has the potential to effectively improve the rate and quality of wound healing. However, significant additional basic and clinical research must be performed to optimize cell therapy, such as further elucidation of the therapeutic mechanisms of stem cells and standardization of clinical trial guidelines.
Collapse
Affiliation(s)
- Nina Kosaric
- a Hagey Laboratory for Pediatric Regenerative Medicine; Division of Plastic and Reconstructive Surgery, Department of Surgery , Stanford University School of Medicine , Stanford , CA , USA
| | - Harriet Kiwanuka
- a Hagey Laboratory for Pediatric Regenerative Medicine; Division of Plastic and Reconstructive Surgery, Department of Surgery , Stanford University School of Medicine , Stanford , CA , USA
| | - Geoffrey C Gurtner
- a Hagey Laboratory for Pediatric Regenerative Medicine; Division of Plastic and Reconstructive Surgery, Department of Surgery , Stanford University School of Medicine , Stanford , CA , USA
| |
Collapse
|
428
|
Harvestine JN, Sheaff CS, Li C, Haudenschild AK, Gionet-Gonzales MA, Hu JC, Athanasiou KA, Marcu L, Leach JK. Multimodal Label-Free Imaging for Detecting Maturation of Engineered Osteogenic Grafts. ACS Biomater Sci Eng 2019; 5:1956-1966. [PMID: 33405522 PMCID: PMC8594456 DOI: 10.1021/acsbiomaterials.9b00007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There is a critical need to develop noninvasive, nondestructive methods for assessing the quality of engineered constructs prior to implantation. Currently, the composition and maturity of engineered tissues are assessed using destructive, costly, and time-consuming biochemical and mechanical analyses. The goal of this study was to use noninvasive, multimodal imaging to monitor osteogenic differentiation and matrix deposition by human mesenchymal stem/stromal cells (MSCs) during in vitro culture. MSCs were encapsulated in alginate hydrogels and cultured in osteogenic conditions for 4 weeks. Samples were evaluated using fluorescence lifetime imaging (FLIm) and ultrasound backscatter microscopy (UBM) prior to traditional biochemical and mechanical testing. Using linear regression analysis, we identified strong correlations between imaging parameters (e.g., fluorescence lifetime and acoustic attenuation coefficient) and destructive mechanical and biochemical tests to assess the maturation of osteogenically induced constructs. These data demonstrate the promise of nondestructive label-free imaging techniques to noninvasively ascertain the progression and maturity of tissue engineered bone grafts.
Collapse
Affiliation(s)
- Jenna N. Harvestine
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Clay S. Sheaff
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Cai Li
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Anne K. Haudenschild
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Marissa A. Gionet-Gonzales
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine California 92697, United States
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine California 92697, United States
| | - Laura Marcu
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, California 95817, United States
| |
Collapse
|
429
|
Zappa Villar MF, Lehmann M, García MG, Mazzolini G, Morel GR, Cónsole GM, Podhajcer O, Reggiani PC, Goya RG. Mesenchymal stem cell therapy improves spatial memory and hippocampal structure in aging rats. Behav Brain Res 2019; 374:111887. [PMID: 30951751 DOI: 10.1016/j.bbr.2019.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
There is a growing interest in the potential of mesenchymal stem cells (MSCs) for implementing regenerative medicine in the brain as they have shown neurogenic and immunomodulatory activities. We assessed the effect of intracerebroventricular (icv) administration of human bone marrow-derived MSCs (hBM-MSCs) on spatial memory and hippocampal morphology of senile (27 months) female rats, using 3-months-old counterparts as young controls. Half of the animals were injected in the lateral ventricles (LV) with a suspension containing 5 × 105hBM-MSCs in 8 μl per side. The other half received no treatment (senile controls). Spatial memory performance was assessed with a modified version of the Barnes maze test. We employed one probe trial, one day after training in order to evaluate learning ability as well as spatial memory retention. Neuroblast (DCX) and microglial (Iba-1 immunoreactive) markers were also immunohistochemically quantitated in the animals by means of an unbiased stereological approach. In addition, hippocampal presynaptic protein expression was assessed by immunoblotting analysis. After treatment, the senile MSC-treated group showed a significant improvement in spatial memory accuracy and extended permanence in a one- and 3-hole goal sectors as compared with senile controls. The MSC treatment increased the number of neuroblasts in the hippocampal dentate gyrus, reduced the number of reactive microglial cells, and restored presynaptic protein levels as compared to senile controls. We conclude that icv injected hBM-MSCs are effective in improving spatial memory in senile rats and that the strategy improves some functional and morphologic brain features typically altered in aging rats.
Collapse
Affiliation(s)
- Maria F Zappa Villar
- INIBIOLP-Pathology B, School of Medicine, National University of La Plata, Argentina; Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina
| | - Marianne Lehmann
- INIBIOLP-Pathology B, School of Medicine, National University of La Plata, Argentina; Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina
| | - Mariana G García
- Gene Therapy Laboratory, IIMT, School of Medical Science, Austral University, Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, IIMT, School of Medical Science, Austral University, Buenos Aires, Argentina
| | - Gustavo R Morel
- INIBIOLP-Pathology B, School of Medicine, National University of La Plata, Argentina; Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina
| | - Gloria M Cónsole
- Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina
| | - Osvaldo Podhajcer
- Laboratory of Molecular and Cellular Therapy, Fundacion Instituto Leloir, Buenos Aires, Argentina
| | - Paula C Reggiani
- INIBIOLP-Pathology B, School of Medicine, National University of La Plata, Argentina; Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina
| | - Rodolfo G Goya
- INIBIOLP-Pathology B, School of Medicine, National University of La Plata, Argentina; Department of Histology and of Embryology B, School of Medicine, National University of La Plata, Argentina.
| |
Collapse
|
430
|
Born S, Dörfel MJ, Hartjen P, Haschemi Yekani SA, Luecke J, Meutsch JK, Westphal JK, Birkelbach M, Köhnke R, Smeets R, Krueger M. A short-term plastic adherence incubation of the stromal vascular fraction leads to a predictable GMP-compliant cell-product. ACTA ACUST UNITED AC 2019; 9:161-172. [PMID: 31508331 PMCID: PMC6726751 DOI: 10.15171/bi.2019.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
Abstract
![]()
Introduction:Mesenchymal stromal/stem cells (MSCs) derived from fat tissue are an encouraging tool for regenerative medicine. They share properties similar to the bone marrow-derived MSCs, but the amount of MSCs per gram of fat tissue is 500x higher. The fat tissue can easily be digested by collagenase, releasing a heterogeneous cell fraction called stromal vascular fraction (SVF) which contains a variable amount of stromal/stem cells. In Europe, cell products like the SVF derived from fat tissue are considered advanced therapy medicinal product (ATMPs). As a consequence, the manufacturing process has to be approved via GMP-compliant process validation. The problem of the process validation for SVF is the heterogeneity of this fraction.
Methods: Here, we modified existing purification strategies by adding an additional plastic adherence incubation of maximal 20 hours after SVF isolation. The resulting cell fraction was characterized and compared to SVF as well as cultivated adipose-derived stromal/stem cells (ASCs) with respect to viability and cell yield, the expression of surface markers, differentiation potential and cytokine expression.
Results: Short-term incubation significantly reduced the heterogeneity of the resulting cell fraction compared to SVF. The cells were able to differentiate into adipocytes, chondrocytes, and osteoblasts. More importantly, they expressed trophic proteins which have been previously associated with the beneficial effects of MSCs. Furthermore, GMP compliance of the production process described herein was acknowledged by the national regulatory agencies (DE_BB_01_GMP_2017_1018).
Conclusion: Addition of a short purification-step after the SVF isolation is a cheap and fast strategy to isolate a homogeneous uncultivated GMP-compliant cell fraction of ASCs.
Collapse
Affiliation(s)
| | | | - Philip Hartjen
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | - Moritz Birkelbach
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Köhnke
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany.,Division, Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
431
|
Chen X, Sun J, Li X, Mao L, Cui L, Bai W. Transplantation of oral mucosal epithelial cells seeded on decellularized and lyophilized amniotic membrane for the regeneration of injured endometrium. Stem Cell Res Ther 2019; 10:107. [PMID: 30898158 PMCID: PMC6429789 DOI: 10.1186/s13287-019-1179-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
Background Intrauterine adhesion (IUA) is characterized by progressive intrauterine fibrosis as a consequence of trauma to the basal layer of the endometrium. In an attempt to relieve IUA, many approaches have been applied in the clinic but show limited effects. In this study, we investigated the effect of autologous oral mucosal epithelial cells (OMECs) seeded on decellularized and lyophilized amniotic membrane (DL-AM) on preventing the development of IUA in a rat model. Methods IUA model was established by surgical scraping of the endometrium in the left uteri (the right uteri were kept as control) of SD rats. Wounds were randomly treated as unrepaired (IUA group), repaired with DL-AM alone (DL-AM group), and DL-AM seeded with autologous OMECs (DL-AM+OMECs group), respectively, in a total of 54 rats (n = 18 each). Uterus samples were harvested for histological and immunohistochemical evaluation after 3, 7, 14, and 28 days (n = 3 in each time point) of operations. Results After surgery, the uterine cavity was observed to be filled with extensive fibrosis in the IUA and DL-AM groups, respectively, while a lower ratio of the fibrotic area was identified in the DL-AM transplantation group. Transplantation of OMECs seeded on DL-AM significantly reduced fibrosis of IUA with recovered uterine cavity and regenerated epithelium and endometrial glands as determined by CK-18 immunostaining. OMECs transplantation resulted in extensive cellular proliferation as revealed by the Ki-67 immunofluorescent staining exhibited. Meanwhile, microvessel density was significantly increased in uteri that received OMECs transplantation, which was concomitant with elevated expression of vascular endothelial growth factor. The pregnancy test (n = 6 in each group) showed successful conception in the OMEC-transplanted uteri, but not in the IUA and DL-AM groups. Conclusions Engineered epithelium developed from DL-AM seeded with OMECs showed great potential in preventing progression of intrauterine adhesion by improved endometrial epithelium regeneration. Electronic supplementary material The online version of this article (10.1186/s13287-019-1179-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xing Chen
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jingtao Sun
- Department of Plastic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaoyu Li
- Department of Plastic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Lele Mao
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Lei Cui
- Department of Plastic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
432
|
Muniswami DM, Reddy LVK, Venkatesh K, Babu S, Sen D. Neuropotency and Neurotherapeutic Potential of Human Umbilical Cord Stem Cell’s Secretome. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00096-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
433
|
Pilny E, Smolarczyk R, Jarosz-Biej M, Hadyk A, Skorupa A, Ciszek M, Krakowczyk Ł, Kułach N, Gillner D, Sokół M, Szala S, Cichoń T. Human ADSC xenograft through IL-6 secretion activates M2 macrophages responsible for the repair of damaged muscle tissue. Stem Cell Res Ther 2019; 10:93. [PMID: 30867059 PMCID: PMC6417195 DOI: 10.1186/s13287-019-1188-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/25/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (ADSCs) are multipotent stromal cells. The cells secrete a number of cytokines and growth factors and show immunoregulatory and proangiogenic properties. Their properties may be used to repair damaged tissues. The aim of our work is to explain the muscle damage repair mechanism with the utilization of the human adipose-derived mesenchymal stromal cells (hADSCs). METHODS For the hADSCs isolation, we used the subcutaneous adipose tissue collected during the surgery. The murine hind limb ischemia was used as a model. The unilateral femoral artery ligation was performed on 10-12-week-old male C57BL/6NCrl and NOD SCID mice. The mice received PBS- (controls) or 1 × 106 hADSCs. One, 3, 7, 14 and 21 days after the surgery, we collected the gastrocnemius muscles for the immunohistochemical analysis. The results were analyzed with relevant tests using the Statistica software. RESULTS The retention time of hADSCs in the limb lasted about 14 days. In the mice receiving hADSCs, the improvement in the functionality of the damaged limb occurred faster than in the control mice. More new blood vessels were formed in the limbs of the mice receiving hADSCs than in limbs of the control mice. hADSCs also increased the infiltration of the macrophages with the M2 phenotype (7-AAD-/CD45+/F4/80+/CD206+) into the ischemic limbs. hADSCs introduced into the limb of mice secreted interleukin-6. This cytokine stimulates the emergence of the proangiogenic M2 macrophages, involved, among others, in the repair of a damaged tissue. Both macrophage depletion and IL-6 blockage suppressed the therapeutic effect of hADSCs. In the mice treated with hADSCs and liposomes with clodronate (macrophages depletion), the number of capillaries formed was lower than in the mice treated with hADSCs alone. Administration of hADSCs to the mice that received siltuximab (human IL-6 blocker) did not cause an influx of the M2 macrophages, and the number of capillaries formed was at the level of the control group, as in contrast to the mice that received only the hADSCs. CONCLUSIONS The proposed mechanism for the repair of the damaged muscle using hADSCs is based on the activity of IL-6. In our opinion, the cytokine, secreted by the hADSCs, stimulates the M2 macrophages responsible for repairing damaged muscle and forming new blood vessels.
Collapse
Affiliation(s)
- Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland.,Department of Organic Chemistry, Biochemistry and Biotechnology, Silesian University of Technology, Księdza Marcina Strzody 9 Street, 44-100, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Alina Hadyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Agnieszka Skorupa
- Department of Medical Physics Maria Sklodowska-Curie Institute -Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Mateusz Ciszek
- Department of Medical Physics Maria Sklodowska-Curie Institute -Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Łukasz Krakowczyk
- Department of Oncologic and Reconstructive Surgery, Maria Sklodowska-Curie Institute -Oncology Center, Wybrzeże Armii Krajowej 15 Street, 44-101 Gliwice Branch, Gliwice, Poland
| | - Natalia Kułach
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland.,Department of Animal Physiology and Ecotoxicology, Faculty of Biology and Environmental Protection, University of Silesia, Bankowa 12 Street, 40-007, Katowice, Poland
| | - Danuta Gillner
- Department of Organic Chemistry, Biochemistry and Biotechnology, Silesian University of Technology, Księdza Marcina Strzody 9 Street, 44-100, Gliwice, Poland
| | - Maria Sokół
- Department of Medical Physics Maria Sklodowska-Curie Institute -Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Stanisław Szala
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland.
| |
Collapse
|
434
|
Davis MS, Marrero-Berrios I, Perez I, Rabolli CP, Radhakrishnan P, Manchikalapati D, Schianodicola J, Kamath H, Schloss RS, Yarmush J. Alginate encapsulation for bupivacaine delivery and mesenchymal stromal cell immunomodulatory cotherapy. J Inflamm Res 2019; 12:87-97. [PMID: 30881083 PMCID: PMC6419600 DOI: 10.2147/jir.s192749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Mesenchymal stromal cells (MSCs) are used to treat various inflammatory conditions. In parallel, to mitigate pain associated with inflammation, analgesics or opioids are prescribed, often with significant side effects. Local anesthetics (LAs) offer a promising alternative to these medications. However, their short duration and negative effects on anti-inflammatory MSCs have limited their therapeutic effectiveness. To mitigate these negative effects and to move toward developing a cotherapy, we engineered a sustained release bupivacaine alginate-liposomal construct that enables up to 4 days of LA release. By encapsulating MSC in alginate (eMSC), we demonstrate that we can further increase drug concentration to clinically relevant levels, without compromising eMSC viability or anti-inflammatory function. MATERIALS AND METHODS MSCs were freely cultured or encapsulated in alginate microspheres ± TNFα/IFN-γ and were left untreated or dosed with bolus, liposomal, or construct bupivacaine. After 24, 48, and 96 hours, the profiles were assessed to quantify secretory function associated with LA-MSC interactions. To approximate LA exposure over time, a MATLAB model was generated. RESULTS eMSCs secrete similar levels of IL-6 and prostaglandin E2 (PGE2) regardless of LA modality, whereas free MSCs secrete larger amounts of IL-6 and lower amounts of anti-inflammatory PGE2. Modeling the system indicated that higher doses of LA can be used in conjunction with eMSC while retaining eMSC viability and function. In general, eMSC treated with higher doses of LA secreted similar or higher levels of immunomodulatory cytokines. CONCLUSION eMSCs, but not free MSC, are protected from LA, regardless of LA modality. Increasing the LA concentration may promote longer and stronger pain mitigation while the protected eMSCs secrete similar, if not higher, immunomodulatory cytokine levels. Therefore, we have developed an approach, using eMSC and the LA construct that can potentially be used to reduce pain as well as improve MSC anti-inflammatory function.
Collapse
Affiliation(s)
- Mollie S Davis
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA,
| | | | - Isabel Perez
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA,
| | - Charles P Rabolli
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA,
| | | | | | | | | | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA,
| | - Joel Yarmush
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, NY, USA
| |
Collapse
|
435
|
Bie YN, Gu P, Chen YT, Zhou XX, Tian YG, Yang Q, Li HY, Lin X, Guan YH, Lin TY, Lu X, Shen HF, Fang TX, Liu YM, Xiao D, Gu WW. TZAP plays an inhibitory role in the self-renewal of porcine mesenchymal stromal cells and is implicated the regulation of premature senescence via the p53 pathway. J Transl Med 2019; 17:72. [PMID: 30845965 PMCID: PMC6404308 DOI: 10.1186/s12967-019-1820-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 02/21/2019] [Indexed: 01/01/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) were originally characterized by the ability to differentiate into different mesenchymal lineages in vitro, and their immunomodulatory and trophic functions have recently aroused significant interest in the application of MSCs in cell-based regenerative medicine. However, a major problem in clinical practice is the replicative senescence of MSCs, which limits the cell proliferation potential of MSCs after large-scale expansion. Telomeric zinc finger-associated protein (TZAP), a novel specific telomere-binding protein, was recently found to stimulate telomere trimming and prevent excessive telomere elongation. The aim of this study was to elucidate the role of TZAP in regulating MSCs senescence, differentiation and proliferation. Method Primary porcine mesenchymal stromal cells (pMSCs) were isolated from the bone marrow of Tibet minipigs by a noninvasive method in combination with frequent medium changes (FMCs). The deterioration of the pMSCs’ proliferation capacity and their resultant entry into senescence were analyzed by using CCK8 and EdU incorporation assays, SA-β-gal staining and comparisons of the expression levels of cellular senescence markers (p16INK14 and p21) in pMSC cell lines with TZAP overexpression or knockout. The effects of TZAP overexpression or knockout on the differentiation potential of pMSCs were assessed by alizarin red S staining after osteogenic induction or by oil red O staining after adipogenic induction. The effect of TZAP overexpression and the involvement of the p53 signaling pathway were evaluated by detecting changes in ARF, MDM2, P53 and P21 protein levels in pMSCs. Results TZAP levels were significantly elevated in late-passage pMSCs compared to those in early-passage pMSCs. We also observed significantly increased levels of the senescence markers p16INK4A and p21. Overexpression of TZAP reduced the differentiation potential of the cells, leading to premature senescence in early-passage pMSCs, while knockout of TZAP led to the opposite phenotype in late-passage pMSCs. Furthermore, overexpression of TZAP activated the P53 pathway (ARF-MDM2-P53-P21WAF/CDKN1A) in vitro. TZAP also downregulated the expression levels of PPARγ and Cebpα, two key modulators of adipogenesis. Conclusions This study demonstrates that the level of TZAP is closely related to differentiation potential in pMSCs and affects cellular senescence outcomes via the p53 pathway. Therefore, attenuation of intracellular TZAP levels could be a new strategy for improving the efficiency of pMSCs in cell therapy and tissue engineering applications. Electronic supplementary material The online version of this article (10.1186/s12967-019-1820-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ya-Nan Bie
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Peng Gu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Yu-Ting Chen
- Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiao-Xu Zhou
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China.,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China
| | - Yu-Guang Tian
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Qin Yang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-Yan Li
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Xia Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Yan-Hong Guan
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Tao-Yan Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Xun Lu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Hong-Fen Shen
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Ting-Xiao Fang
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Yu-Min Liu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China
| | - Dong Xiao
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University, Guangzhou, China.
| | - Wei-Wang Gu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou, China. .,Songshan Lake Pearl Laboratory Animal Sci & Tech. Co., Ltd., Dongguan, China.
| |
Collapse
|
436
|
Castiglione F, Hedlund P, Weyne E, Hakim L, Montorsi F, Bivalacqua TJ, De Ridder D, Milenkovic U, Ralph D, Garaffa G, Muneer A, Joniau S, Albersen M. Intratunical Injection of Human Adipose Tissue-Derived Stem Cells Restores Collagen III/I Ratio in a Rat Model of Chronic Peyronie's Disease. Sex Med 2019; 7:94-103. [PMID: 30503767 PMCID: PMC6377372 DOI: 10.1016/j.esxm.2018.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/09/2018] [Accepted: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Previous studies have shown that the injection of adipose tissue-derived stem cells (ADSCs) into the tunica albuginea (TA) during the active phase of Peyronie's disease (PD) prevents the development of fibrosis. AIM To investigate, using an animal model, whether local injection of human ADSCs (hADSCs) can alter the degree of fibrosis in the chronic phase of PD. METHODS 27 male, 12-week-old rats were divided into 3 equal groups: sham, PD without treatment, and PD treated with hADSCs 1 month after disease induction. Sham rats underwent 2 injections of vehicle into the TA 1 month apart. PD rats underwent transforming growth factor β1 (TGFβ1) injection and injection of vehicle 1 month later. PD-hADSC rats underwent TGFβ1 injection followed by 1 million hADSCs 1 month later. 1 week after treatment, n = 3 animals/group were euthanized, and the penises were harvested for quantitative polymerase chain reaction. 1 month after treatment, the other animals, n = 6 per group, underwent measurement of intracavernous pressure (ICP) and mean arterial pressure (MAP) during electrostimulation of the cavernous nerve. After euthanasia, penises were again harvested for histology and Western blot. MAIN OUTCOME MEASURE The primary outcome measures included (a) gene expression at one week post-injection; (b) measurement of ICP/MAP upon cavernous nerve stimulation as a measure of erectile function; (c) elastin, collagen I and III protein expression; and (d) Histomorphometric analysis of the penis. Means where compared by analysis of variance (ANOVA) followed by a Student-Newman-Keuls test for post hoc comparisons or Mann-Whitney test when applicable. RESULTS No significant difference was noted in ICP or ICP/MAP in response to cavernous nerve electrostimulation between the 3 groups at 2.5, 5, and 7.5 V (P > .05 for all voltages). PD animals developed tunical and subtunical areas of fibrosis with a significant upregulation of collagen III protein. The collagen III/I ratio was higher in the PD (4.6 ± 0.92) group compared with sham (0.66 ± 0.18) and PD-hADSC (0.86 ± 0.06) groups (P < .05) These fibrotic changes were prevented when treated with hADSCs. Compared with PD rats, PD-hADSC rats demonstrated a decreased expression of several fibrosis-related genes. CONCLUSION Injection of hADSCs reduces collagen III expression in a rat model of chronic PD. Castiglione F, Hedlund P, Weyne E, et al. Intratunical Injection of Human Adipose Tissue-Derived Stem Cells Restores Collagen III/I Ratio in a Rat Model of Chronic Peyronie's Disease. Sex Med 2019;7:94-103.
Collapse
Affiliation(s)
- Fabio Castiglione
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium; The Institute of Urology, University College of London Hospital, London, UK; Division of Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Petter Hedlund
- Department of Clinical and Experimental Pharmacology, Lund University, Sweden; Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Sweden.
| | - Emanuel Weyne
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Lukman Hakim
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium; Department of Urology, Airlangga University/Dr Soetomo General Hospital, Surabaya, Indonesia
| | - Francesco Montorsi
- Division of Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Trinity J Bivalacqua
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Dirk De Ridder
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Uros Milenkovic
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - David Ralph
- The Institute of Urology, University College of London Hospital, London, UK
| | - Giulio Garaffa
- The Institute of Urology, University College of London Hospital, London, UK
| | - Asif Muneer
- The Institute of Urology, University College of London Hospital, London, UK; Division of Surgery and Interventional Science, National Institute for Health Research Biomedical Research Centre, University College London Hospital, London, UK
| | - Steven Joniau
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Maarten Albersen
- Laboratory for Experimental Urology, Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| |
Collapse
|
437
|
Zhang C, Guo H, Yang C, Chen Q, Huang J, Liu L, Zhang Y, Jin S, Song A, Yang P. The biological behavior optimization of human periodontal ligament stem cells via preconditioning by the combined application of fibroblast growth factor-2 and A83-01 in in vitro culture expansion. J Transl Med 2019; 17:66. [PMID: 30819199 PMCID: PMC6396448 DOI: 10.1186/s12967-019-1799-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND As the optimal source of seed cells in periodontal tissue engineering, periodontal ligament stem cells (PDLSCs) have always been researched to improve cell expansion due to their limited resource and spontaneous differentiation in vitro cultivation. Fibroblast growth factor-2 (FGF-2) has been proven to stimulate bone marrow mesenchymal stem cells (BMMSCs) proliferation and maintain their pluripotency when being added to the culture medium. As a small molecule inhibitor of transforming growth factor-beta receptors (TGF-βRs), A83-01 can also promote cell proliferation. Therefore, the aim of this study was to verify whether the combined application of FGF-2 and A83-01 could augment cell quantity and quality during in vitro culture. METHODS PDLSCs were preconditioned with A83-01, FGF-2, or their combination. A cell counting kit-8 (CCK8) assay, cell apoptosis assay, ALP activity assay, Alizarin Red S staining assay, RT-PCR assay, Western blot assay and ELISA were used to determine the sustained effects of different preconditioning strategies on the proliferation, apoptosis, stemness, osteogenic differentiation and paracrine action of PDLSCs. RESULTS The combined application of FGF-2 and A83-01 significantly augmented cell expansion, reduced cell apoptosis, magnified stemness expression, promoted later osteogenic differentiation and mineralization and increased paracrine action of PDLSCs compared with the control. Moreover, the combination presented significant advantages in enhancing proliferation, stemness expression and paracrine action over FGF-2 alone. CONCLUSIONS The combined application of A83-01 and FGF-2 may be an improved strategy for PDLSCs biological behavior optimization in culture expansion and advantageous for reinforcing proliferation, stemness expression and cytokine secretion over FGF-2 alone.
Collapse
Affiliation(s)
- Chunshu Zhang
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, China
| | - Hongmei Guo
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital and Institute of Stomatology, Shandong University, Jinan, 250012 Shandong People’s Republic of China
| | - Qian Chen
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Jiahui Huang
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Lianlian Liu
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Yu Zhang
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Shanshan Jin
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
| | - Aimei Song
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, China
| | - Pishan Yang
- Department of Periodontology, School of Stomatology, Shandong University, 44 West Wenhua Road, Jinan, 250012 Shandong People’s Republic of China
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
438
|
Yao X, Jing X, Guo J, Sun K, Deng Y, Zhang Y, Guo F, Ye Y. Icariin Protects Bone Marrow Mesenchymal Stem Cells Against Iron Overload Induced Dysfunction Through Mitochondrial Fusion and Fission, PI3K/AKT/mTOR and MAPK Pathways. Front Pharmacol 2019; 10:163. [PMID: 30873034 PMCID: PMC6403125 DOI: 10.3389/fphar.2019.00163] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/11/2019] [Indexed: 12/26/2022] Open
Abstract
Iron overload has been reported to contribute to bone marrow mesenchymal stem cells (BMSCs) damage, but the precise mechanism still remains elusive. Icariin, a major bioactive monomer belonging to flavonoid glucosides isolated from Herba Epimedii, has been shown to protect cells from oxidative stress induced apoptosis. The aim of this study was to investigate whether icariin protected against iron overload induced dysfunction of BMSCs and its underlying mechanism. In this study, we found that iron overload induced by 100 μM ferric ammonium citrate (FAC) caused apoptosis of BMSCs, promoted cleaved caspase-3 and BAX protein expressions while inhibited Bcl-2 protein expression, which effects were significantly attenuated by icariin treatment. In addition, iron overload induced significant depolarization of mitochondrial membrane potential (MMP), reactive oxygen species (ROS) generation and inhibition of mitochondrial fusion/fission, which effects were also attenuated by icariin treatment. Meanwhile, we found that iron overload induced by 100 μM FAC significantly inhibited mitochondrial fission protein FIS1 and fusion protein MFN2 expressions, inhibited DRP1 and Cytochrome C protein translocation from the cytoplasm to mitochondria. Icariin at concentration of 1 μM was able to promote mitochondrial fission protein FIS1 and fusion protein MFN2 expressions, and increase DRP1 and cytochrome C protein translocation from the cytoplasm to mitochondria. Further, osteogenic differentiation and proliferation of BMSCs was significantly inhibited by iron overload, but icariin treatment rescued both osteogenic differentiation and proliferation of BMSCs. Further studies showed that icariin attenuated iron overload induced inactivation of the PI3K/AKT/mTOR pathway and activation of the ERK1/2 and JNK pathways. In summary, our study indicated that icariin was able to protect against iron overload induced dysfunction of BMSCs. These effects were potentially related to the modulation of mitochondrial fusion and fission, activation of the PI3K/AKT/mTOR pathway and inhibition of ERK1/2 and JNK pathways.
Collapse
Affiliation(s)
- Xudong Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiachao Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Deng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
439
|
Zhang W, Liu C, Wu D, Liang C, Zhang L, Zhang Q, Liu Y, Xia M, Wang H, Su P, Feng S, Han M, Zhou J, Wang F, Jiang E. Decitabine improves platelet recovery by down-regulating IL-8 level in MDS/AML patients with thrombocytopenia. Blood Cells Mol Dis 2019; 76:66-71. [PMID: 30846359 DOI: 10.1016/j.bcmd.2019.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/02/2019] [Accepted: 02/17/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Wenxia Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Cuicui Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Dan Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Chen Liang
- Hematopoietic Stem Cell Transplant Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Leisheng Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Qiuqiu Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Hematopoietic Stem Cell Transplant Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yiying Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Meijuan Xia
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Hongtao Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Pei Su
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Hematopoietic Stem Cell Transplant Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Hematopoietic Stem Cell Transplant Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences & Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, China
| | - Fuxu Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China; Hematopoietic Stem Cell Transplant Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China.
| |
Collapse
|
440
|
Mikael PE, Willard C, Koyee A, Barlao CG, Liu X, Han X, Ouyang Y, Xia K, Linhardt RJ, Dordick JS. Remodeling of Glycosaminoglycans During Differentiation of Adult Human Bone Mesenchymal Stromal Cells Toward Hepatocytes. Stem Cells Dev 2019; 28:278-289. [PMID: 30572803 PMCID: PMC6389772 DOI: 10.1089/scd.2018.0197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/12/2018] [Indexed: 01/09/2023] Open
Abstract
There is a critical need to generate functional hepatocytes to aid in liver repair and regeneration upon availability of a renewable, and potentially personalized, source of human hepatocytes (hHEP). Currently, the vast majority of primary hHEP are obtained from human tissue through cadavers. Recent advances in stem cell differentiation have opened up the possibility to obtain fully functional hepatocytes from embryonic or induced pluripotent stem cells, or adult stem cells. With respect to the latter, human bone marrow mesenchymal stromal cells (hBMSCs) can serve as a source of autogenetic and allogenic multipotent stem cells for liver repair and regeneration. A major aspect of hBMSC differentiation is the extracellular matrix (ECM) composition and, in particular, the role of glycosaminoglycans (GAGs) in the differentiation process. In this study, we examine the influence of four distinct culture conditions/protocols (T1-T4) on GAG composition and hepatic markers. α-Fetoprotein and hepatocyte nuclear factor-4α were expressed continually over 21 days of differentiation, as indicated by real time quantitative PCR analysis, while albumin (ALB) expression did not begin until day 21. Hepatocyte growth factor (HGF) appears to be more effective than activin A in promoting hepatic-like cells through the mesenchymal-epithelial transition, perhaps due to the former binding to the HGF receptor to form a unique complex that diversifies the biological functions of HGF. Of the four protocols tested, uniform hepatocyte-like morphological changes, ALB secretion, and glycogen storage were found to be highest with protocol T2, which involves both early- and late-stage combinations of growth factors. The total GAG profile of the hBMSC ECM is rich in heparan sulfate (HS) and hyaluronan, both of which fluctuate during differentiation. The GAG profile of primary hHEP showed an HS-rich ECM, and thus, it may be possible to guide hBMSC differentiation to more mature hepatocytes by controlling the GAG profile expressed by differentiating cells.
Collapse
Affiliation(s)
- Paiyz E. Mikael
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Charles Willard
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Aurvan Koyee
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Charmaine-Grace Barlao
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, New York
| | - Xinyue Liu
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York
| | - Xiaorui Han
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York
| | - Yilan Ouyang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Ke Xia
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Robert J. Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York
| | - Jonathan S. Dordick
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Department of Biochemistry and Biophysics, Rensselaer Polytechnic Institute, Troy, New York
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
441
|
Broeckx SY, Seys B, Suls M, Vandenberghe A, Mariën T, Adriaensen E, Declercq J, Van Hecke L, Braun G, Hellmann K, Spaas JH. Equine Allogeneic Chondrogenic Induced Mesenchymal Stem Cells Are an Effective Treatment for Degenerative Joint Disease in Horses. Stem Cells Dev 2019; 28:410-422. [PMID: 30623737 PMCID: PMC6441287 DOI: 10.1089/scd.2018.0061] [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] [Indexed: 12/13/2022] Open
Abstract
Degenerative joint disease is one of the main causes of equine early retirement from pleasure riding or a performance career. The disease is initially triggered by an abnormal loading of normal cartilage or a normal loading of abnormal cartilage. This primary insult is accompanied with joint inflammation, which leads to further progressive degeneration of the articular cartilage and changes in the surrounding tissues. Therefore, in search for an effective treatment, 75 adult horses with early signs of degenerative fetlock joint disease were enrolled in a randomized, multicenter, double-blinded, and placebo-controlled study. Fifty animals were injected intra-articularly with the investigational veterinary product (IVP) consisting of allogeneic chondrogenic induced mesenchymal stem cells (ciMSCs) with equine allogeneic plasma, and 25 horses were injected with 0.9% NaCl (saline) control product. From week 3 to 18 after treatment, lameness scores (P < 0.001), flexion test responses (P < 0.034), and joint effusion scores (P < 0.001) were remarkably superior in IVP-treated horses. Besides nasal discharge in both treatment groups, no adverse events were observed during the entire study period. On long-term follow-up (1 year), significantly more investigational product-treated horses were working at training level or were returned to their previous level of work (P < 0.001).
Collapse
Affiliation(s)
- Sarah Y Broeckx
- 1 Global Stem cell Technology NV, Anacura Group, Evergem, Belgium
| | - Bert Seys
- 2 Equine Veterinary Practice Dr Suls, SP Weert, the Netherlands
| | - Marc Suls
- 2 Equine Veterinary Practice Dr Suls, SP Weert, the Netherlands
| | | | - Tom Mariën
- 4 Equitom Equine Hospital, Meldert, Belgium
| | - Edouard Adriaensen
- 3 Equine Veterinary Service Adriaensen-Vandenberghe, Waasmunster, Belgium.,4 Equitom Equine Hospital, Meldert, Belgium
| | | | - Lore Van Hecke
- 1 Global Stem cell Technology NV, Anacura Group, Evergem, Belgium
| | | | | | - Jan H Spaas
- 1 Global Stem cell Technology NV, Anacura Group, Evergem, Belgium
| |
Collapse
|
442
|
Surface tethering of stem cells with H 2O 2-responsive anti-oxidizing colloidal particles for protection against oxidation-induced death. Biomaterials 2019; 201:1-15. [PMID: 30784768 DOI: 10.1016/j.biomaterials.2019.01.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/08/2019] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells are the new generation of medicine for treating numerous vascular diseases and tissue defects because of their ability to secrete therapeutic factors. Poor cellular survival in an oxidative diseased tissue, however, hinders the therapeutic efficacy. To this end, we hypothesized that tethering the surface of stem cells with colloidal particles capable of discharging antioxidant cargos in response to elevated levels of hydrogen peroxide (H2O2) would maintain survival and therapeutic activity of the stem cells. We examined this hypothesis by encapsulating epigallocatechin gallate (EGCG) and manganese oxide (MnO2) nanocatalysts into particles comprising poly(d,l-lactide-co-glycolide)-block-hyaluronic acid. The MnO2 nanocatalysts catalyzed the decomposition of H2O2 into oxygen gas, which increased the internal pressure of particles and accelerated the release of EGCG by 1.5-fold. Consequently, stem cells exhibited 1.2-fold higher metabolic activity and 2.8-fold higher secretion level of pro-angiogenic factor in sub-lethal H2O2 concentrations. These stem cells, in turn, performed a greater angiogenic potential with doubled number of newly formed mature blood vessels. We envisage that the results of this study will contribute to improving the therapeutic efficacy of a wide array of stem cells.
Collapse
|
443
|
Zhang S, Teo KYW, Chuah SJ, Lai RC, Lim SK, Toh WS. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019; 200:35-47. [PMID: 30771585 DOI: 10.1016/j.biomaterials.2019.02.006] [Citation(s) in RCA: 385] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/11/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
The efficacy of mesenchymal stem cell (MSC) therapies is increasingly attributed to paracrine secretion, particularly exosomes. In this study, we investigated the role of MSC exosomes in the regulation of inflammatory response, nociceptive behaviour, and condylar cartilage and subchondral bone healing in an immunocompetent rat model of temporomandibular joint osteoarthritis (TMJ-OA). We observed that exosome-mediated repair of osteoarthritic TMJs was characterized by early suppression of pain and degeneration with reduced inflammation, followed by sustained proliferation and gradual improvements in matrix expression and subchondral bone architecture, leading to overall joint restoration and regeneration. Using chondrocyte cultures, we could attribute some of the cellular activities during exosome-mediated joint repair to adenosine activation of AKT, ERK and AMPK signalling. Specifically, MSC exosomes enhanced s-GAG synthesis impeded by IL-1β, and suppressed IL-1β-induced nitric oxide and MMP13 production. These effects were partially abrogated by inhibitors of adenosine receptor activation, AKT, ERK and AMPK phosphorylation. Together, our observations suggest that MSC exosomes promote TMJ repair and regeneration in OA through a well-orchestrated mechanism of action that involved multiple cellular processes to restore the matrix and overall joint homeostasis. This study demonstrates the translational potential of a cell-free ready-to-use exosome-based therapeutic for treating TMJ pain and degeneration.
Collapse
Affiliation(s)
- Shipin Zhang
- Faculty of Dentistry, National University of Singapore, Singapore
| | | | | | - Ruenn Chai Lai
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore; Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.
| |
Collapse
|
444
|
Immunohematology Mesenchymal Stromal Cell-based Therapy: From Research to Clinic. Appl Immunohistochem Mol Morphol 2019; 26:e26-e43. [PMID: 29271793 DOI: 10.1097/pai.0000000000000629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stromal cells (MSC) are nonhematopoietic cells that can be isolated from several adult and fetal tissues. MSC present specific features as the capacity to support hematopoiesis and to regulate immune response. Thus, the use of MSC as a cell therapeutic product in the field of immune-hematology is of great importance. In this review, we focused on human MSC and discussed their immune-hematologic properties and their translation toward therapeutic clinical applications. Thus, these features hold great promise for cell-based therapy and are of important relevance for the field.
Collapse
|
445
|
Ljung K, Grönlund A, Felldin U, Rodin S, Corbascio M, Österholm C, Grinnemo KH. Human Fetal Cardiac Mesenchymal Stromal Cells Differentiate In Vivo into Endothelial Cells and Contribute to Vasculogenesis in Immunocompetent Mice. Stem Cells Dev 2019; 28:310-318. [PMID: 30618344 DOI: 10.1089/scd.2018.0198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have shown great potential as a treatment for systemic inflammatory diseases, but their local regenerative properties are highly tissue- and site specific. Previous studies have demonstrated that adult human MSCs respond to inflammatory cytokines through the release of paracrine factors that stimulate angiogenesis, but they do not themselves differentiate into vascular structures in vivo. In this study, we used human fetal cardiac MSCs (hfcMSCs) harvested during the first trimester of heart development and injected them into the subcutaneous tissue of normal immunocompetent mice treated with short-term costimulation blockade for tolerance induction. When hfcMSCs were transplanted subcutaneously together with Matrigel matrix, they contributed to vasculogenesis through differentiation into endothelial cells and generation of the basal membrane protein Laminin α4. These characteristics of hfcMSCs are similar to the mesodermal progenitors giving rise to the developing heart and they may be useful for treatment of ischemic injuries.
Collapse
Affiliation(s)
- Karin Ljung
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,2 Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Grönlund
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Felldin
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sergey Rodin
- 3 Division of Cardiothoracic Surgery and Anaesthesiology, Department of Surgical Sciences, Uppsala University, Akademiska University Hospital, Uppsala, Sweden
| | - Matthias Corbascio
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,2 Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Österholm
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Karl-Henrik Grinnemo
- 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,3 Division of Cardiothoracic Surgery and Anaesthesiology, Department of Surgical Sciences, Uppsala University, Akademiska University Hospital, Uppsala, Sweden
| |
Collapse
|
446
|
Davis AB, Schnabel LV, Gilger BC. Subconjunctival bone marrow-derived mesenchymal stem cell therapy as a novel treatment alternative for equine immune-mediated keratitis: A case series. Vet Ophthalmol 2019; 22:674-682. [PMID: 30715781 DOI: 10.1111/vop.12641] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/11/2018] [Accepted: 11/24/2018] [Indexed: 12/26/2022]
Abstract
Equine immune-mediated keratitis (IMMK) leads to increased corneal opacity and inflammation secondary to an alteration of the local immune system. Bone marrow-derived mesenchymal stem cells (BM-MSC) have been shown to modulate the immune system by downregulating inflammation. Four horses with unilateral IMMK poorly responsive to traditional medical treatments underwent novel, autologous subconjunctival BM-MSC therapy. Bone marrow was harvested and processed as previously described for equine orthopedic disease. Horses received autologous subconjunctival BM-MSC injections approximately every 3-4 weeks for 1-5 treatments total. Horses were maintained on their current medical treatment regimen throughout the BM-MSC treatment period. Three horses had a positive response to therapy as demonstrated by an increase in corneal clarity, a decrease in neovascularization and a reduction in surface irregularity. One horse was nonresponsive to therapy. These experimental results demonstrate the safety and potential efficacy of an innovative solution for IMMK.
Collapse
Affiliation(s)
- Amanda B Davis
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Brian C Gilger
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
447
|
Najar M, Ouhaddi Y, Bouhtit F, Melki R, Afif H, Boukhatem N, Merimi M, Fahmi H. Empowering the immune fate of bone marrow mesenchymal stromal cells: gene and protein changes. Inflamm Res 2019; 68:167-176. [PMID: 30426152 DOI: 10.1007/s00011-018-1198-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/23/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE AND DESIGN Bone marrow mesenchymal stromal cells (BM-MSCs) are referred as a promising immunotherapeutic cell product. New approaches using empowered MSCs should be developed as for the treatment or prevention of different immunological diseases. Such preconditioning by new licensing stimuli will empower the immune fate of BM-MSCs and, therefore, promote a better and more efficient biological. Here, our main goal was to establish the immunological profile of BM-MSCs following inflammatory priming and in particular their capacity to adjust their immune-related proteome and transcriptome. MATERIAL AND METHODS To run this study, we have used BM-MSC cell cultures, a pro-inflammatory cytokine cocktail priming, flow cytometry analysis, qPCR and ELISA techniques. RESULTS Different expression levels of several immunological mediators such as COX-1, COX-2, LIF, HGF, Gal-1, HO-1, IL-11, IL-8, IL-6 and TGF-β were constitutively observed in BM-MSCs. Inflammation priming substantially but differentially modulated the gene and protein expression profiles of these mediators. Thus, expressions of COX-2, LIF, HGF, IL-11, IL-8 and IL-6 were highly increased/induced and those of COX-1, Gal-1, and TGF-β were reduced. CONCLUSIONS Collectively, we demonstrated that BM-MSCs are endowed with a specific and modular regulatory machinery which is potentially involved in immunomodulation. Moreover, BM-MSCs are highly sensitive to inflammation and respond to such signal by properly adjusting their gene and protein expression of regulatory factors. Using such preconditioning may empower the immune fate of MSCs and, therefore, enhance their value for cell-based immunotherapy.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada.
| | - Yassine Ouhaddi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Fatima Bouhtit
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Rahma Melki
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Hassan Afif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Noureddine Boukhatem
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Makram Merimi
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
448
|
Kaita Y, Tarui T, Yoshino H, Matsuda T, Yamaguchi Y, Nakagawa T, Asahi M, Ii M. Sufficient therapeutic effect of cryopreserved frozen adipose-derived regenerative cells on burn wounds. Regen Ther 2019; 10:92-103. [PMID: 30766897 PMCID: PMC6360408 DOI: 10.1016/j.reth.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/22/2018] [Accepted: 01/06/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction The purpose of this study was to evaluate whether cryopreserved (frozen) adipose-derived regenerative cells (ADRCs) have a therapeutic effect on burn wound healing as well as freshly isolated (fresh) ADRCs. Methods Full thickness burns were created on dorsum of nude mice and burn wound was excised. The wound was covered by artificial dermis with; (i) fresh ADRCs, (ii) frozen ADRCs, and (iii) PBS (control). The assessment for wound healing was performed by morphological, histopathological and immunohistochemical analyses. Results In vivo analyses exhibited the significant therapeutic effect of frozen ADRCs on burn wound healing up to the similar or higher level of fresh ADRCs. There were significant differences of wound closure, epithelized tissue thickness, and neovascularization between the treatment groups and control group. Although there was no significant difference of therapeutic efficacy between fresh ADRC group and frozen ADRC group, frozen ADRCs improved burn wound healing process in dermal regeneration with increased great type I collagen synthesis compared with fresh ADRCs. Conclusions These findings indicate that frozen ADRCs allow us to apply not only quickly but also for multiple times, and the cryopreserved ADRCs could therefore be useful for the treatment of burn wounds in clinical settings. Frozen ADRCs had a therapeutic effect on burn wounds as well as fresh ADRCs. Frozen ADRCs promoted tissue regeneration by paracrine factors. Frozen ADRCs could be useful for the treatment of burn wounds in clinical setting.
Collapse
Key Words
- ADRCs, adipose-derived regenerative cells
- AdSCs, adipose-derived stem cells
- Burn
- CM, conditioned medium
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM-F12, Dulbecco's modified Eagle's medium supplemented with F12
- EGF, Epidermal growth factor
- FBS, Fetal bovine serum
- FGF2, Fibroblast growth factor 2
- GAPDH, Glyceraldehyde 3-phosphate dehydrogenase
- HGF, Hepatocyte growth factor
- IGF-1, Insulin-like growth factor-1
- ILB4, Isolectin B4
- MSC, Mesenchymal stem cell
- NHDF, Normal human dermal fibroblast
- NHEK, Normal human epithelial keratinocyte
- PBS, Phosphate-buffered saline
- PVDF, Poly vinylidene di-fluoride
- SDS-PAGE, Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- SVF, Stromal vascular fraction
- Stem cells
- VEGF, Vascular endothelial growth factor
- Wound healing
- qRT-PCR, Quantitative real-time reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Yasuhiko Kaita
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Takehiko Tarui
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Hideaki Yoshino
- Division of Cardiology, Second Department of Internal Medicine, Kyorin University, School of Medicine, Tokyo, Japan
| | - Takeaki Matsuda
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Yoshihiro Yamaguchi
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Takatoshi Nakagawa
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Ii
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, Osaka, Japan
| |
Collapse
|
449
|
Examining the therapeutic potential of various stem cell sources for differentiation into insulin-producing cells to treat diabetes. ANNALES D'ENDOCRINOLOGIE 2019; 80:47-53. [DOI: 10.1016/j.ando.2018.06.1084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/24/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
|
450
|
Sahu A, Foulsham W, Amouzegar A, Mittal SK, Chauhan SK. The therapeutic application of mesenchymal stem cells at the ocular surface. Ocul Surf 2019; 17:198-207. [PMID: 30695735 DOI: 10.1016/j.jtos.2019.01.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 12/12/2022]
Abstract
The therapeutic potential of mesenchymal stem cells (MSCs) has been heralded by their multipotentiality and immunomodulatory capacity. MSCs migrate toward sites of tissue damage, where specific pro-inflammatory factors 'license' their immunosuppressive functions. Recent studies in animal models of ocular surface disease have demonstrated the potential of MSC-derived therapies to limit inflammation and promote tissue repair. Herein, we review the immunoregulatory mechanisms of MSCs, as well as strategies to harness their regenerative function at the cornea. We examine reports of the therapeutic application of MSCs in the setting of ocular surface inflammation; including corneal injury, transplantation, ocular surface autoimmunity and allergy.
Collapse
Affiliation(s)
- Anuradha Sahu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA; Institute of Ophthalmology, University College London, London, UK
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|