501
|
Damia E, Chicharro D, Lopez S, Cuervo B, Rubio M, Sopena JJ, Vilar JM, Carrillo JM. Adipose-Derived Mesenchymal Stem Cells: Are They a Good Therapeutic Strategy for Osteoarthritis? Int J Mol Sci 2018; 19:ijms19071926. [PMID: 29966351 PMCID: PMC6073660 DOI: 10.3390/ijms19071926] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of disability in elderly population around the world. More than one-third of people over 65 years old shows either clinical or radiological evidence of OA. There is no effective treatment for this degenerative disease, due to the limited capacity for spontaneous cartilage regeneration. Regarding the use of regenerative therapies, it has been reported that one option to restore degenerated cartilage are adipose-derived mesenchymal stem cells (ASCs). The purpose of this review is to describe and compare the efficacy of ASCs versus other therapies in OA. Methods: Recent studies have shown that ASCs exert paracrine effects protecting against degenerative changes in chondrocytes. According to the above, we have carried out a review of the literature using a combination of osteoarthritis, stem cells, and regenerative therapies as keywords. Results: Conventional pharmacological therapies for OA treatment are considered before the surgical option, however, they do not stop the progression of the disease. Moreover, total joint replacement is not recommended for patients under 55 years, and high tibia osteotomy (HTO) is a viable solution to address lower limb malalignment with concomitant OA, but some complications have been described. In recent years, the use of mesenchymal stem cells (MSCs) as a treatment strategy for OA is increasing considerably, thanks to their capacity to improve symptoms together with joint functionality and, therefore, the patients’ quality of life. Conclusions: ASC therapy has a positive effect on patients with OA, although there is limited evidence and little long-term follow-up.
Collapse
Affiliation(s)
- 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Sergio Lopez
- Department of Animal Pathology. Instituto Universitario de Investigaciones Biomédicas y Sanitarias. University of Las Palmas de Gran Canaria, 35416 Las Palmas de Gran Canaria, 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Jose Manuel Vilar
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
- Department of Animal Pathology. Instituto Universitario de Investigaciones Biomédicas y Sanitarias. University of Las Palmas de Gran Canaria, 35416 Las Palmas de Gran Canaria, Spain.
| | - Jose Maria Carrillo
- 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.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| |
Collapse
|
502
|
Combination of MSC spheroids wrapped within autologous composite sheet dually protects against immune rejection and enhances stem cell transplantation efficacy. Tissue Cell 2018; 53:93-103. [PMID: 30060833 DOI: 10.1016/j.tice.2018.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in transplantation therapy due to their multilineage differentiation potential, abundance, and immuno-modulating ability. However, the risk of allograft rejection limits their application. Here, we proposed a novel method to facilitate MSC transplantation with enhanced applicability and efficacy. We cultured human adipose-derived MSCs in a 3D culture under in vitro expansion conditions and under conventional 2D adherent culture conditions. MSC spheroids promoted extracellular matrix molecules that stimulate MSC proliferation, and produced more angiogenic cytokines such as vascular endothelial growth factor, hepatocyte growth factor, and fibroblast growth factor than 2D-cultured MSCs. Further, MSC spheroids showed increased IDO expression, increased proportion of M2 macrophages, and decreased macrophage proliferation, compared to 2D-cultured MSCs. Next, we proposed the wrapping of autologous cell sheets from the recipient around in-vitro-grown MSC spheroids to prevent allogenic immune rejection during transplantation. Myoblasts from C57BL/6 mice were used to prepare a stem cell composite sheet containing human-derived MSC spheres. The transplantation of MSC spheroids increased the survival rate and decreased the inflammatory response of the immunocompetent C57BL/6 ischemic mice. Thus, combining 3D-cultured MSC spheroid technology with immune evasion stem cell composite sheet improved the outcome and strengthened the protection against allogenic immune rejection.
Collapse
|
503
|
Fiore EJ, Domínguez LM, Bayo J, García MG, Mazzolini GD. Taking advantage of the potential of mesenchymal stromal cells in liver regeneration: Cells and extracellular vesicles as therapeutic strategies. World J Gastroenterol 2018; 24:2427-2440. [PMID: 29930465 PMCID: PMC6010941 DOI: 10.3748/wjg.v24.i23.2427] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/08/2018] [Accepted: 06/02/2018] [Indexed: 02/06/2023] Open
Abstract
Cell-based therapies for acute and chronic liver diseases are under continuous progress. Mesenchymal stem/stromal cells (MSCs) are multipotent cells able to migrate selectively to damaged tissue and contribute to its healing and regeneration. The MSC pro-regenerative effect occurs due to their immunomodulatory capacity and their ability to produce factors that promote cell protection and survival. Likewise, it has been observed that part of their paracrine effect is mediated by MSC-derived extracellular vesicles (EVs). EVs contain proteins, lipids and nucleic acids (DNA, mRNA, miRNA, lncRNA) from the cell of origin, allowing for intercellular communication. Recently, different studies have demonstrated that MSC-derived EVs could reproduce, at least in part, the biological effects obtained by MSC-based therapies. Moreover, due to EVs' stability for long periods of time and easy isolation methods they have become a therapeutic option to MSCs treatments. This review summarizes the latest results achieved in clinical trials using MSCs as cell therapy for liver regeneration, the role of EVs in liver physiopathology and the potential of MSCderived EVs as intercellular mediators and therapeutic tools in liver diseases.
Collapse
Affiliation(s)
- Esteban Juan Fiore
- Laboratory of Gene Therapy, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires 999071, Argentina
| | - Luciana María Domínguez
- Laboratory of Gene Therapy, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires 999071, Argentina
| | - Juan Bayo
- Laboratory of Gene Therapy, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires 999071, Argentina
| | - Mariana Gabriela García
- Laboratory of Gene Therapy, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires 999071, Argentina
| | - Guillermo Daniel Mazzolini
- Laboratory of Gene Therapy, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires 999071, Argentina
| |
Collapse
|
504
|
Egger D, Tripisciano C, Weber V, Dominici M, Kasper C. Dynamic Cultivation of Mesenchymal Stem Cell Aggregates. Bioengineering (Basel) 2018; 5:E48. [PMID: 29921755 PMCID: PMC6026937 DOI: 10.3390/bioengineering5020048] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/24/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered as primary candidates for cell-based therapies due to their multiple effects in regenerative medicine. Pre-conditioning of MSCs under physiological conditions—such as hypoxia, three-dimensional environments, and dynamic cultivation—prior to transplantation proved to optimize their therapeutic efficiency. When cultivated as three-dimensional aggregates or spheroids, MSCs display increased angiogenic, anti-inflammatory, and immunomodulatory effects as well as improved stemness and survival rates after transplantation, and cultivation under dynamic conditions can increase their viability, proliferation, and paracrine effects, alike. Only few studies reported to date, however, have utilized dynamic conditions for three-dimensional aggregate cultivation of MSCs. Still, the integration of dynamic bioreactor systems, such as spinner flasks or stirred tank reactors might pave the way for a robust, scalable bulk expansion of MSC aggregates or MSC-derived extracellular vesicles. This review summarizes recent insights into the therapeutic potential of MSC aggregate cultivation and focuses on dynamic generation and cultivation techniques of MSC aggregates.
Collapse
Affiliation(s)
- Dominik Egger
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| | - Carla Tripisciano
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria.
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria.
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via Università 4, 41121 Modena, Italy.
- Technopole of Mirandola TPM, 41037 Mirandola, Modena, Italy.
| | - Cornelia Kasper
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
505
|
Sun J, Lv J, Zhang W, Li L, Lv J, Geng Y, Yin A. Combination with miR-124a improves the protective action of BMSCs in rescuing injured rat podocytes from abnormal apoptosis and autophagy. J Cell Biochem 2018; 119:7166-7176. [PMID: 29904949 DOI: 10.1002/jcb.26771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
This in vitro study was performed to identify the role of miR-124a in bone marrow stromal stem cells (BMSCs) therapy for H2 O2 -induced rat podocyte injury, and determine whether combination treatment with miR-124a could improve the protective effect of BMSCs. Cell viability of podocytes was detected by CCK-8 assay. Detection of ROS level, apoptotic rate, and autophagy rate was carried out using flow cytometry assays. Oxidative stress parameters were analyzed using the ELISA assays. MiR-124a and mRNA levels were determined using real-time PCR. Protein expression was detected using Western blotting. Our study revealed a pivotal role of miR-124a in the protective action of BMSCs on podocyte injury driven by oxidative stress. BMSCs could rescue injured podocytes from aberrant apoptosis and autophagy by regulating cleaved caspase-3, Bax, Bcl-2, LC3-II/I, and p62. Suppression of the PI3 K/Akt/mTOR signaling pathway is likely one of the main mechanisms underlying the protective action of BMSCs transfected with miR-124a. Our study revealed that miR-124a further improves the protective effect of BMSCs in injured podocytes. Thus, the combination of BMSCs and microRNAs could be a beneficial treatment for renal diseases in the near future.
Collapse
Affiliation(s)
- Jiping Sun
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenjing Zhang
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lili Li
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jia Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingzhou Geng
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Aiping Yin
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
506
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
507
|
Romeo L, Diomede F, Gugliandolo A, Scionti D, Lo Giudice F, Lanza Cariccio V, Iori R, Bramanti P, Trubiani O, Mazzon E. Moringin Induces Neural Differentiation in the Stem Cell of the Human Periodontal Ligament. Sci Rep 2018; 8:9153. [PMID: 29904155 PMCID: PMC6002387 DOI: 10.1038/s41598-018-27492-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/04/2018] [Indexed: 12/26/2022] Open
Abstract
The therapeutic strategies for neurodegenerative diseases still represent a vast research field because of the lack of targeted, effective and resolutive treatment for neurodegenerative diseases. The use of stem cell-based therapy is an alternative approach that could lead to the replacement of damaged neuronal tissue. For this purpose, adult mesenchymal stem cells (MSC), including periodontal ligament stem cells (PDLSCs), could be very useful for their differentiation capacity, easy isolation and the ability to perform an autologous implant. The aim of this work was to test whether the Moringin [4-(α-L-rhamnosyloxy) benzyl isothiocyanate; GMG-ITC], an isothiocyanate extracted from Moringa oleifera seeds, was able to induce PDLSCs toward neural progenitor differentiation. Next-generation transcriptomics sequencing showed that moringin treatment increased the expression of genes involved in neuron cortical development and in particular in neuron belonging to upper and deep cortical layers. Moreover, moringin treatment upregulated genes involved in osteogenesis and adipogenesis although with a lower fold change compared to upregulated genes involved in neuronal differentiation. Finally, moringin did not induce the expression of oncogenes resulting in a safe treatment.
Collapse
Affiliation(s)
- Letizia Romeo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Francesca Diomede
- Department of medical, oral and biotechnological sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Domenico Scionti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Fabrizio Lo Giudice
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Veronica Lanza Cariccio
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Renato Iori
- Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria, Centro di ricerca Agricoltura e Ambiente (CREA-AA), Via di Corticella 133, 40128, Bologna, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy
| | - Oriana Trubiani
- Department of medical, oral and biotechnological sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124, Messina, Italy.
| |
Collapse
|
508
|
Bellavia D, Raimondi L, Costa V, De Luca A, Carina V, Maglio M, Fini M, Alessandro R, Giavaresi G. Engineered exosomes: A new promise for the management of musculoskeletal diseases. Biochim Biophys Acta Gen Subj 2018; 1862:1893-1901. [PMID: 29885361 DOI: 10.1016/j.bbagen.2018.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/16/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exosomes are nanovesicles actively secreted by potentially all cell types, including tumour cells, with the primary role of extracellular systemic communication mediators, both at autocrine and paracrine levels, at short and long distances. Recently, different studies have used exosomes as a delivery system for a plethora of different molecules, such as drugs, microRNAs and proteins. This has been made possible thanks to the simplicity in exosomes engineering, their great stability and versatility for applications in oncology as well as in regenerative medicine. SCOPE OF REVIEW The aim of this review is to provide information on the state-of-the-art and possible applications of engineered exosomes, both for cargo and specific cell-targeting, in different pathologies related to the musculoskeletal system. MAJOR CONCLUSIONS The use of exosomes as therapeutic agents is rapidly evolving, different studies explore drug delivery with exosomes using different molecules, showing an enormous potential in various research fields such as oncology and regenerative medicine. GENERAL SIGNIFICANCE However, despite the significant progress made by the different studies carried out, currently, the use of exosomes is not a therapeutic reality for the considerable difficulties to overcome.
Collapse
Affiliation(s)
- D Bellavia
- IRCCS Istituto ortopedico Rizzoli, Bologna, Italy.
| | - L Raimondi
- IRCCS Istituto ortopedico Rizzoli, Bologna, Italy
| | - V Costa
- IRCCS Istituto ortopedico Rizzoli, Bologna, Italy
| | - A De Luca
- IRCCS Istituto ortopedico Rizzoli, Bologna, Italy
| | - V Carina
- IRCCS Istituto ortopedico Rizzoli, Bologna, Italy
| | - M Maglio
- IRCCS Istituto ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - M Fini
- IRCCS Istituto ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - R Alessandro
- Department of Biopathology and Medical Biotechnologies, Section of Biology and Genetics, University of Palermo, Palermo 90133, Italy; Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy
| | - G Giavaresi
- IRCCS Istituto ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| |
Collapse
|
509
|
Barrachina L, Remacha AR, Romero A, Zaragoza P, Vázquez FJ, Rodellar C. Differentiation of equine bone marrow derived mesenchymal stem cells increases the expression of immunogenic genes. Vet Immunol Immunopathol 2018; 200:1-6. [DOI: 10.1016/j.vetimm.2018.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/14/2018] [Accepted: 04/09/2018] [Indexed: 12/27/2022]
|
510
|
Yang H, Tian W, Wang S, Liu X, Wang Z, Hou L, Ge J, Zhang X, He Z, Wang X. TSG-6 secreted by bone marrow mesenchymal stem cells attenuates intervertebral disc degeneration by inhibiting the TLR2/NF-κB signaling pathway. J Transl Med 2018; 98:755-772. [PMID: 29483622 DOI: 10.1038/s41374-018-0036-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/11/2022] Open
Abstract
Inflammation has been correlated with intervertebral disc degeneration (IDD). Recent evidence suggests that TNF-α-stimulated gene 6 protein (TSG-6) secreted by bone marrow mesenchymal stem cells (BMSCs) displays a remarkable ability to inhibit inflammatory processes in a variety of diseases. However, it is unknown whether BMSCs exert their therapeutic effect against IDD by secreting TSG-6. Here we investigated the effects of BMSCs and TSG-6 on IDD and explored the possible underlying mechanisms in vitro and in vivo. We found that BMSCs and TSG-6 reduced the expression of MMP-3 and MMP-13, and increased the expression of collagen II and aggrecan in the IL-1β-treated nucleus pulposus cells (NPCs), but the protective effects of BMSCs and TSG-6 were attenuated when TSG-6 expression was silenced. We also found that the activation of the TLR2/NF-κB pathway was inhibited by BMSCs and TSG-6. The levels of IL-6 and TNF-α in the degenerated NPCs were reduced and the proliferation of IL-1β-treated NPCs was increased in the presence of BMSCs and TSG-6. Furthermore, in vivo experiments showed that BMSCs and TSG-6 restored the MRI T2-weighted signal intensity and increased collagen II and aggrecan expression in the degenerated nucleus pulposus (NP) tissues. Finally, our results showed that BMSCs and TSG-6 downregulated the TLR2/NF-κB signaling and reduced the expression of MMPs and inflammatory cytokines in the degenerated NP tissues. The present study is the first to demonstrate the involvement of TLR2/NF-κB pathway in the potential anti-IDD therapeutic effect of TSG-6, and the results provide new insight into the beneficial effect of BMSCs in the treatment of IDD.
Collapse
Affiliation(s)
- Hao Yang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Weitian Tian
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shaocheng Wang
- Department of Anesthesiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaohua Liu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhankui Wang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lei Hou
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiaxi Ge
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao Zhang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhengyu He
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Xiangrui Wang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
511
|
Brandão JS, Alvarenga ML, Pfeifer JPH, Dos Santos VH, Fonseca-Alves CE, Rodrigues M, Laufer-Amorim R, Castillo JAL, Alves ALG. Allogeneic mesenchymal stem cell transplantation in healthy equine superficial digital flexor tendon: A study of the local inflammatory response. Res Vet Sci 2018; 118:423-430. [PMID: 29715649 DOI: 10.1016/j.rvsc.2018.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/13/2018] [Accepted: 03/18/2018] [Indexed: 01/01/2023]
Abstract
The superficial digital flexor tendon (SDFT) is a structure frequently affected by injuries in high-performance athletic horses, and there are limited therapeutic options. Regenerative medicine has evolved significantly in treating different illnesses. However, understanding the cellular behaviour during mesenchymal stem cell (MSC) transplantation in healthy tissues is not fully known yet. To address the inflammatory response induced by allogeneic MSC transplantation, this study evaluated the local inflammatory response after the application of allogeneic adipose tissue-derived mesenchymal stem cells (AT-MSCs) in the equine tendon compared to an autologous transplant and the control group. Eighteen thoracic limbs (TL) in nine animals were divided into three groups and subjected to the application of AT-MSCs in the healthy tendon. In the allogeneic group (Gallog), the animals received an allogeneic AT-MSC application in the TL. The autologous group (Gauto) received an application of autologous cells in the TL, and in the control group (Gcont), phosphate-buffered saline (PBS) was applied. There were no significant differences among the evaluated groups in the physical, morphological, thermography, and ultrasonography analyses. A higher number of CD3-positive lymphocytes was observed in the Gauto group compared to the control (P < 0.05). Additionally, we did not observe different expressions of CD172 and microvascular density among the groups. The allogeneic transplantation of AT-MSCs did not result in an adverse or inflammatory reaction that compromised the use of these cells in this experiment. Their behaviour was similar to that of autologous transplantation.
Collapse
Affiliation(s)
- Jaqueline Souza Brandão
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Marina Landim Alvarenga
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - João Pedro Hubbe Pfeifer
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Vitor Hugo Dos Santos
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Carlos Eduardo Fonseca-Alves
- Department de Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Mirian Rodrigues
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Renée Laufer-Amorim
- Department de Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - José Antonio Lucas Castillo
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil
| | - Ana Liz Garcia Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Distrito de Rubião Júnior s/n, Botucatu, SP 18618-9070, Brazil.
| |
Collapse
|
512
|
Mohamed MT, Embaby EA, Labib A, El-Husseiny M, Khamis H, El-Demery A, Shoukry MM. Effects of exercise in combination with autologous bone marrow stem cell transplantation for patients with type 1 diabetes. Physiother Theory Pract 2018; 35:1233-1242. [PMID: 29775119 DOI: 10.1080/09593985.2018.1474511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Stem cell therapy is a promising approach for the treatment of type 1 diabetes mellitus (T1D). Previous studies recommended regular exercise for the control of T1D. Experimental studies showed that a combination of stem cells and exercise yielded a better outcome. Yet, the effect of exercise programs following stem cell transplantation in patients with T1D has not been investigated. Thus, the current study aimed to examine the effect of a combined exercise program on measures of glycemic control in patients with T1D who received autologous bone marrow stem cell transplantation (ABMSCT). Thirty patients with controlled T1D were assigned into two equal groups. Both groups underwent ABMSCT and received insulin therapy and a diabetic diet regime. Only the exercise group followed the combined exercise program. Outcome measures of glycemic control (i.e. fasting blood glucose level [FBG], post-prandial blood glucose level [PPG], HbA1c, daily insulin dosage, and C-peptide levels) were tested before and after a 3-month rehabilitation period. There were significant (p < 0.05) decreases in all outcome measures except C-peptides after ABMSCT compared with before in both groups. Moreover, there was a significant decrease in the mean value of HbA1c in the exercise group compared with the control group after rehabilitation. Overall, this study strengthens the idea that adding exercise to ABMSCT is important to help control diabetes in patients with T1D.
Collapse
Affiliation(s)
- Marwa Taher Mohamed
- Department of Basic Science, Faculty of Physical Therapy, Cairo University, Giza, Egypt.,Department of Physical Therapy, Wadi El-Neel Hospital, Cairo, Egypt
| | - Eman Ahmed Embaby
- Department of Basic Science, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Awatif Labib
- Department of Basic Science, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | | | - Hazem Khamis
- Department of Cardiology, 6th October University, Giza, Egypt
| | - Ahmed El-Demery
- Department of Medical Biocemistry, 6th October University, Elmehawar Almarkazy, Giza, Egypt
| | - Mohamed Mounir Shoukry
- Faculty of Pharmacy, MTI University, Cairo, Egypt.,Department of Pharmacology, Wadi El-Neel Hospital, Cairo, Egypt
| |
Collapse
|
513
|
Park MJ, Lee J, Byeon JS, Jeong DU, Gu NY, Cho IS, Cha SH. Effects of three-dimensional spheroid culture on equine mesenchymal stem cell plasticity. Vet Res Commun 2018; 42:171-181. [PMID: 29721754 DOI: 10.1007/s11259-018-9720-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 04/02/2018] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) are useful candidates for tissue engineering and cell therapy fields. We optimize culture conditions of equine adipose tissue-derived MSCs (eAD-MSCs) for treatment of horse fractures. To investigate enhancing properties of three-dimensional (3D) culture system in eAD-MSCs, we performed various sized spheroid formation and determined changes in gene expression levels to obtain different sized spheroid for cell therapy. eAD-MSCs were successfully isolated from horse tailhead. Using hanging drop method, spheroid formation was generated for three days. Quantitative real-time PCR was performed to analyze gene expression. As results, expression levels of pluripotent markers were increased depending on spheroid size and the production of PGE2 was increased in spheroid formation compared to that in monolayer. Ki-67 showed a remarkable increase in the spheroid formed with 2.0 × 105 cells/drop as compared to that in the monolayer. Expression levels of angiogenesis-inducing factors such as VEGF, IL-6, IL-8, and IL-18 were significantly increased in spheroid formation compared to those in the monolayer. Expression levels of bone morphogenesis-inducing factors such as Cox-2 and TGF-β1 were also significantly increased in spheroid formation compared to those in the monolayer. Expression levels of osteocyte-specific markers such as RUNX2, osteocalcin, and differentiation potential were also significantly increased in spheroid formation compared to those in the monolayer. Therefore, spheroid formation of eAD-MSCs through the hanging drop method can increases the expression of angiogenesis-inducing and bone morphogenesis-inducing factors under optimal culture conditions.
Collapse
Affiliation(s)
- Mi Jeong Park
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jeong Su Byeon
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Da-Un Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - In-Soo Cho
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Sang-Ho Cha
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea.
| |
Collapse
|
514
|
Mesenchymal Stromal Cells: From Discovery to Manufacturing and Commercialization. Stem Cells Int 2018; 2018:4083921. [PMID: 30057622 PMCID: PMC6051015 DOI: 10.1155/2018/4083921] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/01/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
Over the last decades, mesenchymal stromal cells (MSC) have been the focus of intense research by academia and industry due to their unique features. MSC can be easily isolated and expanded through in vitro culture by taking full advantage of their self-renewing capacity. In addition, MSC exert immunomodulatory effects and can be differentiated into various lineages, which makes them highly attractive for clinical applications in cell-based therapies. In this review, we attempt to provide a brief historical overview of MSC discovery, characterization, and the first clinical studies conducted. The current MSC manufacturing platforms are reviewed with special attention regarding the use of bioreactors for the production of GMP-compliant clinically relevant cell numbers. The first commercial MSC-based products are also addressed, as well as the remaining challenges to the widespread use of MSC-derived products.
Collapse
|
515
|
Comparison of trophic factors secreted from human adipose-derived stromal vascular fraction with those from adipose-derived stromal/stem cells in the same individuals. Cytotherapy 2018; 20:589-591. [DOI: 10.1016/j.jcyt.2018.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
|
516
|
Planchon SM, Lingas KT, Reese Koç J, Hooper BM, Maitra B, Fox RM, Imrey PB, Drake KM, Aldred MA, Lazarus HM, Cohen JA. Feasibility of mesenchymal stem cell culture expansion for a phase I clinical trial in multiple sclerosis. Mult Scler J Exp Transl Clin 2018; 4:2055217318765288. [PMID: 29623216 PMCID: PMC5881997 DOI: 10.1177/2055217318765288] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/22/2018] [Indexed: 12/17/2022] Open
Abstract
Background Multiple sclerosis is an inflammatory, neurodegenerative disease of the central nervous system for which therapeutic mesenchymal stem cell transplantation is under study. Published experience of culture-expanding multiple sclerosis patients' mesenchymal stem cells for clinical trials is limited. Objective To determine the feasibility of culture-expanding multiple sclerosis patients' mesenchymal stem cells for clinical use. Methods In a phase I trial, autologous, bone marrow-derived mesenchymal stem cells were isolated from 25 trial participants with multiple sclerosis and eight matched controls, and culture-expanded to a target single dose of 1-2 × 106 cells/kg. Viability, cell product identity and sterility were assessed prior to infusion. Cytogenetic stability was assessed by single nucleotide polymorphism analysis of mesenchymal stem cells from 18 multiple sclerosis patients and five controls. Results One patient failed screening. Mesenchymal stem cell culture expansion was successful for 24 of 25 multiple sclerosis patients and six of eight controls. The target dose was achieved in 16-62 days, requiring two to three cell passages. Growth rate and culture success did not correlate with demographic or multiple sclerosis disease characteristics. Cytogenetic studies identified changes on one chromosome of one control (4.3%) after extended time in culture. Conclusion Culture expansion of mesenchymal stem cells from multiple sclerosis patients as donors is feasible. However, culture time should be minimized for cell products designated for therapeutic administration.
Collapse
Affiliation(s)
| | - Karen T Lingas
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Jane Reese Koç
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Brittney M Hooper
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Basabi Maitra
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Robert M Fox
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | - Peter B Imrey
- Mellen Center, Neurological Institute, USA.,Quantitative Health Sciences, Lerner Research Institute, USA
| | - Kylie M Drake
- Genomic Medicine Institute, Lerner Research Institute, USA
| | | | - Hillard M Lazarus
- Case Comprehensive Cancer Center and National Center for Regenerative Medicine, Case Western Reserve University and Seidman Cancer Center, USA
| | | |
Collapse
|
517
|
Alagesan S, Sanz-Nogués C, Chen X, Creane M, Ritter T, Ceredig R, O'Brien T, Griffin MD. Anti-donor antibody induction following intramuscular injections of allogeneic mesenchymal stromal cells. Immunol Cell Biol 2018; 96:536-548. [PMID: 29446493 DOI: 10.1111/imcb.12024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/10/2018] [Accepted: 02/11/2018] [Indexed: 12/20/2022]
Abstract
Allogeneic mesenchymal stromal cells (allo-MSC) are a promising "off-the-shelf" therapy with anti-inflammatory and pro-repair properties. This study investigated humoral immune responses to intramuscular (IM) injections of allo-MSC. Total and isotype-specific anti-donor IgG and donor-specific complement-mediated lysis were determined in sera from healthy mice 2 weeks after single or repeated IM injections of fully mismatched-MHC allo-MSC with comparison to mice receiving syngeneic MSC, allogeneic splenocytes or saline. In mice subjected to hind limb ischemia (HLI), anti-donor IgG was analyzed following IM allo-MSC injection with and without administration of the T-cell immunosuppressant tacrolimus. Recipients of single and repeated IM allo-MSC developed readily-detectable anti-donor IgG. Serum anti-donor IgG levels were similar to those of allo-splenocyte recipients but had higher IgG1/IgG2a ratio and variable capacity for complement-mediated lysis of donor cells. The induced anti-donor IgG bound readily to allo-MSC and this binding was increased following allo-MSC pretreatment with interferon gamma. In mice with HLI, IM injection of allo-MSC into the ischemic limb was also associated with induction of anti-donor IgG but this was abrogated by tacrolimus (FK-506). The results indicate that allo-MSC are inherently immunogenic when delivered intramuscularly to healthy and ischemic mouse hind limb, but induce an IgG1-skewed humoral response that is suppressed by tacrolimus.
Collapse
Affiliation(s)
- Senthilkumar Alagesan
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Clara Sanz-Nogués
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Xizhe Chen
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Michael Creane
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Rhodri Ceredig
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Galway, Ireland
| |
Collapse
|
518
|
Gharaei MA, Xue Y, Mustafa K, Lie SA, Fristad I. Human dental pulp stromal cell conditioned medium alters endothelial cell behavior. Stem Cell Res Ther 2018; 9:69. [PMID: 29562913 PMCID: PMC5861606 DOI: 10.1186/s13287-018-0815-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/01/2018] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Angiogenesis is of utmost importance for tissue regeneration and repair. Human dental pulp stromal cells (hDPSCs) possess angiogenic potential, as they secrete paracrine factors that may alter the host microenvironment. However, more insight into how hDPSCs guide endothelial cells (ECs) in a paracrine fashion is yet to be obtained. Therefore, the current study aimed to investigate the effect(s) of conditioned medium derived from hDPSCs (hDPSC-CM) on EC behavior in vitro. Methods hDPSCs were harvested from third molars scheduled for surgical removal under informed consent. The angiogenic profile of hDPSC-CM was identified using human angiogenesis antibody array and enzyme-linked immunosorbent assay (ELISA). Using real-time reverse transcription-polymerase chain reaction (RT-PCR) and ELISA, the mRNA and protein expression level of specific angiogenic biomarkers was determined in human umbilical vein endothelial cells (HUVECs) exposed to hDPSC-CM. The effect of hDPSC-CM on HUVEC attachment, proliferation and migration was evaluated by crystal violet staining, MTT, transwell migration along with real-time cell monitoring assays (xCELLigence; ACEA Biosciences, Inc.). A Matrigel assay was included to examine the influence of hDPSC-CM on HUVEC network formation. Endothelial growth medium (EGM-2) and EGM-2 supplemented with hDPSC-CM served as experimental groups, whereas endothelial basal medium (EBM-2) was set as negative control. Results A wide range of proangiogenic and antiangiogenic factors, including vascular endothelial growth factor, tissue inhibitor of metalloproteinase protein 1, plasminogen activator inhibitor (serpin E1), urokinase plasminogen activator and stromal cell-derived factor 1, was abundantly detected in hDPSC-CM by protein profiling array and ELISA. hDPSC-CM significantly accelerated the adhesion phases, from sedimentation to attachment and spreading, the proliferation rate and migration of HUVECs as shown in both endpoint assays and real-time cell analysis recordings. Furthermore, Matrigel assay demonstrated that hDPSC-CM stimulated tubulogenesis, affecting angiogenic parameters such as the number of nodes, meshes and total tube length. Conclusions The sustained proangiogenic and promaturation effects of hDPSC-CM shown in this in vitro study strongly suggest that the trophic factors released by hDPSCs are able to trigger pronounced angiogenic responses, even beyond EGM-2 considered as an optimal culture condition for ECs.
Collapse
Affiliation(s)
- M A Gharaei
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Y Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - K Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - S A Lie
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - I Fristad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway.
| |
Collapse
|
519
|
Orekhov PY, Konoplyannikov MA, Baklaushev VP, Kalsin VAA, Averyanov AV, Konopliannikov AG, Habazov RI, Troitskiy AV. Bone marrow stem cells for the critical limb ischemia treatment: biological aspects and clinical application. GENES & CELLS 2018; 13:20-34. [DOI: 10.23868/201805002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
Cell therapy is one of the most promising directions in the treatment of critical limb ischemia (CLI). In spite of certain advances achieved in this field in the last decades, which are related to application of bone marrow stem cells (BMSC), a large number of problems still remain unsolved. In this review, we discuss the BMSC biology, mechanisms of their therapeutic effect in the CLI treatment and results of the most notable BMSC-based clinical studies in detail.
Collapse
|
520
|
Kim JS, Chung H, Byun N, Kang SJ, Lee S, Shin JS, Park CG. Construction of EMSC-islet co-localizing composites for xenogeneic porcine islet transplantation. Biochem Biophys Res Commun 2018; 497:506-512. [DOI: 10.1016/j.bbrc.2018.02.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 11/25/2022]
|
521
|
Jia H, Yan Y, Liang Z, Tandra N, Zhang B, Wang J, Xu W, Qian H. Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review). Mol Med Rep 2018; 17:3439-3447. [PMID: 29257336 DOI: 10.3892/mmr.2017.8311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/09/2017] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) is a common and serious medical condition associated with poor health outcomes. Autophagy is a conserved multistep pathway that serves a major role in many biological processes and diseases. Recent studies have demonstrated that autophagy is induced in proximal tubular cells during AKI. Autophagy serves a pro‑survival or pro‑death role under certain conditions. Furthermore, mesenchymal stem cells (MSCs) have therapeutic potential in the repair of renal injury. This review summarizes the recent progress on the role of autophagy in AKI and MSCs‑based therapy for AKI. Further research is expected to prevent and treat acute kidney injury.
Collapse
Affiliation(s)
- Haoyuan Jia
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongmin Yan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Nitin Tandra
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Juanjuan Wang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
522
|
Xiang C, Yang K, Liang Z, Wan Y, Cheng Y, Ma D, Zhang H, Hou W, Fu P. Sphingosine-1-phosphate mediates the therapeutic effects of bone marrow mesenchymal stem cell-derived microvesicles on articular cartilage defect. Transl Res 2018; 193:42-53. [PMID: 29324234 DOI: 10.1016/j.trsl.2017.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 12/07/2017] [Accepted: 12/10/2017] [Indexed: 01/22/2023]
Abstract
Microvesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular components to target cells, yet their function in disease is just being explored. However, the therapeutic effects of MVs in cartilage injury and degeneration remain unknown. We found MVs contained high levels of sphingosine-1-phosphate (S1P) compared with the original bone marrow mesenchymal stem cells (MSCs). The enrichment of S1P in MVs was mediated by sphingosine kinase 1 (SphK1), but not by sphingosine kinase 2 (SphK2). Co-culture of human chondrocytes with MVs resulted in increased proliferation of chondrocytes in vitro, which was mediated by activation of S1P receptor 1 (S1PR1) expressed on chondrocytes. Meanwhile, MVs inhibited interleukin 1 beta-induced human chondrocytes apoptosis in a dose dependent manner. Furthermore, uptake of MVs by primary cultures of human chondrocytes was mediated by CD44 expressed by MVs. Anti-CD44 antibody significantly reduced the uptake of fluorescent protein-labeled MVs by chondrocytes. Further, blocking S1P by its neutralizing antibody significantly inhibited the therapeutic effects of MVs in vivo. Taken together, MVs showed therapeutic potential for treatment of clinical cartilage injury. This therapeutic potential is due to CD44-mediated uptake of MVs by chondrocytes and the S1P/S1PR1 axis-mediated proliferative effects of MVs on chondrocytes.
Collapse
Affiliation(s)
- Chuan Xiang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Kun Yang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhiyong Liang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yulong Wan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanwei Cheng
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dong Ma
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Heng Zhang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weiyu Hou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Panfeng Fu
- Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
523
|
Melly L, Cerino G, Frobert A, Cook S, Giraud MN, Carrel T, Tevaearai Stahel HT, Eckstein F, Rondelet B, Marsano A, Banfi A. Myocardial infarction stabilization by cell-based expression of controlled Vascular Endothelial Growth Factor levels. J Cell Mol Med 2018; 22:2580-2591. [PMID: 29478261 PMCID: PMC5908097 DOI: 10.1111/jcmm.13511] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/23/2017] [Indexed: 01/24/2023] Open
Abstract
Vascular Endothelial Growth Factor (VEGF) can induce normal or aberrant angiogenesis depending on the amount secreted in the microenvironment around each cell. Towards a possible clinical translation, we developed a Fluorescence Activated Cell Sorting (FACS)-based technique to rapidly purify transduced progenitors that homogeneously express a desired specific VEGF level from heterogeneous primary populations. Here, we sought to induce safe and functional angiogenesis in ischaemic myocardium by cell-based expression of controlled VEGF levels. Human adipose stromal cells (ASC) were transduced with retroviral vectors and FACS purified to generate two populations producing similar total VEGF doses, but with different distributions: one with cells homogeneously producing a specific VEGF level (SPEC), and one with cells heterogeneously producing widespread VEGF levels (ALL), but with an average similar to that of the SPEC population. A total of 70 nude rats underwent myocardial infarction by coronary artery ligation and 2 weeks later VEGF-expressing or control cells, or saline were injected at the infarction border. Four weeks later, ventricular ejection fraction was significantly worsened with all treatments except for SPEC cells. Further, only SPEC cells significantly increased the density of homogeneously normal and mature microvascular networks. This was accompanied by a positive remodelling effect, with significantly reduced fibrosis in the infarcted area. We conclude that controlled homogeneous VEGF delivery by FACS-purified transduced ASC is a promising strategy to achieve safe and functional angiogenesis in myocardial ischaemia.
Collapse
Affiliation(s)
- Ludovic Melly
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Giulia Cerino
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Aurélien Frobert
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | - Stéphane Cook
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | | | - Thierry Carrel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Hendrik T Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Friedrich Eckstein
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Benoît Rondelet
- Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Anna Marsano
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
524
|
Ziaei M, Greene C, Green CR. Wound healing in the eye: Therapeutic prospects. Adv Drug Deliv Rev 2018; 126:162-176. [PMID: 29355667 DOI: 10.1016/j.addr.2018.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/06/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
In order to maintain a smooth optical surface the corneal epithelium has to continuously renew itself so as to maintain its function as a barrier to fluctuating external surroundings and various environmental insults. After trauma, the cornea typically re-epithelializes promptly thereby minimizing the risk of infection, opacification or perforation. A persistent epithelial defect (PED) is usually referred to as a non-healing epithelial lesion after approximately two weeks of treatment with standard therapies to no avail. They occur following exposure to toxic agents, mechanical injury, and ocular surface infections and are associated with significant clinical morbidity in patients, resulting in discomfort or visual loss. In the case of deeper corneal injury and corneal pathology the wound healing cascade can also extend to the corneal stroma, the layer below the epithelium. Although significant progress has been made in recent years, pharmaco-therapeutic agents that promote corneal healing remain limited. This article serves as a review of current standard therapies, recently introduced alternative therapies gaining in popularity, and a look into the newest developments into ocular wound healing.
Collapse
|
525
|
Wang Y, Pati S, Schreiber M. Cellular therapies and stem cell applications in trauma. Am J Surg 2018; 215:963-972. [PMID: 29502858 DOI: 10.1016/j.amjsurg.2018.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND As the leading cause of mortality in the United States, trauma management have improved drastically over the past few decades with improved resuscitation and hemorrhage control. Stem cells are being used in an attempt to augment healing from trauma. DATA SOURCES PubMed and ClinicalTrials.gov were searched for published and registered pre-clinical and clinical trials for the application of stem cells to AKI, ARDS, shock, infection, TBI, wound healing, and bone healing. CONCLUSIONS Stem cell therapy for augmentation of healing traumatic injuries appears safe, as demonstrated by completed phase I/II trials. Further large scale studies are needed to assess the clinical efficacy.
Collapse
Affiliation(s)
- Yuxuan Wang
- Oregon Health and Science University, Department of Trauma, Surgical Critical Care, and Acute Care Surgery, USA.
| | - Shibani Pati
- University of California, San Francisco, Department of Laboratory Medicine, USA
| | - Martin Schreiber
- Oregon Health and Science University, Department of Trauma, Surgical Critical Care, and Acute Care Surgery, USA
| |
Collapse
|
526
|
Damania A, Jaiman D, Teotia AK, Kumar A. Mesenchymal stromal cell-derived exosome-rich fractionated secretome confers a hepatoprotective effect in liver injury. Stem Cell Res Ther 2018; 9:31. [PMID: 29409540 PMCID: PMC5801895 DOI: 10.1186/s13287-017-0752-6] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/18/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are an attractive therapeutic agent in regenerative medicine. Recently, there has been a paradigm shift from differentiation of MSCs to their paracrine effects at the injury site. Several reports elucidate the role of trophic factors secreted by MSCs toward the repair of injured tissues. We hypothesize that fractionating the MSC secretome will enrich exosomes containing soluble bioactive molecules, improving its therapeutic potential for liver failure. METHODS Rat bone marrow MSCs were isolated and the conditioned media filtered, concentrated and ultracentrifuged to generate fractionated secretome. This secretome was characterized for the presence of exosomes and recovery from liver injury assessed in in-vitro liver injury models. The results were further validated in vivo. RESULTS Studies on in-vitro liver injury models using acetaminophen and hydrogen peroxide show better cell recovery and reduced cytotoxicity in the presence of fractionated as opposed to unfractionated secretome. Further, the cells showed reduced oxidative stress in the presence of fractionated secretome, suggesting a potential antioxidative effect. These results were further validated in vivo in liver failure models, wherein improved liver regeneration in the presence of fractionated secretome (0.819 ± 0.035) was observed as compared to unfractionated secretome (0.718 ± 0.042). CONCLUSIONS The work presented is a proof of concept that fractionating the secretome enriches certain bioactive molecules involved in the repair and recovery of injured liver tissue. Exosome enriched mesenchymal stromal cell-derived fractionated secretome potentiates recovery upon injection in injured liver.
Collapse
Affiliation(s)
- Apeksha Damania
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 UP India
| | - Deepika Jaiman
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 UP India
| | - Arun Kumar Teotia
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 UP India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 UP India
| |
Collapse
|
527
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
528
|
Harris VK, Stark J, Vyshkina T, Blackshear L, Joo G, Stefanova V, Sara G, Sadiq SA. Phase I Trial of Intrathecal Mesenchymal Stem Cell-derived Neural Progenitors in Progressive Multiple Sclerosis. EBioMedicine 2018; 29:23-30. [PMID: 29449193 PMCID: PMC5925446 DOI: 10.1016/j.ebiom.2018.02.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system and is one of the leading causes of disability in young adults. Cell therapy is emerging as a therapeutic strategy to promote repair and regeneration in patients with disability associated with progressive MS. METHODS We conducted a phase I open-label clinical trial investigating the safety and tolerability of autologous bone marrow mesenchymal stem cell-derived neural progenitor (MSC-NP) treatment in 20 patients with progressive MS. MSC-NPs were administered intrathecally (IT) in three separate doses of up to 1 × 107 cells per dose, spaced three months apart. The primary endpoint was to assess safety and tolerability of the treatment. Expanded disability status scale (EDSS), timed 25-ft walk (T25FW), muscle strength, and urodynamic testing were used to evaluate treatment response. This trial is registered with ClinicalTrials.gov, number NCT01933802. FINDINGS IT MSC-NP treatment was safe and well tolerated. The 20 enrolled subjects completed all 60 planned treatments without serious adverse effects. Minor adverse events included transient fever and mild headaches usually resolving in <24 h. Post-treatment disability score analysis demonstrated improved median EDSS suggesting possible efficacy. Positive trends were more frequently observed in the subset of SPMS patients and in ambulatory subjects (EDSS ≤ 6.5). In addition, 70% and 50% of the subjects demonstrated improved muscle strength and bladder function, respectively, following IT MSC-NP treatment. INTERPRETATION The possible reversal of disability that was observed in a subset of patients warrants a larger phase II placebo-controlled study to establish efficacy of IT MSC-NP treatment in patients with MS. FUNDING SOURCE The Damial Foundation.
Collapse
Affiliation(s)
- Violaine K Harris
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA
| | - James Stark
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA
| | - Tamara Vyshkina
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA
| | - Leslie Blackshear
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA
| | - Gloria Joo
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA
| | | | - Gabriel Sara
- Department of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY, USA
| | - Saud A Sadiq
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, USA.
| |
Collapse
|
529
|
Zhang S, Chuah SJ, Lai RC, Hui JHP, Lim SK, Toh WS. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials 2018; 156:16-27. [DOI: 10.1016/j.biomaterials.2017.11.028] [Citation(s) in RCA: 642] [Impact Index Per Article: 91.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/17/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
|
530
|
Sato C, Yamamoto Y, Funayama E, Furukawa H, Oyama A, Murao N, Hosono H, Kawakubo K, Sakamoto N, Ohnishi S. Conditioned Medium Obtained from Amnion-Derived Mesenchymal Stem Cell Culture Prevents Activation of Keloid Fibroblasts. Plast Reconstr Surg 2018; 141:390-398. [PMID: 29369991 DOI: 10.1097/prs.0000000000004068] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenchymal stem cells are a valuable cell source in regenerative medicine, and conditioned medium obtained from mesenchymal stem cells reportedly inhibits inflammation. Keloids are characterized by abnormal fibrosis, caused by fibroblasts in response to inflammation. In this study, the authors evaluated whether conditioned medium obtained from amnion-derived mesenchymal stem cells suppressed activation of keloid fibroblasts. METHODS Keloid (n = 7), mature (n = 5), and normal (n = 5) fibroblasts were harvested from patients. Fibroblasts were stimulated with transforming growth factor (TGF)-β, and the effects of conditioned medium obtained from amnion-derived mesenchymal stem cells on cell proliferation, activation, and expression of extracellular matrix-related genes were analyzed. The effect of concentrating the conditioned medium by ultrafiltration on fibroblast activation was also analyzed. RESULTS Conditioned medium obtained from amnion-derived mesenchymal stem cells significantly up-regulated proliferation of mature fibroblasts but tended to suppress that of keloid fibroblasts. Conditioned medium obtained from amnion-derived mesenchymal stem cells significantly suppressed the TGF-β-induced up-regulation of α-smooth muscle actin in keloid and normal fibroblasts and collagen I in keloid fibroblasts, but not in mature fibroblasts. The conditioned medium obtained from amnion-derived mesenchymal stem cells concentrated by ultrafiltration and the filtrate significantly suppressed TGF-β-induced α-smooth muscle actin expression. CONCLUSION Conditioned medium obtained from amnion-derived mesenchymal stem cells prevents proliferation and activation of keloid fibroblasts and is a promising keloid treatment for administration as a topical agent. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
Affiliation(s)
- Chigusa Sato
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Yuhei Yamamoto
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Emi Funayama
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Hiroshi Furukawa
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Akihiko Oyama
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Naoki Murao
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Hidetaka Hosono
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Kazumichi Kawakubo
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Naoya Sakamoto
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shunsuke Ohnishi
- Sapporo, Japan
- From the Departments of Plastic and Reconstructive Surgery, and Gastroenterology and Hepatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| |
Collapse
|
531
|
Davis MS, Marrero-Berrios I, Perez I, Maguire T, Radhakrishnan P, Manchikalapati D, SchianodiCola J, Kamath H, Schloss RS, Yarmush J. Alginate-liposomal construct for bupivacaine delivery and MSC function regulation. Drug Deliv Transl Res 2018; 8:226-238. [PMID: 29204926 PMCID: PMC6218803 DOI: 10.1007/s13346-017-0454-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cell (MSC) therapies have become potential treatment options for multiple ailments and traumatic injuries. In the clinical setting, MSC are likely to be co-administered with local anesthetics (LA) which have been shown to have dose- and potency-dependent detrimental effects on the viability and function of cells. We previously developed and characterized a sustained-release LA delivery formulation comprised of alginate-encapsulated liposomal bupivacaine. The current studies were designed to evaluate the effect of this formulation on the secretion of three key MSC regulatory molecules, interleukin 6 (IL-6), prostaglandin E2 (PGE2), and transforming growth factor-beta 1 (TGF-β1). MSCs were treated with several bupivacaine formulations-bolus, liposome, or alginate-liposome construct (engineered construct)-in the presence or absence of inflammatory stimulus to stimulate an injured tissue environment. Our results indicated that compared to bolus or liposomal bupivacaine, the engineered construct preserved or promoted MSC anti-inflammatory PGE2 secretion; however, the engineered construct did not increase TGF-β1 secretion. Bupivacaine release profile analyses indicated that mode of drug delivery controlled the LA concentration over time and pathway analysis identified several shared and cytokine-specific molecular mediators for IL-6, PGE2, and TGF-β1 which could explain differential MSC secretion responses in the presence of bupivacaine. Collectively, these studies support the potential utility of alginate-encapsulated LA constructs for anti-inflammatory cell therapy co-administration and indicate that mode of local anesthetic delivery can significantly alter MSC secretome function.
Collapse
Affiliation(s)
- Mollie S Davis
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08805, USA
| | - Ileana Marrero-Berrios
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08805, USA
| | - Isabel Perez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08805, USA
| | - Timothy Maguire
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08805, USA
| | | | | | | | | | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08805, USA.
| | - Joel Yarmush
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, NY, USA
| |
Collapse
|
532
|
Ko DY, Patel M, Lee HJ, Jeong B. Coordinating Thermogel for Stem Cell Spheroids and Their Cyto-Effectiveness. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1706286. [DOI: 10.1002/adfm.201706286] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Du Young Ko
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| |
Collapse
|
533
|
Spasovski D, Spasovski V, Baščarević Z, Stojiljković M, Vreća M, Anđelković M, Pavlović S. Intra-articular injection of autologous adipose-derived mesenchymal stem cells in the treatment of knee osteoarthritis. J Gene Med 2018; 20. [PMID: 29243283 DOI: 10.1002/jgm.3002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/04/2017] [Accepted: 12/04/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative joint disease and is considered to be the fourth leading cause of disability and the second cause of inability to work in men. Recently, adipose-derived mesenchymal stem cells (AD-MSCs) came into focus for regenerative medicine as a promising tool for the treatment of OA. The administration of stem cells into impaired joints results in pain relief and improves quality of life, accompanied by restoration of hyaline articular cartilage. METHODS In the present study, nine patients (including two patients with bilateral symptoms) diagnosed with osteoarthritis (International Knee Documentation grade B in 5 and grade D in six knees) were treated using a single injection of AD-MSCs at a concentration of 0.5-1.0 × 107 cells and were followed up for 18 months. During follow-up, all the cases were evaluated clinically by Knee Society score (KSS), Hospital for Special Surgery knee score (HSS-KS), Tegner-Lysholm (T-L) score and visual analogue scale (VAS) of pain, as well as by plain radiography and by magnetic resonance imaging visualization with 2D Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score assessment. RESULTS Significant improvement of all four clinical scores was observed within the first 6 months (KSS for 41.4 points, HSS-KS for 33.9 points, T-L score for 44.8 points, VAS of pain from 54.5 to 9.3) and improvement persisted throughout the rest of the follow-up. MOCART score showed significant cartilage restoration (from 43 ± 7.2 to 63 ± 17.1), whereas radiography showed neither improvement, nor further joint degeneration. CONCLUSIONS The results obtained in the present study provide good basis for prospective randomized controlled clinical trials with respect to the use of AD-MSCs in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Duško Spasovski
- Institute for Orthopaedic Surgery 'Banjica', Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Spasovski
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Zoran Baščarević
- Institute for Orthopaedic Surgery 'Banjica', Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Maja Stojiljković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miša Vreća
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marina Anđelković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Sonja Pavlović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
534
|
Regeneration of functional alveoli by adult human SOX9 + airway basal cell transplantation. Protein Cell 2018; 9:267-282. [PMID: 29344809 PMCID: PMC5829276 DOI: 10.1007/s13238-018-0506-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/24/2017] [Indexed: 12/04/2022] Open
Abstract
Irreversible destruction of bronchi and alveoli can lead to multiple incurable lung diseases. Identifying lung stem/progenitor cells with regenerative capacity and utilizing them to reconstruct functional tissue is one of the biggest hopes to reverse the damage and cure such diseases. Here we showed that a rare population of SOX9+ basal cells (BCs) located at airway epithelium rugae can regenerate adult human lung. Human SOX9+ BCs can be readily isolated by bronchoscopic brushing and indefinitely expanded in feeder-free condition. Expanded human SOX9+ BCs can give rise to alveolar and bronchiolar epithelium after being transplanted into injured mouse lung, with air-blood exchange system reconstructed and recipient’s lung function improved. Manipulation of lung microenvironment with Pirfenidone to suppress TGF-β signaling could further boost the transplantation efficiency. Moreover, we conducted the first autologous SOX9+ BCs transplantation clinical trial in two bronchiectasis patients. Lung tissue repair and pulmonary function enhancement was observed in patients 3–12 months after cell transplantation. Altogether our current work indicated that functional adult human lung structure can be reconstituted by orthotopic transplantation of tissue-specific stem/progenitor cells, which could be translated into a mature regenerative therapeutic strategy in near future.
Collapse
|
535
|
Najar M, Dollé L, Crompot E, Verhulst S, van Grunsven LA, Busser H, Lagneaux L. Isolation and Characterization of Bone Marrow Mesenchymal Stromal Cell Subsets in Culture Based on Aldehyde Dehydrogenase Activity. Tissue Eng Part C Methods 2018; 24:89-98. [PMID: 29241418 DOI: 10.1089/ten.tec.2017.0312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have particular properties that allow their use as therapeutic strategies for several cell-based applications. Historically, bone marrow (BM)-MSCs are isolated by culture adherence since specific cell surface markers are yet to be developed. This original work aimed to identify and characterize isolating expanded BM-MSCs based on their aldehyde dehydrogenase (ALDH) activity known to be a hallmark of stem cells and relevant for their isolation. We thus isolated by fluorescence-activated cell sorting technology two functionally different populations of BM-MSCs depending on their ALDH activity (ALDH+ and ALDH-). Transcriptome analysis and profiling clearly demonstrated that both populations of BM-MSCs present distinct pattern of genes related to the main properties of MSCs (proliferation, response to hypoxia, angiogenesis, phenotype, stemness, multilineage, hematopoiesis, immunomodulation) in an ALDH activity dependent manner. Both BM-MSC populations look to significantly differ in terms of biological responses and functionalities. More functional analyses are needed to understand and characterize the properties of these ALDH populations. Collectively, our results highlight ALDH activity as a potential feature for isolating and segregating functional and/or competent subset of BM-MSC populations, which may account for better and more efficient therapeutic issue.
Collapse
Affiliation(s)
- Mehdi Najar
- 1 Department of Haematology, Laboratory of Clinical Cell Therapy (LTCC), Jules Bordet Institute , Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Brussels, Belgium
| | - Laurent Dollé
- 2 Department of Cell Biology (CYTO-VUB), Liver Cell Biology Laboratory, Vrije Universiteit Brussel , Brussels, Belgium
| | - Emerence Crompot
- 1 Department of Haematology, Laboratory of Clinical Cell Therapy (LTCC), Jules Bordet Institute , Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Brussels, Belgium
| | - Stefaan Verhulst
- 2 Department of Cell Biology (CYTO-VUB), Liver Cell Biology Laboratory, Vrije Universiteit Brussel , Brussels, Belgium
| | - Leo A van Grunsven
- 2 Department of Cell Biology (CYTO-VUB), Liver Cell Biology Laboratory, Vrije Universiteit Brussel , Brussels, Belgium
| | - Hélène Busser
- 1 Department of Haematology, Laboratory of Clinical Cell Therapy (LTCC), Jules Bordet Institute , Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Brussels, Belgium
| | - Laurence Lagneaux
- 1 Department of Haematology, Laboratory of Clinical Cell Therapy (LTCC), Jules Bordet Institute , Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Brussels, Belgium
| |
Collapse
|
536
|
Kim DS, Jang IK, Lee MW, Ko YJ, Lee DH, Lee JW, Sung KW, Koo HH, Yoo KH. Enhanced Immunosuppressive Properties of Human Mesenchymal Stem Cells Primed by Interferon-γ. EBioMedicine 2018; 28:261-273. [PMID: 29366627 PMCID: PMC5898027 DOI: 10.1016/j.ebiom.2018.01.002] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/02/2018] [Accepted: 01/02/2018] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of particular interest for the treatment of immune-related diseases owing to their immunosuppressive properties. In this study, we aimed to identify the effect of interferon (IFN)-γ priming on immunomodulation by MSCs and elucidate the possible mechanism underlying their properties for the clinical treatment of allogeneic conflicts. Infusion of MSCs primed with IFN-γ significantly reduced the symptoms of graft-versus-host disease (GVHD) in NOD-SCID mice, thereby increasing survival rate when compared with naïve MSC-infused mice. However, infusion of IFN-γ-primed MSCs in which indoleamine 2,3-dioxygenase (IDO) was downregulated did not elicit this effect. The IDO gene was expressed in MSCs via the IFN-γ-Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) pathway, and the infusion of IDO-over-expressing MSCs increased survival rate in an in vivo GVHD model, similar to infusion of IFN-γ-primed MSCs. These data indicate that IFN-γ production by activated T-cells is correlated with the induction of IDO expression in MSCs via the IFN-γ-JAK-STAT1 pathway, which in turn results in the suppression of T-cell proliferation. Our findings also suggest that cell therapy based on MSCs primed with IFN-γ can be used for the clinical treatment of allogeneic conflicts, including GVHD. IFN-γ priming enhances the immunosuppressive properties of human MSCs in in vitro and in vivo models. IFN-γ priming induces IDO expression in MSCs via the JAK/STAT1 signaling pathway, but TLR3 activation does not. Cell therapy using MSCs primed with IFN-γ could be highly effective in treating allogeneic diseases, including GVHD.
It is necessary to improve the function of mesenchymal stem cells (MSCs) to maximize their treatment potential beyond what is currently achieved in cell therapy studies using naïve heterogeneous MSCs. The preclinical study of a candidate cell therapy based on MSCs primed with interferon-γ as reported in this study, could lay the foundation for the use of cell therapy for the treatment of graft-versus-host disease (GVHD), and is very important for the initiation of clinical trials. Our findings also suggest that cell therapy based on functionally improved MSCs could be used for the clinical treatment of allogeneic conflicts.
Collapse
Affiliation(s)
- Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - In Keun Jang
- Biomedical Research Institute, LIFELIVER. Co., LTD., Yongin, Gyeonggi-do, Republic of Korea
| | - Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.
| | - Young Jong Ko
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Doo-Hoon Lee
- Biomedical Research Institute, LIFELIVER. Co., LTD., Yongin, Gyeonggi-do, Republic of Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea; Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
537
|
Yan Z, Yin H, Nerlich M, Pfeifer CG, Docheva D. Boosting tendon repair: interplay of cells, growth factors and scaffold-free and gel-based carriers. J Exp Orthop 2018; 5:1. [PMID: 29330711 PMCID: PMC5768579 DOI: 10.1186/s40634-017-0117-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022] Open
Abstract
Background Tendons are dense connective tissues and critical components for the integrity and function of the musculoskeletal system. Tendons connect bone to muscle and transmit forces on which locomotion entirely depends. Due to trauma, overuse and age-related degeneration, many people suffer from acute or chronic tendon injuries. Owing to their hypovascularity and hypocellularity, tendinopathies remain a substantial challenge for both clinicians and researchers. Surgical treatment includes suture or transplantation of autograft, allograft or xenograft, and these serve as the most common technique for rescuing tendon injuries. However, the therapeutic efficacies are limited by drawbacks including inevitable donor site morbidity, poor graft integration, adhesion formations and high rates of recurrent tearing. This review summarizes the literature of the past 10 y concerning scaffold-free and gel-based approaches for treating tendon injuries, with emphasis on specific advantages of such modes of application, as well as the obtained results regarding in vitro and in vivo tenogenesis. Results The search was focused on publications released after 2006 and 83 articles have been analysed. The main results are summarizing and discussing the clear advantages of scaffold-free and hydrogels carriers that can be functionalized with cells alone or in combination with growth factors. Conclusion The improved understanding of tissue resident adult stem cells has made a significant progress in recent years as well as strategies to steer their fate toward tendon lineage, with the help of growth factors, have been identified. The field of tendon tissue engineering is exploring diverse models spanning from hard scaffolds to gel-based and scaffold-free approaches seeking easier cell delivery and integration in the site of injury. Still, the field needs to consider a multifactorial approach that is based on the combination and fine-tuning of chemical and biomechanical stimuli. Taken together, tendon tissue engineering has now excellent foundations and enters the period of precision and translation to models with clinical relevance on which better treatment options of tendon injuries can be shaped up.
Collapse
Affiliation(s)
- Zexing Yan
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Heyong Yin
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Michael Nerlich
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Christian G Pfeifer
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Denitsa Docheva
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany. .,Director of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
538
|
Exploring Stem Cells and Inflammation in Tendon Repair and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1089:37-46. [PMID: 30088259 DOI: 10.1007/5584_2018_258] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tendon injuries are frequent and are responsible for substantial morbidity both in sports and in the workplace. Despite the endogenous mechanisms of tendon repair and regeneration, tendon healing upon injury is slow and often insufficient to restore complete biomechanics functionality.Inflammation has a pivotal role in tendon healing and failed healing responses contribute to the progression of tendinopathies. However, the molecular and cellular mechanisms involved are poorly understood requiring further insights.During inflammation, bioactive molecules such as cytokines secreted locally at the injury site, influence resident stem cells that contribute as modulatory agents over the niche towards homeostasis, holding great promise as therapeutic agents for tendon pathological conditions associated to unresolved inflammation and failed healing.This review overviews the role of cytokines and resident cells, focusing on the participation of tendon stem cell population in inflammation and tendon healing upon injury and their potential action in resolution of pathological conditions.
Collapse
|
539
|
Emerging Concepts in Treating Cartilage, Osteochondral Defects, and Osteoarthritis of the Knee and Ankle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:25-62. [PMID: 29736568 DOI: 10.1007/978-3-319-76735-2_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The management and treatment of cartilage lesions, osteochondral defects, and osteoarthritis remain a challenge in orthopedics. Moreover, these entities have different behaviors in different joints, such as the knee and the ankle, which have inherent differences in function, biology, and biomechanics. There has been a huge development on the conservative treatment (new technologies including orthobiologics) as well as on the surgical approach. Some surgical development upraises from technical improvements including advanced arthroscopic techniques but also from increased knowledge arriving from basic science research and tissue engineering and regenerative medicine approaches. This work addresses the state of the art concerning basic science comparing the knee and ankle as well as current options for treatment. Furthermore, the most promising research developments promising new options for the future are discussed.
Collapse
|
540
|
Cavallini C, Zannini C, Olivi E, Tassinari R, Taglioli V, Rossi M, Poggi P, Chatgilialoglu A, Simonazzi G, Alviano F, Bonsi L, Ventura C. Restoring In Vivo-Like Membrane Lipidomics Promotes Exosome Trophic Behavior from Human Placental Mesenchymal Stromal/Stem Cells. Cell Transplant 2018; 27:55-69. [PMID: 29562775 PMCID: PMC6434476 DOI: 10.1177/0963689717723016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/19/2017] [Accepted: 06/16/2017] [Indexed: 12/20/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an effective tool in regenerative medicine notably for their intrinsic plentiful paracrine activity rather than differentiating properties. The hMSC secretome includes a wide spectrum of regulatory and trophic factors, encompassing several naked molecules as well as different kinds of extracellular vesicles (EVs). Among EVs, exosomes represent an intriguing population, able to shuttle proteins, transcription factors, and genetic materials, with a relevant role in cell-to-cell communication, modulating biological responses in recipient cells. In this context, the extracellular milieu can greatly impact the paracrine activity of stem cells, modifying their metabolism, and the dynamics of vesicle secretion. In the present study, we investigated the effects elicited on exosome patterning by tailored, ad hoc formulated lipid supplementation (Refeed®) in MSCs derived from human fetal membranes (hFM-MSCs). Wound healing experiments revealed that stem cell exposure to exosomes obtained from Refeed®-supplemented hFM-MSCs increased their migratory capability, although the amount of exosomes released after Refeed® supplementation was lower than that yielded from non-supplemented cells. We found that such a decrease was mainly due to a different rate of exosomal exocytosis rather than to an effect of the lipid supplement on the endocytic pathway. Endoplasmic reticulum homeostasis was modified by supplementation, through the upregulation of PKR-like ER kinase (PERK) and inositol-requiring enzyme 1α (IRE1α). Increased expression of these proteins did not lead to stress-induced, unfolded protein response (UPR)-mediated apoptosis, nor did it affect phosphorylation of p38 kinase, suggesting that PERK and IRE1α overexpression was due to augmented metabolic activities mediated by optimization of a cellular feeding network afforded through lipid supplementation. In summary, these results demonstrate how tailored lipid supplementation can successfully modify the paracrine features in hFM-MSCs, impacting both intracellular vesicle trafficking and secreted exosome number and function.
Collapse
Affiliation(s)
- Claudia Cavallini
- GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), Rome, Italy
- Ettore Sansavini Health Science Foundation ONLUS—Lab SWITH, Lugo, Italy
| | - Chiara Zannini
- Ettore Sansavini Health Science Foundation ONLUS—Lab SWITH, Lugo, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Nephrology, Dialysis and Renal Transplant, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elena Olivi
- GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), Rome, Italy
- Ettore Sansavini Health Science Foundation ONLUS—Lab SWITH, Lugo, Italy
| | - Riccardo Tassinari
- GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), Rome, Italy
- Ettore Sansavini Health Science Foundation ONLUS—Lab SWITH, Lugo, Italy
| | - Valentina Taglioli
- National Institute of Biostructures and Biosystems (NIBB), Rome, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, Laboratory of Experimental Cardiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Martina Rossi
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | | | | | - Giuliana Simonazzi
- Division of Obstetrics and Prenatal Medicine, Department of Medical and Surgical Sciences, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Laura Bonsi
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Carlo Ventura
- GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), Rome, Italy
- CNR, Institute of Organic Synthesis and Photoreactivity (Istituto per la Sintesi Organica e la Fotoreattività ISOF), Via Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
541
|
Rahmati M, Pennisi CP, Mobasheri A, Mozafari M. Bioengineered Scaffolds for Stem Cell Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018:73-89. [DOI: 10.1007/5584_2018_215] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
542
|
Therapeutic Benefit for Late, but Not Early, Passage Mesenchymal Stem Cells on Pain Behaviour in an Animal Model of Osteoarthritis. Stem Cells Int 2017; 2017:2905104. [PMID: 29434641 PMCID: PMC5757143 DOI: 10.1155/2017/2905104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/07/2017] [Indexed: 12/29/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have a therapeutic potential for the treatment of osteoarthritic (OA) joint pathology and pain. The aims of this study were to determine the influence of a passage number on the effects of MSCs on pain behaviour and cartilage and bone features in a rodent model of OA. Methods Rats underwent either medial meniscal transection (MNX) or sham surgery under anaesthesia. Rats received intra-articular injection of either 1.5 × 106 late passage MSCs labelled with 10 μg/ml SiMAG, 1.5 × 106 late passage mesenchymal stem cells, the steroid Kenalog (200 μg/20 μL), 1.5 × 106 early passage MSCs, or serum-free media (SFM). Sham-operated rats received intra-articular injection of SFM. Pain behaviour was quantified until day 42 postmodel induction. Magnetic resonance imaging (MRI) was used to localise the labelled cells within the knee joint. Results Late passage MSCs and Kenalog attenuated established pain behaviour in MNX rats, but did not alter MNX-induced joint pathology at the end of the study period. Early passage MSCs exacerbated MNX-induced pain behaviour for up to one week postinjection and did not alter joint pathology. Conclusion Our data demonstrate for the first time the role of a passage number in influencing the therapeutic effects of MSCs in a model of OA pain.
Collapse
|
543
|
Berglund AK, Fortier LA, Antczak DF, Schnabel LV. Immunoprivileged no more: measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell Res Ther 2017; 8:288. [PMID: 29273086 PMCID: PMC5741939 DOI: 10.1186/s13287-017-0742-8] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Autologous and allogeneic adult mesenchymal stem/stromal cells (MSCs) are increasingly being investigated for treating a wide range of clinical diseases. Allogeneic MSCs are especially attractive due to their potential to provide immediate care at the time of tissue injury or disease diagnosis. The prevailing dogma has been that allogeneic MSCs are immune privileged, but there have been very few studies that control for matched or mismatched major histocompatibility complex (MHC) molecule expression and that examine immunogenicity in vivo. Studies that control for MHC expression have reported both cell-mediated and humoral immune responses to MHC-mismatched MSCs. The clinical implications of immune responses to MHC-mismatched MSCs are still unknown. Pre-clinical and clinical studies that document the MHC haplotype of donors and recipients and measure immune responses following MSC treatment are necessary to answer this critical question. Conclusions This review details what is currently known about the immunogenicity of allogeneic MSCs and suggests contemporary assays that could be utilized in future studies to appropriately identify and measure immune responses to MHC-mismatched MSCs.
Collapse
Affiliation(s)
- Alix K Berglund
- Department of Clinical Sciences, College of Veterinary Medicine and the Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.
| | - Lisa A Fortier
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Douglas F Antczak
- Baker Institute for Animal Health, Cornell University, Ithaca, NY, 14853, USA
| | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine and the Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.
| |
Collapse
|
544
|
Huntsman HD, Rendeiro C, Merritt JR, Pincu Y, Cobert A, De Lisio M, Kolyvas E, Dvoretskiy S, Dobrucki IT, Kemkemer R, Jensen T, Dobrucki LW, Rhodes JS, Boppart MD. The impact of mechanically stimulated muscle-derived stromal cells on aged skeletal muscle. Exp Gerontol 2017; 103:35-46. [PMID: 29269268 DOI: 10.1016/j.exger.2017.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/16/2017] [Accepted: 12/14/2017] [Indexed: 01/06/2023]
Abstract
Perivascular stromal cells, including mesenchymal stem/stromal cells (MSCs), secrete paracrine factor in response to exercise training that can facilitate improvements in muscle remodeling. This study was designed to test the capacity for muscle-resident MSCs (mMSCs) isolated from young mice to release regenerative proteins in response to mechanical strain in vitro, and subsequently determine the extent to which strain-stimulated mMSCs can enhance skeletal muscle and cognitive performance in a mouse model of uncomplicated aging. Protein arrays confirmed a robust increase in protein release at 24h following an acute bout of mechanical strain in vitro (10%, 1Hz, 5h) compared to non-strain controls. Aged (24month old), C57BL/6 mice were provided bilateral intramuscular injection of saline, non-strain control mMSCs, or mMSCs subjected to a single bout of mechanical strain in vitro (4×104). No significant changes were observed in muscle weight, myofiber size, maximal force, or satellite cell quantity at 1 or 4wks between groups. Peripheral perfusion was significantly increased in muscle at 4wks post-mMSC injection (p<0.05), yet no difference was noted between control and preconditioned mMSCs. Intramuscular injection of preconditioned mMSCs increased the number of new neurons and astrocytes in the dentate gyrus of the hippocampus compared to both control groups (p<0.05), with a trend toward an increase in water maze performance noted (p=0.07). Results from this study demonstrate that acute injection of exogenously stimulated muscle-resident stromal cells do not robustly impact aged muscle structure and function, yet increase the survival of new neurons in the hippocampus.
Collapse
Affiliation(s)
- Heather D Huntsman
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Catarina Rendeiro
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Nutrition, Learning and Memory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Jennifer R Merritt
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Psychology and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yair Pincu
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Adam Cobert
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Psychology and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Emily Kolyvas
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Svyatoslav Dvoretskiy
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Iwona T Dobrucki
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ralf Kemkemer
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Tor Jensen
- Division of Biomedical Sciences, Carle Hospital, Urbana, IL 61801, USA
| | - Lawrence W Dobrucki
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Justin S Rhodes
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Nutrition, Learning and Memory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Psychology and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Nutrition, Learning and Memory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
545
|
Teixeira FG, Serra SC, Salgado AJ. Tips on How to Collect and Administer the Mesenchymal Stem Cell Secretome for Central Nervous System Applications. Methods Mol Biol 2017; 1416:457-65. [PMID: 27236689 DOI: 10.1007/978-1-4939-3584-0_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have been proposed as possible therapeutic agents for central nervous system (CNS) disorders. Recently, it has been suggested that their effects are mostly mediated through their secretome, which contains a number of neuroregulatory molecules capable of increasing cell proliferation, differentiation, and survival in different physiological conditions. Here, we present an overview of the hMSC secretome as a possible candidate in the creation of new cell-free therapies, demonstrating the process of its collection and route of administration, focusing our attention on their effects in CNS regenerative medicine.
Collapse
Affiliation(s)
- F G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S C Serra
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
546
|
Yao Y, Zheng Z, Song Q. Mesenchymal stem cells: A double-edged sword in radiation-induced lung injury. Thorac Cancer 2017; 9:208-217. [PMID: 29235254 PMCID: PMC5792737 DOI: 10.1111/1759-7714.12573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/06/2023] Open
Abstract
Radiation therapy is an important treatment modality for multiple thoracic malignancies. However, radiation‐induced lung injury (RILI), which is the term generally used to describe damage to the lungs caused by exposure to ionizing radiation, remains a critical issue affecting both tumor control and patient quality of life. Despite tremendous effort, there is no current consensus regarding the optimal treatment approach for RILI. Because of a number of functional advantages, including self‐proliferation, multi‐differentiation, injury foci chemotaxis, anti‐inflammation, and immunomodulation, mesenchymal stem cells (MSCs) have been a focus of research for many years. Accumulating evidence indicates the therapeutic potential of transplantation of MSCs derived from adipose tissue, umbilical cord blood, and bone marrow for inflammatory diseases, including RILI. However, reports have also shown that MSCs, including fibrocytes, lung hematopoietic progenitor cells, and ABCG2+ MSCs, actually enhance the progression of lung injuries. These contradictory results suggest that MSCs may have dual effects and that caution should be taken when using MSCs to treat RILI. In this review, we present and discuss recent evidence of the double‐edged function of MSCs and provide comments on the prospects of these findings.
Collapse
Affiliation(s)
- Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongliang Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
547
|
Zeppieri M, Salvetat ML, Beltrami A, Cesselli D, Russo R, Alcalde I, Merayo-Lloves J, Brusini P, Parodi PC. Adipose Derived Stem Cells for Corneal Wound Healing after Laser Induced Corneal Lesions in Mice. J Clin Med 2017; 6:115. [PMID: 29206194 PMCID: PMC5742804 DOI: 10.3390/jcm6120115] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/24/2017] [Accepted: 11/30/2017] [Indexed: 02/05/2023] Open
Abstract
The aim of our study was to assess the clinical effectiveness of topical adipose derived stem cell (ADSC) treatment in laser induced corneal wounds in mice by comparing epithelial repair, inflammation, and histological analysis between treatment arms. Corneal lesions were performed on both eyes of 40 mice by laser induced photorefractive keratectomy. All eyes were treated with topical azythromycin bid for three days. Mice were divided in three treatment groups (n = 20), which included: control, stem cells and basic serum; which received topical treatment three times daily for five consecutive days. Biomicroscope assessments and digital imaging were performed by two masked graders at 30, 54, 78, 100, and 172 h to analyze extent of fluorescein positive epithelial defect, corneal inflammation, etc. Immunohistochemical techniques were used in fixed eyes to assess corneal repair markers Ki67, α Smooth Muscle Actin (α-SMA) and E-Cadherin. The fluorescein positive corneal lesion areas were significantly smaller in the stem cells group on days 1 (p < 0.05), 2 (p < 0.02) and 3. The stem cell treated group had slightly better and faster re-epithelization than the serum treated group in the initial phases. Comparative histological data showed signs of earlier and better corneal repair in epithelium and stromal layers in stem cell treated eyes, which showed more epithelial layers and enhanced wound healing performance of Ki67, E-Cadherin, and α-SMA. Our study shows the potential clinical and histological advantages in the topical ADSC treatment for corneal lesions in mice.
Collapse
Affiliation(s)
- Marco Zeppieri
- Department of Ophthalmology, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| | - Maria Letizia Salvetat
- Department of Ophthalmology, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| | - Antonio Beltrami
- Department of Pathology, University of Udine, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| | - Daniela Cesselli
- Department of Pathology, University of Udine, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| | - Rossella Russo
- Department of Pharmacobiology, University of Calabria, Cosenza 87036, Italy.
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, Oviedo 33006, Spain.
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, Oviedo 33006, Spain.
| | - Paolo Brusini
- Department of Ophthalmology, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University of Udine, Azienda Ospedaliero Universitaria Santa Maria della Misericordia, Udine 33100, Italy.
| |
Collapse
|
548
|
Mesenchymal Stem Cell Benefits Observed in Bone Marrow Failure and Acquired Aplastic Anemia. Stem Cells Int 2017; 2017:8076529. [PMID: 29333168 PMCID: PMC5733198 DOI: 10.1155/2017/8076529] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/15/2017] [Accepted: 09/10/2017] [Indexed: 12/16/2022] Open
Abstract
Acquired aplastic anemia (AA) is a type of bone marrow failure (BMF) syndrome characterized by partial or total bone marrow (BM) destruction resulting in peripheral blood (PB) pancytopenia, which is the reduction in the number of red blood cells (RBC) and white blood cells (WBC), as well as platelets (PLT). The first-line treatment option of AA is given by hematopoietic stem cell (HSCs) transplant and/or immunosuppressive (IS) drug administration. Some patients did not respond to the treatment and remain pancytopenic following IS drugs. The studies are in progress to test the efficacy of adoptive cellular therapies as mesenchymal stem cells (MSCs), which confer low immunogenicity and are reliable allogeneic transplants in refractory severe aplastic anemia (SAA) cases. Moreover, bone marrow stromal cells (BMSC) constitute an essential component of the hematopoietic niche, responsible for stimulating and enhancing the proliferation of HSCs by secreting regulatory molecules and cytokines, providing stimulus to natural BM microenvironment for hematopoiesis. This review summarizes scientific evidences of the hematopoiesis improvements after MSC transplant, observed in acquired AA/BMF animal models as well as in patients with acquired AA. Additionally, we discuss the direct and indirect contribution of MSCs to the pathogenesis of acquired AA.
Collapse
|
549
|
Radwan RR, Mohamed HA. Nigella sativa oil modulates the therapeutic efficacy of mesenchymal stem cells against liver injury in irradiated rats. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 178:447-456. [PMID: 29216568 DOI: 10.1016/j.jphotobiol.2017.11.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/17/2017] [Accepted: 11/27/2017] [Indexed: 01/05/2023]
Abstract
Stem cell transplantation is a novel strategy for regenerative medicine in liver disease. This study was conducted to explore the modulatory effect of Nigella sativa oil (NSO) on the therapeutic potential of mesenchymal stem cells (MSCs) against irradiation-induced liver damage in rats. Liver damage was induced by a total body exposure to a single dose of 7Gy. NSO (2mg/kg/day) was then given orally for 4 consecutive weeks starting 24h after irradiation with or without a single intravenous MSCs administration, then rats were sacrificed four weeks after exposure to γ radiation. Data revealed that irradiation elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities in serum, increased hepatic malondialdehyde (MDA) content and reduced hepatic superoxide dismutase (SOD) activity. Furthermore, it caused elevation in pro-inflammatory mediators such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) associated with reduction in anti-inflammatory cytokine interleukin-10 (IL-10) and it increased fibrogenic marker transforming growth factor-β (TGF-β) in liver tissues. It was observed that combined NSO/MSCs therapy provided more beneficial tissue repair comparable to MSCs alone as demonstrated by modulating the tested parameters. Finally, these results were confirmed by histopathological examination. In conclusion, dual therapy with NSO and MSCs could serve as a promising approach for alleviating radiation-induced liver injury in patients with radiotherapy.
Collapse
Affiliation(s)
- Rasha R Radwan
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), PO Box 29, Nasr City, Cairo, Egypt.
| | - Heba A Mohamed
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), PO Box 29, Nasr City, Cairo, Egypt
| |
Collapse
|
550
|
Tribe HC, McEwan J, Taylor H, Oreffo ROC, Tare RS. Mesenchymal Stem Cells: Potential Role in the Treatment of Osteochondral Lesions of the Ankle. Biotechnol J 2017; 12:1700070. [PMID: 29068173 PMCID: PMC5765412 DOI: 10.1002/biot.201700070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Given articular cartilage has a limited repair potential, untreated osteochondral lesions of the ankle can lead to debilitating symptoms and joint deterioration necessitating joint replacement. While a wide range of reparative and restorative surgical techniques have been developed to treat osteochondral lesions of the ankle, there is no consensus in the literature regarding which is the ideal treatment. Tissue engineering strategies, encompassing stem cells, somatic cells, biomaterials, and stimulatory signals (biological and mechanical), have a potentially valuable role in the treatment of osteochondral lesions. Mesenchymal stem cells (MSCs) are an attractive resource for regenerative medicine approaches, given their ability to self-renew and differentiate into multiple stromal cell types, including chondrocytes. Although MSCs have demonstrated significant promise in in vitro and in vivo preclinical studies, their success in treating osteochondral lesions of the ankle is inconsistent, necessitating further clinical trials to validate their application. This review highlights the role of MSCs in cartilage regeneration and how the application of biomaterials and stimulatory signals can enhance chondrogenesis. The current treatments for osteochondral lesions of the ankle using regenerative medicine strategies are reviewed to provide a clinical context. The challenges for cartilage regeneration, along with potential solutions and safety concerns are also discussed.
Collapse
Affiliation(s)
- Howard C. Tribe
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Josephine McEwan
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Heath Taylor
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Rahul S. Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Bioengineering Science, Mechanical Engineering DepartmentFaculty of Engineering and the EnvironmentUniversity of SouthamptonSouthamptonSO17 1BJUK
| |
Collapse
|