1
|
Gabr MM, El-Halawani SM, Refaie AF, Khater SM, Ismail AM, Karras MS, Magar RW, Sayed SE, Kloc M, Uosef A, Sabek OM, Ghoneim MA. Modulation of naïve mesenchymal stromal cells by extracellular vesicles derived from insulin-producing cells: an in vitro study. Sci Rep 2024; 14:17844. [PMID: 39090166 PMCID: PMC11294623 DOI: 10.1038/s41598-024-68104-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
This study was to determine whether extracellular vesicles (EVs) derived from insulin-producing cells (IPCs) can modulate naïve mesenchymal stromal cells (MSCs) to become insulin-secreting. MSCs were isolated from human adipose tissue. The cells were then differentiated to generate IPCs by achemical-based induction protocol. EVs were retrieved from the conditioned media of undifferentiated (naïve) MSCs (uneducated EVs) and from that of MSC-derived IPCs (educated EVs) by sequential ultracentrifugation. The obtained EVs were co-cultured with naïve MSCs.The cocultured cells were evaluated by immunofluorescence, flow cytometry, C-peptide nanogold silver-enhanced immunostaining, relative gene expression and their response to a glucose challenge.Immunostaining for naïve MSCs cocultured with educated EVs was positive for insulin, C-peptide, and GAD65. By flow cytometry, the median percentages of insulin-andC-peptide-positive cells were 16.1% and 14.2% respectively. C-peptide nanogoldimmunostaining providedevidence for the intrinsic synthesis of C-peptide. These cells released increasing amounts of insulin and C-peptide in response to increasing glucose concentrations. Gene expression of relevant pancreatic endocrine genes, except for insulin, was modest. In contrast, the results of naïve MSCs co-cultured with uneducated exosomes were negative for insulin, C-peptide, and GAD65. These findings suggest that this approach may overcome the limitations of cell therapy.
Collapse
Affiliation(s)
- Mahmoud M Gabr
- Biotechnology Department, Urology and Nephrology Center, Mansoura, Egypt
| | | | - Ayman F Refaie
- Nephrology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Sherry M Khater
- Pathology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Amani M Ismail
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Mary S Karras
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Raghda W Magar
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Shorouk El Sayed
- Microbiology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
- Department of Genetics, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ahmed Uosef
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Omaima M Sabek
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | | |
Collapse
|
2
|
Lyu Z, Xin M, Oyston DR, Xue T, Kang H, Wang X, Wang Z, Li Q. Cause and consequence of heterogeneity in human mesenchymal stem cells: Challenges in clinical application. Pathol Res Pract 2024; 260:155354. [PMID: 38870711 DOI: 10.1016/j.prp.2024.155354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
Human mesenchymal stem cells (hMSCs) are mesoderm-derived adult stem cells with self-proliferation capacity, pluripotent differentiation potency, and excellent histocompatibility. These advantages make hMSCs a promising tool in clinical application. However, the majority of clinical trials using hMSC therapy for diverse human diseases do not achieve expectations, despite the prospective pre-clinical outcomes in animal models. This is partly attributable to the intrinsic heterogeneity of hMSCs. In this review, the cause of heterogeneity in hMSCs is systematically discussed at multiple levels, including isolation methods, cultural conditions, donor-to-donor variation, tissue sources, intra-tissue subpopulations, etc. Additionally, the effect of hMSCs heterogeneity on the contrary role in tumor progression and immunomodulation is also discussed. The attempts to understand the cellular heterogeneity of hMSCs and its consequences are important in supporting and improving therapeutic strategies for hMSCs.
Collapse
Affiliation(s)
- Zhao Lyu
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Miaomiao Xin
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shaanxi, China; University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Vodnany, Czech Republic
| | - Dale Reece Oyston
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool, UK
| | - Tingyu Xue
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Hong Kang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Xiangling Wang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Zheng Wang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing, Sichuan, China.
| | - Qian Li
- Changsha Medical University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Ning M, Hua S, Ma Y, Liu Y, Wang D, Xu K, Yu H. Microvesicles facilitate the differentiation of mesenchymal stem cells into pancreatic beta-like cells via miR-181a-5p/150-5p. Int J Biol Macromol 2024; 254:127719. [PMID: 37918601 DOI: 10.1016/j.ijbiomac.2023.127719] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
Transplantation of pancreatic islet cells is a promising strategy for the long-term treatment of type 1 diabetes (T1D). The stem cell-derived beta cells showed great potential as substitute sources of transplanted pancreatic islet cells. However, the current efficiency of stem cell differentiation still cannot match the requirements for clinical transplantation. Here, we report that microvesicles (MVs) from insulin-producing INS-1 cells could induce mesenchymal stem cell (MSC) differentiation into pancreatic beta-like cells. The combination of MVs with small molecules, nicotinamide and insulin-transferrin-selenium (ITS), dramatically improved the efficiency of MSC differentiation. Notably, the function of MVs in MSC differentiation requires their entry into MSCs through giant pinocytosis. The MVs-treated or MVs combined with small molecules-treated MSCs show pancreatic beta-like cell morphology and response to glucose stimulation in insulin secretion. Using high throughput small RNA-sequencing, we found that MVs induced MSC differentiation into the beta-like cells through miR-181a-5p/150-5p. Together, our findings reveal the role of MVs or the MV-enriched miR-181a-5p/150-5p as a class of biocompatible reagents to differentiate MSCs into functional beta-like cells and demonstrate that the combined usage of MVs or miR-181a-5p/150-5p with small molecules can potentially be used in making pancreatic islet cells for future clinical purposes.
Collapse
Affiliation(s)
- Mingming Ning
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shanshan Hua
- Department of Spine Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266071, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yunpeng Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Dianliang Wang
- Stem cell and tissue engineering research laboratory, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China.
| | - Kai Xu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
4
|
Lopes D, Lopes J, Pereira-Silva M, Peixoto D, Rabiee N, Veiga F, Moradi O, Guo ZH, Wang XD, Conde J, Makvandi P, Paiva-Santos AC. Bioengineered exosomal-membrane-camouflaged abiotic nanocarriers: neurodegenerative diseases, tissue engineering and regenerative medicine. Mil Med Res 2023; 10:19. [PMID: 37101293 PMCID: PMC10134679 DOI: 10.1186/s40779-023-00453-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
A bio-inspired strategy has recently been developed for camouflaging nanocarriers with biomembranes, such as natural cell membranes or subcellular structure-derived membranes. This strategy endows cloaked nanomaterials with improved interfacial properties, superior cell targeting, immune evasion potential, and prolonged duration of systemic circulation. Here, we summarize recent advances in the production and application of exosomal membrane-coated nanomaterials. The structure, properties, and manner in which exosomes communicate with cells are first reviewed. This is followed by a discussion of the types of exosomes and their fabrication methods. We then discuss the applications of biomimetic exosomes and membrane-cloaked nanocarriers in tissue engineering, regenerative medicine, imaging, and the treatment of neurodegenerative diseases. Finally, we appraise the current challenges associated with the clinical translation of biomimetic exosomal membrane-surface-engineered nanovehicles and evaluate the future of this technology.
Collapse
Affiliation(s)
- Daniela Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Joana Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 374-37515, Iran
| | - Zhan-Hu Guo
- Integrated Composites Laboratory (ICL), Department of Mechanical and Construction Engineering, Northumbria University, Newcastle Upon Tyne, NE1 8ST, UK
| | - Xiang-Dong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| | - João Conde
- Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
| |
Collapse
|
5
|
Extracellular Vesicles Derived from Three-Dimensional-Cultured Human Umbilical Cord Blood Mesenchymal Stem Cells Prevent Inflammation and Dedifferentiation in Pancreatic Islets. Stem Cells Int 2023; 2023:5475212. [PMID: 36860546 PMCID: PMC9970714 DOI: 10.1155/2023/5475212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 12/16/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
It is unclear whether extracellular vesicles (EVs) from mesenchymal stem cells (MSCs) have a direct protective effect on pancreatic islets. In addition, whether culturing MSCs in three dimensions (3D) instead of a monolayer (2D) can induce changes in the cargo of EVs that facilitate the polarization of macrophages into an M2 phenotype has not been investigated. We sought to determine whether EVs from MSCs cultured in 3D can prevent inflammation and dedifferentiation in pancreatic islets and, if so, whether the protective effect is superior to that of EVs from 2D MSCs. Human umbilical cord blood- (hUCB-) MSCs cultured in 3D were optimized according to cell density, exposure to hypoxia, and cytokine treatment based on the ability of the hUCB-MSC-derived EVs to induce the M2 polarization of macrophages. Islets isolated from human islet amyloid polypeptide (hIAPP) heterozygote transgenic mice were cultured in serum-deprived conditions with hUCB-MSC-derived EVs. EVs derived from 3D hUCB-MSCs had more abundant microRNAs involved in M2 polarization of macrophages and had an enhanced M2 polarization ability on macrophages, which was optimized when the 3D culture condition was 2.5 × 104 cells per spheroid without preconditioning with hypoxia and cytokine exposure. When islets isolated from hIAPP heterozygote transgenic mice were cultured in serum-deprived conditions with hUCB-MSC-derived EVs, the EVs derived from 3D hUCB-MSCs suppressed the expression of proinflammatory cytokines and caspase-1 in pancreatic islets and increased the proportion of M2-polarized islet-resident macrophages. They improved glucose-stimulated insulin secretion, reduced the expression of Oct4 and NGN3, and induced the expression of Pdx1 and FoxO1. The greater suppression of IL-1β, NLRP3 inflammasome, caspase-1, and Oct4 and induction of Pdx1 and FoxO1 were found in islets cultured with the EVs derived from 3D hUCB-MSCs. In conclusion, EVs derived from 3D hUCB-MSCs optimized for M2 polarization attenuated nonspecific inflammation and preserved β-cell identity of pancreatic islets.
Collapse
|
6
|
Hackel A, Vollmer S, Bruderek K, Lang S, Brandau S. Immunological priming of mesenchymal stromal/stem cells and their extracellular vesicles augments their therapeutic benefits in experimental graft-versus-host disease via engagement of PD-1 ligands. Front Immunol 2023; 14:1078551. [PMID: 36875112 PMCID: PMC9978482 DOI: 10.3389/fimmu.2023.1078551] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/09/2023] [Indexed: 02/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) and their extracellular vesicles (EVs) exert profound anti-inflammatory and regenerative effects in inflammation and tissue damage, which makes them an attractive tool for cellular therapies. In this study we have assessed the inducible immunoregulatory properties of MSCs and their EVs upon stimulation with different combinations of cytokines. First, we found that MSCs primed with IFN-γ, TNF-α and IL-1β, upregulate the expression of PD-1 ligands, as crucial mediators of their immunomodulatory activity. Further, primed MSCs and MSC-EVs, compared to unstimulated MSCs and MSC-EVs, had increased immunosuppressive effects on activated T cells and mediated an enhanced induction of regulatory T cells, in a PD-1 dependent manner. Importantly, EVs derived from primed MSCs reduced the clinical score and prolonged the survival of mice in a model of graft-versus-host disease. These effects could be reversed in vitro and in vivo by adding neutralizing antibodies directed against PD-L1 and PD-L2 to both, MSCs and their EVs. In conclusion, our data reveal a priming strategy that potentiates the immunoregulatory function of MSCs and their EVs. This concept also provides new opportunities to improve the clinical applicability and efficiency of cellular or EV-based therapeutic MSC products.
Collapse
Affiliation(s)
- Alexander Hackel
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Vollmer
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
7
|
Lyu Z, Li H, Li X, Wang H, Jiao H, Wang X, Zhao J, Lin H. Fibroblast growth factor 23 inhibits osteogenic differentiation and mineralization of chicken bone marrow mesenchymal stem cells. Poult Sci 2022; 102:102287. [PMID: 36442309 PMCID: PMC9706642 DOI: 10.1016/j.psj.2022.102287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23), a bone-derived hormone, is involved in the reabsorption of phosphate (P) and the production of vitamin D hormones in the kidney. However, whether and how FGF23 regulates chicken bone metabolism remains largely unknown. In the present study, we investigated the effect of FGF23 on osteogenic differentiation and mineralization of chicken bone marrow mesenchymal stem cells (BMSCs). First, we found that the transcription of FGF23 was inhibited by β-glycerophosphate sodium (GPS, 5 mM, 10 mM, 20 mM) and 10-9 M 1, 25-dihydroxyvitamin D3 (1, 25(OH)2D3), but was stimulated by 10-7 M 1, 25(OH)2D3 and parathyroid hormone (PTH, 10-9 M, 10-8 M, 10-7 M). Second, overexpression of FGF23 by the FGF23 adenovirus (Adv-FGF23) suppressed the formation of mineralized nodules (P < 0.001) and alkaline phosphatase (ALP) activity (P < 0.05) in both differentiated and mineralized osteoblasts. Administration of FGF receptor 3 (FGFR3) inhibitor (50 nM) was sufficient to restore the FGF23-decreased ALP activity (P < 0.05), but not for the formation of mineralized nodules. In addition, the phosphorylation of ERK increased considerably with Adv-FGF23 overexpression (P < 0.05). Administration of an ERK-specific inhibitor (10 μM) could down-regulate the phosphorylation of ERK (P-ERK) (P < 0.05) and slightly restored the Adv-FGF23-reduction of ALP activity (P = 0.08). In summary, our data suggest that GPS, 1, 25(OH)2D3, and PTH could regulate FGF23 mRNA expression in vitro. FGF23 is a negative regulator of bone remodeling. FGF23 not only inhibits BMSCs osteogenesis through the FGFR3-ERK signaling pathway but also suppresses the mineralization of mature osteoblasts.
Collapse
Affiliation(s)
- Zhengtian Lyu
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Haifang Li
- Department of Life Science, Shandong Agricultural University, Taian City, Shandong Province, 271018, China
| | - Xin Li
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Hui Wang
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Hongchao Jiao
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Xiaojuan Wang
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Jingpeng Zhao
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China.
| |
Collapse
|
8
|
Pelizzo G, Silvestro S, Avanzini MA, Zuccotti G, Mazzon E, Calcaterra V. Mesenchymal Stromal Cells for the Treatment of Interstitial Lung Disease in Children: A Look from Pediatric and Pediatric Surgeon Viewpoints. Cells 2021; 10:3270. [PMID: 34943779 PMCID: PMC8699409 DOI: 10.3390/cells10123270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been proposed as a potential therapy to treat congenital and acquired lung diseases. Due to their tissue-regenerative, anti-fibrotic, and immunomodulatory properties, MSCs combined with other therapy or alone could be considered as a new approach for repair and regeneration of the lung during disease progression and/or after post- surgical injury. Children interstitial lung disease (chILD) represent highly heterogeneous rare respiratory diseases, with a wild range of age of onset and disease expression. The chILD is characterized by inflammatory and fibrotic changes of the pulmonary parenchyma, leading to gas exchange impairment and chronic respiratory failure associated with high morbidity and mortality. The therapeutic strategy is mainly based on the use of corticosteroids, hydroxychloroquine, azithromycin, and supportive care; however, the efficacy is variable, and their long-term use is associated with severe toxicity. The role of MSCs as treatment has been proposed in clinical and pre-clinical studies. In this narrative review, we report on the currently available on MSCs treatment as therapeutical strategy in chILD. The progress into the therapy of respiratory disease in children is mandatory to ameliorate the prognosis and to prevent the progression in adult age. Cell therapy may be a future therapy from both a pediatric and pediatric surgeon's point of view.
Collapse
Affiliation(s)
- Gloria Pelizzo
- Pediatric Surgery Department, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, 20157 Milan, Italy;
| | - Serena Silvestro
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (E.M.)
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, 20157 Milan, Italy;
- Department of Pediatrics, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy;
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (E.M.)
| | - Valeria Calcaterra
- Department of Pediatrics, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy;
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
9
|
Mesenchymal Stem Cells in the Treatment of COVID-19, a Promising Future. Cells 2021; 10:cells10102588. [PMID: 34685567 PMCID: PMC8533906 DOI: 10.3390/cells10102588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have a potent self-renewal capacity and can differentiate into multiple cell types. They also affect the ambient tissue by the paracrine secretion of numerous factors in vivo, including the induction of other stem cells’ differentiation. In vitro, the culture media supernatant is named secretome and contains soluble molecules and extracellular vesicles that retain potent biological function in tissue regeneration. MSCs are considered safe for human treatment; their use does not involve ethical issues, as embryonic stem cells do not require genetic manipulation as induced pluripotent stem cells, and after intravenous injection, they are mainly found in the lugs. Therefore, these cells are currently being tested in various preclinical and clinical trials for several diseases, including COVID-19. Several affected COVID-19 patients develop induced acute respiratory distress syndrome (ARDS) associated with an uncontrolled inflammatory response. This condition causes extensive damage to the lungs and may leave serious post-COVID-19 sequelae. As the disease may cause systemic alterations, such as thromboembolism and compromised renal and cardiac function, the intravenous injection of MSCs may be a therapeutic alternative against multiple pathological manifestations. In this work, we reviewed the literature about MSCs biology, focusing on their function in pulmonary regeneration and their use in COVID-19 treatment.
Collapse
|
10
|
Li B, Xing Y, Gan Y, He J, Hua H. Labial gland-derived mesenchymal stem cells and their exosomes ameliorate murine Sjögren's syndrome by modulating the balance of Treg and Th17 cells. Stem Cell Res Ther 2021; 12:478. [PMID: 34446113 PMCID: PMC8390194 DOI: 10.1186/s13287-021-02541-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022] Open
Abstract
Background Sjögren's syndrome (SS) is a chronic, systemic autoimmune disorder characterized by sicca syndrome and/or systemic manifestations. The disease severely affects the health and life of patients, and the treatment of SS has always been a clinical challenge and essentially palliative. Mesenchymal stem cells (MSCs) have been reported to exert immunomodulatory effects and as a potential novel therapeutic strategy for SS. Labial gland-derived MSCs (LGMSCs) are a population of resident stem cells in the labial gland, first isolated by our group. Exosomes released by MSCs contain a large variety of bioactive molecules and considered to function as an extension of MSCs. Methods LGMSCs were isolated from patients who were needed surgery to remove the lip mucocele and LGMSCs derived exosomes (LGMSC-Exos) were isolated by ultracentrifugation. The non-obese diabetic (NOD) mice were treated with LGMSCs or LGMSC-Exos by tail vein injection. The saliva flow rate of mice was determined and salivary glands were dissected and stained with hematoxylin and eosin. In vitro, peripheral blood mononuclear cells (PBMCs) from SS patients were cocultured with LGMSCs or LGMSC-Exos. Percentage of T helper 17 (Th17) cells and regulatory T (Treg) cells were determined by flow cytometry. The serum levels of cytokines in NOD mice and in the supernatant of the co-culture system by ELISA. Results Treatment with LGMSCs or LGMSC-Exos reduced inflammatory infiltration in the salivary glands, and restored salivary gland secretory function in NOD mice. Importantly, LGMSCs or LGMSC-Exos were demonstrated to inhibit the differentiation of Th17 cells but promote the induction of Treg cells in NOD mice and PBMCs from SS patients in vitro, accompanied by reduced interleukin 17 (IL-17), interferon gamma, and IL-6 levels and enhanced transforming growth factor beta and IL-10 secretion by T cells. Conclusions LGMSCs are potential candidates for MSCs-based therapy and LGMSC-Exos might be utilized for establishing a new cell-free therapy against SS. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02541-0.
Collapse
Affiliation(s)
- Boya Li
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.,Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yixiao Xing
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.,Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yehua Gan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.,Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, People's Republic of China.
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China. .,Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| |
Collapse
|
11
|
Hoseinnia S, Ghane M, Norouzi J, Hosseini F. Mesenchymal stem cell and endothelial progenitor cells coinjection improves LPS-induced lung injury via Tie2 activation and downregulation of the TLR4/MyD88 pathway. J Cell Biochem 2021; 122:1791-1804. [PMID: 34397115 DOI: 10.1002/jcb.30133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is one of the most important complications of infection with a high mortality rate. Recently, cell therapy has been widely used to reduce the symptoms of sepsis. It has been previously reported that mesenchymal stem cell (MSC) and endothelial progenitor cells (EPC) therapy have beneficial effects in experimental models of sepsis. The effects of coculture of MSC and EPC have not yet been used to treat sepsis. Therefore, the aim of this study was to investigate the therapeutic potential of EPC + MSC coculture on the residual effects of sepsis in a lipopolysaccharide (LPS)-induced mice model. Coinjections of EPC + MSC significantly enhanced the survival rate of LPS-induced mice, decreased concentrations of pro-inflammatory cytokines, and increased the level of anti-inflammatory cytokine. The LPS-induced mice that were treated with EPC + MSC showed a notable reduction in pulmonary edema, hepatic enzymes, and C-reactive protein level compared with the control group. Our results showed that coinjection of EPC + MSC up and downregulates Tie2 and TLR4/MyD88 signaling pathways in LPS-induced mice, respectively. Also, in vitro study showed that viability, adhesion, and migration in coculture cells is significantly decreased after being induced with 10 μg/ml LPS. Our results showed that LPS impaired the functional activity of the cocultured EPC + MSC via upregulation of the TLR4/MyD88 signaling pathway, which may be associated with decreased pTie2/Tie2 expression. In conclusion, coinjection of EPC and MSC modulated the TLR4/MyD88 signaling pathway that leads to reduce the inflammatory response. This study may provide promising results for the introduction of cocultured cells to manage infectious diseases and balance the immune response through immune regulatory function.
Collapse
Affiliation(s)
- Sadaf Hoseinnia
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Ghane
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| | - Jamile Norouzi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Farzaneh Hosseini
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
12
|
Li L, Cheng D, An X, Liao G, Zhong L, Liu J, Chen Y, Yuan Y, Lu Y. Mesenchymal stem cells transplantation attenuates hyperuricemic nephropathy in rats. Int Immunopharmacol 2021; 99:108000. [PMID: 34352566 DOI: 10.1016/j.intimp.2021.108000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/05/2021] [Accepted: 07/18/2021] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs), due to their multi-directional differentiation, paracrine and immunomodulation potentials, and the capacity of homing to target organ, have been reported to facilitate regeneration and repair of kidney and improve kidney function in acute or chronic kidney injury. The present study was aimed to evaluate whether MSCs could have a protective effect in hyperuricemic nephropathy (HN) and the underlying mechanisms. A rat HN model was established by oral administration of a mixture of potassium oxonate (PO, 1.5 g/kg) and adenine (Ad, 50 mg/kg) daily for 4 weeks. For MSCs treatment, MSCs (3 × 106 cells/kg per week) were injected via tail vein from the 2nd week for 3 times. The results showed that along with the elevated uric acid (UA) in HN rats, creatinine (CREA), blood urea nitrogen (BUN), microalbuminuria (MAU) and 24-hour urinary protein levels were significantly increased comparing with the normal control rats, while decreased after MSCs treatment. Moreover, the mRNA levels of inflammation and fibrosis-related gene were reduced in UA + MSCs group. Consistently, hematoxylin-eosin (HE) staining results showed the destruction of kidney structure and fibrosis were significantly alleviated after MSCs administration. Similarly, in vitro, NRK-52Es cells were treated with high concentration UA (10 mg/dL) in the presence of MSCs, and we found that MSCs co-culture could inhibited UA-induced cell injury, characterized as improvement of cell viability and proliferation, inhibition of apoptosis, inflammation, and fibrosis. Collectively, MSCs treatment could effectively attenuate UA-induced renal injury, and thus it might be a potential therapy to hyperuricemia-related renal diseases.
Collapse
Affiliation(s)
- Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongqi Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xingxing An
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China; Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Guangneng Liao
- Experimental Animal Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Zhong
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China; Department of Clinical and Experimental Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Yan YY, Zhou WM, Wang YQ, Guo QR, Zhao FX, Zhu ZY, Xing YX, Zhang HY, Aljofan M, Jarrahi AM, Makabel B, Zhang JY. The Potential Role of Extracellular Vesicles in COVID-19 Treatment: Opportunity and Challenge. Front Mol Biosci 2021; 8:699929. [PMID: 34368228 PMCID: PMC8345113 DOI: 10.3389/fmolb.2021.699929] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 infection has become an urgent public health concern worldwide, severely affecting our society and economy due to the long incubation time and high prevalence. People spare no effort on the rapid development of vaccine and treatment all over the world. Amongst the numerous ways of tackling this pandemic, some approaches using extracellular vesicles (EVs) are emerging. In this review, we summarize current prevalence and pathogenesis of COVID-19, involving the combination of SARS-CoV-2 and virus receptor ACE2, endothelial dysfunction and micro thrombosis, together with cytokine storm. We also discuss the ongoing EVs-based strategies for the treatment of COVID-19, including mesenchymal stem cell (MSC)-EVs, drug-EVs, vaccine-EVs, platelet-EVs, and others. This manuscript provides the foundation for the development of targeted drugs and vaccines for SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Yan-yan Yan
- School of Medicine, Shanxi Datong University, Datong, China
| | - Wen-min Zhou
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yu-qing Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiao-ru Guo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Xinjiang Institute of Materia Medica, Urumqi, China
| | - Fu-xi Zhao
- School of Medicine, Shanxi Datong University, Datong, China
| | - Zhuang-yan Zhu
- School of Medicine, Shanxi Datong University, Datong, China
| | - Yan-xia Xing
- School of Medicine, Shanxi Datong University, Datong, China
| | - Hai-yan Zhang
- School of Medicine, Shanxi Datong University, Datong, China
| | - Mohamad Aljofan
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | | | | - Jian-ye Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Pranskunas M, Šimoliūnas E, Alksne M, Martin V, Gomes PS, Puisys A, Kaupinis A, Juodzbalys G. Assessment of the Bone Healing Process Mediated by Periosteum-Derived Mesenchymal Stem Cells' Secretome and a Xenogenic Bioceramic-An In Vivo Study in the Rabbit Critical Size Calvarial Defect Model. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3512. [PMID: 34202509 PMCID: PMC8269548 DOI: 10.3390/ma14133512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022]
Abstract
The mesenchymal stem cell (MSC) secretome has been considered an innovative therapeutic biological approach, able to modulate cellular crosstalk and functionality for enhanced tissue repair and regeneration. This study aims to evaluate the functionality of the secretome isolated from periosteum-derived MSCs, from either basal or osteogenic-induced conditions, in the healing of a critical size calvarial bone defect in the rabbit model. A bioceramic xenograft was used as the vehicle for secretome delivery, and the biological response to the established biocomposite system was assessed by clinical, histological, histomorphometric, and microtomographic analysis. A comparative analysis revealed that the osteogenic-induced secretome presented an increased diversity of proteins, with emphasis on those related to osteogenesis. Microtomographic and histological morphometric analysis revealed that bioceramic xenografts implanted with secretomes enhanced the new bone formation process, with the osteogenic-induced secretome inducing the highest bone tissue formation. The application of the MSC secretome, particularly from osteogenic-induced populations, may be regarded as an effective therapeutic approach to enhance bone tissue healing and regeneration.
Collapse
Affiliation(s)
- Mindaugas Pranskunas
- Department of Oral and Maxillofacial Surgery, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
- 32:Balti Dental Clinic, LT-09235 Vilnius, Lithuania
| | - Egidijus Šimoliūnas
- Life Sciences Center, Department of Biological Models, Institute of Biochemistry, Vilnius University, LT-10257 Vilnius, Lithuania; (E.Š.); (M.A.)
| | - Milda Alksne
- Life Sciences Center, Department of Biological Models, Institute of Biochemistry, Vilnius University, LT-10257 Vilnius, Lithuania; (E.Š.); (M.A.)
| | - Victor Martin
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U. Porto, 4200-393 Porto, Portugal; (V.M.); (P.S.G.)
- LAQV/REQUIMTE—U. Porto, 4200-393 Porto, Portugal
| | - Pedro Sousa Gomes
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U. Porto, 4200-393 Porto, Portugal; (V.M.); (P.S.G.)
- LAQV/REQUIMTE—U. Porto, 4200-393 Porto, Portugal
| | - Algirdas Puisys
- Vilnius Implantology Center, LT-03162 Vilnius, Lithuania;
- Vilnius Research Group, LT-02233 Vilnius, Lithuania
| | - Algirdas Kaupinis
- Proteomics Centre, Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio av. 7, LT-10257 Vilnius, Lithuania;
| | - Gintaras Juodzbalys
- Department of Oral and Maxillofacial Surgery, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| |
Collapse
|
15
|
Wszoła M, Nitarska D, Cywoniuk P, Gomółka M, Klak M. Stem Cells as a Source of Pancreatic Cells for Production of 3D Bioprinted Bionic Pancreas in the Treatment of Type 1 Diabetes. Cells 2021; 10:1544. [PMID: 34207441 PMCID: PMC8234129 DOI: 10.3390/cells10061544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is the third most common autoimmune disease which develops due to genetic and environmental risk factors. Often, intensive insulin therapy is insufficient, and patients require a pancreas or pancreatic islets transplant. However, both solutions are associated with many possible complications, including graft rejection. The best approach seems to be a donor-independent T1D treatment strategy based on human stem cells cultured in vitro and differentiated into insulin and glucagon-producing cells (β and α cells, respectively). Both types of cells can then be incorporated into the bio-ink used for 3D printing of the bionic pancreas, which can be transplanted into T1D patients to restore glucose homeostasis. The aim of this review is to summarize current knowledge about stem cells sources and their transformation into key pancreatic cells. Last, but not least, we comment on possible solutions of post-transplant immune response triggered stem cell-derived pancreatic cells and their potential control mechanisms.
Collapse
Affiliation(s)
- Michał Wszoła
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (M.W.); (P.C.); (M.G.)
- Polbionica Ltd., 01-793 Warsaw, Poland;
- Medispace Medical Centre, 01-044 Warsaw, Poland
| | | | - Piotr Cywoniuk
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (M.W.); (P.C.); (M.G.)
| | - Magdalena Gomółka
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (M.W.); (P.C.); (M.G.)
| | - Marta Klak
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (M.W.); (P.C.); (M.G.)
- Polbionica Ltd., 01-793 Warsaw, Poland;
| |
Collapse
|
16
|
Grieco GE, Fignani D, Formichi C, Nigi L, Licata G, Maccora C, Brusco N, Sebastiani G, Dotta F. Extracellular Vesicles in Immune System Regulation and Type 1 Diabetes: Cell-to-Cell Communication Mediators, Disease Biomarkers, and Promising Therapeutic Tools. Front Immunol 2021; 12:682948. [PMID: 34177928 PMCID: PMC8219977 DOI: 10.3389/fimmu.2021.682948] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are generated by cells of origin through complex molecular mechanisms and released into extracellular environment. Hence, the presence of EVs has been described in multiple biological fluids and in most cases their molecular cargo, which includes non-coding RNAs (ncRNA), messenger RNAs (mRNA), and proteins, has been reported to modulate distinct biological processes. EVs release and their molecular cargo have been demonstrated to be altered in multiple diseases, including autoimmune diseases. Notably, numerous evidence showed a relevant crosstalk between immune system and interacting cells through specific EVs release. The crosstalk between insulin-producing pancreatic β cells and immune system through EVs bidirectional trafficking has yet started to be deciphered, thus uncovering an intricate communication network underlying type 1 diabetes (T1D) pathogenesis. EVs can also be found in blood plasma or serum. Indeed, the assessment of circulating EVs cargo has been shown as a promising advance in the detection of reliable biomarkers of disease progression. Of note, multiple studies showed several specific cargo alterations of EVs collected from plasma/serum of subjects affected by autoimmune diseases, including T1D subjects. In this review, we discuss the recent literature reporting evidence of EVs role in autoimmune diseases, specifically focusing on the bidirectional crosstalk between pancreatic β cells and immune system in T1D and highlight the relevant promising role of circulating EVs as disease biomarkers.
Collapse
Affiliation(s)
- Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Daniela Fignani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy.,UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy.,UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Carla Maccora
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy.,UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Noemi Brusco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.,Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy.,UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| |
Collapse
|
17
|
The emerging roles of exosomes in autoimmune diseases, with special emphasis on microRNAs in exosomes. Pharmacol Res 2021; 169:105680. [PMID: 34010670 DOI: 10.1016/j.phrs.2021.105680] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/29/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases include rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic vasculitis, dermatomyositis, systemic sclerosis (SSc), mixed connective tissue disease, autoimmune hemolytic anemia, autoimmune thyroiditis (AITD) and ulcerative colitis. Exosomes exist in body fluids, including blood, saliva, urine, cerebrospinal fluid and milk. They are mainly derived from the invagination of intracellular lysosomal particles, which are released into the extracellular matrix after fusion of the outer membrane of the exosomes with the cell membrane. Exosomes mediate intercellular communication and regulate the biological activity of receptor cells by carrying proteins, nucleic acids and lipids. Evidences show that exosomes are involved in the pathogenesis of various autoimmune diseases. In view of the important roles of exosomes in autoimmune diseases, this work systematically reviewed the effects of exosomes on the pathogenesis of autoimmune diseases, especially the regulatory roles of exosome derived microRNAs (miRNAs) in the pathogenesis of RA, SLE, dermatomyositis, SSc, AITD and ulcerative colitis. The review of the roles of exosomes in autoimmune diseases will help to clarify the pathogenesis of these diseases and explore new diagnostic markers and therapeutic targets.
Collapse
|
18
|
Costa de Oliveira Souza CM, de Souza CF, Mogharbel BF, Irioda AC, Cavichiolo Franco CR, Sierakowski MR, Athayde Teixeira de Carvalho K. Nanostructured Cellulose-Gellan-Xyloglucan-Lysozyme Dressing Seeded with Mesenchymal Stem Cells for Deep Second-Degree Burn Treatment. Int J Nanomedicine 2021; 16:833-850. [PMID: 33584096 PMCID: PMC7875079 DOI: 10.2147/ijn.s289868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In deep burns, wound contraction and hypertrophic scar formation can generate functional derangement and debilitation of the affected part. In order to improve the quality of healing in deep second-degree burns, we developed a new treatment in a preclinical model using nanostructured membranes seeded with mesenchymal stem cells (MSCs). METHODS Membranes were obtained by reconstitution of bacterial cellulose (reconstituted membrane [RM]) and produced by a dry-cast process, then RM was incorporated with 10% tamarind xyloglucan plus gellan gum 1:1 and 10% lysozyme (RMGT-LZ) and with 10% gellan gum and 10% lysozyme (RMG-LZ). Membrane hydrophobic/hydrophilic characteristics were investigated by static/dynamic contact-angle measurements. They were cultivated with MSCs, and cell adhesion, proliferation, and migration capacity was analyzed with MTT assays. Morphological and topographic characteristics were analyzed by scanning electron microscopy. MSC patterns in flow cytometry and differentiation into adipocytes and osteocytes were checked. In vivo assays used RMG-LZ and RMGT-LZ (with and without MSCs) in Rattus norvegicus rats submitted to burn protocol, and histological sections and collagen deposits were analyzed and immunocytochemistry assay performed. RESULTS In vitro results demonstrated carboxyl and amine groups made the membranes moderately hydrophobic and xyloglucan inclusion decreased wettability, favoring MSC adhesion, proliferation, and differentiation. In vivo, we obtained 40% and 60% reduction in acute/chronic inflammatory infiltrates, 96% decrease in injury area, increased vascular proliferation and collagen deposition, and complete epithelialization after 30 days. MSCs were detected in burned tissue, confirming they had homed and proliferated in vivo. CONCLUSION Nanostructured cellulose-gellan-xyloglucan-lysozyme dressings, especially when seeded with MSCs, improved deep second-degree burn regeneration.
Collapse
Affiliation(s)
- Carolina Maria Costa de Oliveira Souza
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | - Clayton Fernandes de Souza
- Chemistry Undergraduate Program, School of Education and Humanities, Pontifícia Universidade Católica Do Paraná, Curitiba, Paraná, Brazil
- BioPol, Chemistry Department, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Bassam Felipe Mogharbel
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | - Ana Carolina Irioda
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | | | | | - Katherine Athayde Teixeira de Carvalho
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| |
Collapse
|
19
|
Baumgartner JE, Baumgartner LS, Baumgartner ME, Moore EJ, Messina SA, Seidman MD, Shook DR. Progenitor cell therapy for acquired pediatric nervous system injury: Traumatic brain injury and acquired sensorineural hearing loss. Stem Cells Transl Med 2021; 10:164-180. [PMID: 33034162 PMCID: PMC7848325 DOI: 10.1002/sctm.20-0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
While cell therapies hold remarkable promise for replacing injured cells and repairing damaged tissues, cell replacement is not the only means by which these therapies can achieve therapeutic effect. For example, recent publications show that treatment with varieties of adult, multipotent stem cells can improve outcomes in patients with neurological conditions such as traumatic brain injury and hearing loss without directly replacing damaged or lost cells. As the immune system plays a central role in injury response and tissue repair, we here suggest that multipotent stem cell therapies achieve therapeutic effect by altering the immune response to injury, thereby limiting damage due to inflammation and possibly promoting repair. These findings argue for a broader understanding of the mechanisms by which cell therapies can benefit patients.
Collapse
Affiliation(s)
- James E. Baumgartner
- Advent Health for ChildrenOrlandoFloridaUSA
- Department of Neurological SurgeryUniversity of Central Florida College of MedicineOrlandoFloridaUSA
| | | | | | - Ernest J. Moore
- Department of Audiology and Speech Language PathologyUniversity of North TexasDentonTexasUSA
| | | | - Michael D. Seidman
- Advent Health CelebrationCelebrationFloridaUSA
- Department of OtorhinolaryngologyUniversity of Central FloridaOrlandoFloridaUSA
| | | |
Collapse
|
20
|
Porzionato A, Zaramella P, Dedja A, Guidolin D, Bonadies L, Macchi V, Pozzobon M, Jurga M, Perilongo G, De Caro R, Baraldi E, Muraca M. Intratracheal administration of mesenchymal stem cell-derived extracellular vesicles reduces lung injuries in a chronic rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2021; 320:L688-L704. [PMID: 33502939 DOI: 10.1152/ajplung.00148.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Arben Dedja
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Diego Guidolin
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Michela Pozzobon
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Marcin Jurga
- The Cell Factory BVBA (Esperite NV), Niel, Belgium
| | - Giorgio Perilongo
- Institute of Pediatric Research, Padua, Italy.,Pediatric Clinic, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy.,Institute of Pediatric Research, Padua, Italy
| | - Maurizio Muraca
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| |
Collapse
|
21
|
Wei W, Ao Q, Wang X, Cao Y, Liu Y, Zheng SG, Tian X. Mesenchymal Stem Cell-Derived Exosomes: A Promising Biological Tool in Nanomedicine. Front Pharmacol 2021; 11:590470. [PMID: 33716723 PMCID: PMC7944140 DOI: 10.3389/fphar.2020.590470] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022] Open
Abstract
As nano-scale biological vesicles, extracellular vesicles (EVs)/exosomes, in particular, exosomes derived from mesenchymal stem cells (MSC-exosomes), have been studied in the diagnosis, prevention, and treatment of many diseases. In addition, through the combination of nanotechnology and biotechnology, exosomes have emerged as innovative tools for the development of nanomedicine. This review focuses on a profound summarization of MSC-exosomes as a powerful tool in bionanomedicine. It systemically summarizes the role of MSC-exosomes as a nanocarrier, drug loading and tissue engineering, and their potential contribution in a series of diseases as well as the advantages of exosomes over stem cells and synthetic nanoparticles and potential disadvantages. The in-depth understanding of the functions and mechanisms of exosomes provides insights into the basic research and clinical transformation in the field of nanomedicine.
Collapse
Affiliation(s)
- Wumei Wei
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Qiang Ao
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Yue Cao
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Yanying Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, United States
| | - Xiaohong Tian
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| |
Collapse
|
22
|
Battafarano G, Rossi M, De Martino V, Marampon F, Borro L, Secinaro A, Del Fattore A. Strategies for Bone Regeneration: From Graft to Tissue Engineering. Int J Mol Sci 2021; 22:ijms22031128. [PMID: 33498786 PMCID: PMC7865467 DOI: 10.3390/ijms22031128] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/08/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a regenerative organ characterized by self-renewal ability. Indeed, it is a very dynamic tissue subjected to continuous remodeling in order to preserve its structure and function. However, in clinical practice, impaired bone healing can be observed in patients and medical intervention is needed to regenerate the tissue via the use of natural bone grafts or synthetic bone grafts. The main elements required for tissue engineering include cells, growth factors and a scaffold material to support them. Three different materials (metals, ceramics, and polymers) can be used to create a scaffold suitable for bone regeneration. Several cell types have been investigated in combination with biomaterials. In this review, we describe the options available for bone regeneration, focusing on tissue engineering strategies based on the use of different biomaterials combined with cells and growth factors.
Collapse
Affiliation(s)
- Giulia Battafarano
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (G.B.); (M.R.)
| | - Michela Rossi
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (G.B.); (M.R.)
| | - Viviana De Martino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, 00161 Rome, Italy;
| | - Francesco Marampon
- Department of Radiotherapy, “Sapienza” University of Rome, 00161 Rome, Italy;
| | - Luca Borro
- Advanced Cardiovascular Imaging Unit, Department of Imaging, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.B.); (A.S.)
| | - Aurelio Secinaro
- Advanced Cardiovascular Imaging Unit, Department of Imaging, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (L.B.); (A.S.)
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (G.B.); (M.R.)
- Correspondence: ; Tel.: +39-066-859-3740
| |
Collapse
|
23
|
Herrmann M, Diederichs S, Melnik S, Riegger J, Trivanović D, Li S, Jenei-Lanzl Z, Brenner RE, Huber-Lang M, Zaucke F, Schildberg FA, Grässel S. Extracellular Vesicles in Musculoskeletal Pathologies and Regeneration. Front Bioeng Biotechnol 2021; 8:624096. [PMID: 33553127 PMCID: PMC7855463 DOI: 10.3389/fbioe.2020.624096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence of musculoskeletal diseases is steadily increasing with aging of the population. In the past years, extracellular vesicles (EVs) have gained attention in musculoskeletal research. EVs have been associated with various musculoskeletal pathologies as well as suggested as treatment option. EVs play a pivotal role in communication between cells and their environment. Thereby, the EV cargo is highly dependent on their cellular origin. In this review, we summarize putative mechanisms by which EVs can contribute to musculoskeletal tissue homeostasis, regeneration and disease, in particular matrix remodeling and mineralization, pro-angiogenic effects and immunomodulatory activities. Mesenchymal stromal cells (MSCs) present the most frequently used cell source for EV generation for musculoskeletal applications, and herein we discuss how the MSC phenotype can influence the cargo and thus the regenerative potential of EVs. Induced pluripotent stem cell-derived mesenchymal progenitor cells (iMPs) may overcome current limitations of MSCs, and iMP-derived EVs are discussed as an alternative strategy. In the last part of the article, we focus on therapeutic applications of EVs and discuss both practical considerations for EV production and the current state of EV-based therapies.
Collapse
Affiliation(s)
- Marietta Herrmann
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Svitlana Melnik
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Drenka Trivanović
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Shushan Li
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Susanne Grässel
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Gao X, Cao Z. Gingiva-derived Mesenchymal Stem Cells and Their Potential Applications in Oral and Maxillofacial Diseases. Curr Stem Cell Res Ther 2020; 15:43-53. [PMID: 31702517 DOI: 10.2174/1574888x14666191107100311] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/02/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stem cells are undifferentiated cells with multilineage differentiation potential. They can be collected from bone marrow, fat, amniotic fluid, and teeth. Stem cell-based therapies have been widely used to treat multiple diseases, such as cardiac disease, and hematological disorders. The cells may also be beneficial for controlling the disease course and promoting tissue regeneration in oral and maxillofacial diseases. Oral-derived gingival mesenchymal stem cells are easy to access and the donor sites heal rapidly without a scar. Such characteristics demonstrate the beneficial role of GMSCs in oral and maxillofacial diseases. OBJECTIVE We summarize the features of GMSCs, including their self-renewal, multipotent differentiation, immunomodulation, and anti-inflammation properties. We also discuss their applications in oral and maxillofacial disease treatment and tissue regeneration. CONCLUSION GMSCs are easily harvestable adult stem cells with outstanding proliferation, differentiation, and immunomodulation characteristics. A growing body of evidence indicates that GMSCs have strong potential use in accelerating wound healing and promoting the regeneration of bone defects, periodontium, oral neoplasms, salivary glands, peri-implantitis, and nerves. Moreover, alginate, polylactic acid and polycaprolactone can be used as biodegradable scaffolds for GMSC encapsulation. Various growth factors can be applied to the corresponding scaffolds to obtain the desired GMSC differentiation and phenotypes. Three-dimensional spheroid culture systems could optimize GMSC properties and improve the performance of the cells in tissue engineering. The immunomodulatory property of GMSCs in controlling oral and maxillofacial inflammation needs further research.
Collapse
Affiliation(s)
- Xudong Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
25
|
Han Q, Niu X, Hou R, Li J, Liu Y, Li X, Li J, Li Y, Zhang K, Wu Y. Dermal mesenchymal stem cells promoted adhesion and migration of endothelial cells by integrin in psoriasis. Cell Biol Int 2020; 45:358-367. [PMID: 33079476 DOI: 10.1002/cbin.11492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 07/09/2020] [Accepted: 10/18/2020] [Indexed: 01/12/2023]
Abstract
The unusual dilatation of dermal capillaries and angiogenesis played important roles in psoriasis. Some genes and proteins of dermal mesenchymal stem cells (DMSCs) from psoriasis are abnormal and related to the function of endothelial cells (ECs). The present study was aimed to evaluate whether psoriatic DMSCs could affect adhesion and migration of ECs through neovascularization-related integrins in psoriasis. Human DMSCs, collected from psoriasis lesions and healthy skin, respectively, were cocultured with human umbilical vein endothelial cells (HUVECs). The expression levels of three integrins, that is, αvβ3, αvβ5, and α5β1 in HUVECs were tested by quantitative real-time polymerase chain reaction and Western blot analysis. The adhesion and migration of HUVECs were detected by adhesion assay and migration assay. The results showed that in psoriasis group, the expression of αVβ3 and α5β1 of HUVECs markedly increased 2.50- and 3.71-fold in messenger RNA levels, and significantly increased 1.63- and 1.92-fold in protein levels, comparing to healthy control group (all p < .05). But β5 was not significantly different between the two groups (p > .05). In addition, compared with control, psoriatic DMSCs promoted HUVECs adhesion by 1.62-fold and migration by 2.91-fold (all p < .05). In conclusion, psoriatic DMSCs impact HUVECs adhesion and migration by upregulating the expression of integrins αVβ3 and α5β1.
Collapse
Affiliation(s)
- Qixin Han
- Dermatology Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruixia Hou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yamin Liu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaofang Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Li
- English Department, School of Fundamental Sciences, China Medical University, Shenyang, Liaoning, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Wu
- Dermatology Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Bazzoni R, Takam Kamga P, Tanasi I, Krampera M. Extracellular Vesicle-Dependent Communication Between Mesenchymal Stromal Cells and Immune Effector Cells. Front Cell Dev Biol 2020; 8:596079. [PMID: 33240892 PMCID: PMC7677193 DOI: 10.3389/fcell.2020.596079] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells residing in the stromal tissues of the body and capable of promoting tissue repair and attenuating inflammatory processes through their immunomodulatory properties. Preclinical and clinical observations revealed that not only direct intercellular communication mediates MSC properties; in fact, a pivotal role is also played by the release of soluble and bioactive factors, such as cytokines, growth factor and extracellular vesicles (EVs). EVs are membrane-coated vesicles containing a large variety of bioactive molecules, including lipids, proteins, and nucleic acids, such as RNA. EVs release their contents into target cells, thus influencing cell fate through the control of intracellular processes. In addition, MSC-derived EVs can mediate modulatory effects toward different effector cells belonging to both innate and adaptive immunity. In this review, we will discuss the literature data concerning MSC-derived EVs, including the current standardized methods for their isolation and characterization, the mechanisms supporting their immunoregulatory properties, and their potential clinical application as alternative to MSC-based therapy for inflammatory reactions, such as graft-versus-host disease (GvHD).
Collapse
Affiliation(s)
- Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
27
|
Zhao Y, Qin Y, Wu S, Huang D, Hu H, Zhang X, Hao D. Mesenchymal stem cells regulate inflammatory milieu within degenerative nucleus pulposus cells via p38 MAPK pathway. Exp Ther Med 2020; 20:22. [PMID: 32934687 PMCID: PMC7471866 DOI: 10.3892/etm.2020.9150] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
It has been established that excessive apoptosis of nucleus pulposus cells (NPCs) are responsible for pathogenesis of human intervertebral disc degeneration (IDD). The present study aimed to shed light on the molecular mechanisms underlying the protective effects of mesenchymal stem cells (MSCs) on NPCs in an inflammatory environment. NPCs were treated with TNF-α to induce inflammation and then co-cultured with Wharton's Jelly-derived MSCs (WJ-MSCs)without direct interaction. The levels of inflammation markers (IL-1β, IL-6 and IL-8) in NPCs were detected by performing enzyme-linked immunosorbent assay (ELISA), and expression of metalloproteases and aggrecan, as well as the activity of p38 MAPK pathway were determined through immunoblotting. SB-203580 was used to inhibit p38 signaling, prior to evaluation of the effects of Wharton's Jelly-derived MSCs (WJ-MSCs) on inflammatory response within the co-cultured NPCs. After TNF-α treatment, the levels of inflammatory cytokines, MMP-3, and MMP-13 in NPCs were increased whereas aggrecan was decreased, which was then dramatically reversed by WJ-MSCs co-culture. Likewise, WJ-MSCs suppressed TNF-α-induced phosphorylation of p38 MAPK signaling components including p38, ASK-1, MKK-3 and MKK-6. Blocking p38 MAPK pathway enhanced the anti-inflammatory impact of WJ-MSCs, and there was no significant difference between NPCs co-cultured with WJ-MSCs or the cells cultured alone. WJ-MSCs co-culture mitigate TNF-α-induced inflammatory response and ECM degeneration in NPCs, the major pathological events are implicated in IDD development, probably by suppressing the p38 MAPK signaling cascade.
Collapse
Affiliation(s)
- Yuanting Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Yue Qin
- Department of Anesthesiology, Honghui Hospital, Xi'an University, Xi'an, Shaanxi 710054, P.R. China
| | - Shufang Wu
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dageng Huang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Huimin Hu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Xinliang Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
28
|
Bajetto A, Thellung S, Dellacasagrande I, Pagano A, Barbieri F, Florio T. Cross talk between mesenchymal and glioblastoma stem cells: Communication beyond controversies. Stem Cells Transl Med 2020; 9:1310-1330. [PMID: 32543030 PMCID: PMC7581451 DOI: 10.1002/sctm.20-0161] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from bone marrow or other adult tissues (adipose tissue, dental pulp, amniotic fluid, and umbilical cord). In vitro, MSCs grow as adherent cells, display fibroblast-like morphology, and self-renew, undergoing specific mesodermal differentiation. High heterogeneity of MSCs from different origin, and differences in preparation techniques, make difficult to uniform their functional properties for therapeutic purposes. Immunomodulatory, migratory, and differentiation ability, fueled clinical MSC application in regenerative medicine, whereas beneficial effects are currently mainly ascribed to their secretome and extracellular vesicles. MSC translational potential in cancer therapy exploits putative anti-tumor activity and inherent tropism toward tumor sites to deliver cytotoxic drugs. However, controversial results emerged evaluating either the therapeutic potential or homing efficiency of MSCs, as both antitumor and protumor effects were reported. Glioblastoma (GBM) is the most malignant brain tumor and its development and aggressive nature is sustained by cancer stem cells (CSCs) and the identification of effective therapeutic is required. MSC dualistic action, tumor-promoting or tumor-targeting, is dependent on secreted factors and extracellular vesicles driving a complex cross talk between MSCs and GBM CSCs. Tumor-tropic ability of MSCs, besides providing an alternative therapeutic approach, could represent a tool to understand the biology of GBM CSCs and related paracrine mechanisms, underpinning MSC-GBM interactions. In this review, recent findings on the complex nature of MSCs will be highlighted, focusing on their elusive impact on GBM progression and aggressiveness by direct cell-cell interaction and via secretome, also facing the perspectives and challenges in treatment strategies.
Collapse
Affiliation(s)
- Adriana Bajetto
- Dipartimento di Medicina InternaUniversità di GenovaGenovaItaly
| | | | | | - Aldo Pagano
- Dipartimento di Medicina SperimentaleUniversità di GenovaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | | | - Tullio Florio
- Dipartimento di Medicina InternaUniversità di GenovaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
29
|
Qian X, An N, Ren Y, Yang C, Zhang X, Li L. Immunosuppressive Effects of Mesenchymal Stem Cells-derived Exosomes. Stem Cell Rev Rep 2020; 17:411-427. [PMID: 32935222 DOI: 10.1007/s12015-020-10040-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) have become important seed cells in therapy because of their immunosuppressive function and anti-inflammatory effects. MSCs exert immunosuppressive effects through direct contact or paracrine action. The paracrine functions of MSCs are at least partially mediated by exosomes, which are membrane vesicles, carrying abundant proteins, nucleic acids and other active molecules. MSC-exos have heterogeneity. The exosomes from different donors, tissues generations of MSCs carry different bioactive molecules. These cargos are transferred to recipient cells by endocytosis or binding to proteins on the receptor surface to mediate intercellular communication between different cell types and affect the functions of the recipient cells. Exosomes play an important role in the regulation of the immune system. Exosomes derived from MSCs (MSC-exos) carry immunomodulatory effectors or transmit active signal molecules to regulate the biological activities of immune cells and thus mediating immune suppression, especially on macrophages and T cells. Mitochondria and autophagy-related pathways are also associated with MSC-exos immunosuppressive effects. Graphical Abstract.
Collapse
Affiliation(s)
- Xiaoli Qian
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Nan An
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Yifan Ren
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Chenxin Yang
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
30
|
Taghavi-Farahabadi M, Mahmoudi M, Mahdaviani SA, Baghaei K, Rayzan E, Hashemi SM, Rezaei N. Improving the function of neutrophils from chronic granulomatous disease patients using mesenchymal stem cells' exosomes. Hum Immunol 2020; 81:614-624. [PMID: 32891471 DOI: 10.1016/j.humimm.2020.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/10/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
In chronic granulomatous disease (CGD) patients, reactive oxygen species (ROS) production by neutrophils is impaired. So, they are susceptible to infections. Studies showed that, mesenchymal stem cells (MSCs) have protective effects on the function of neutrophils and an approach that MSCs use to apply their effects, is secreting soluble factors and exosomes. So, we investigated the effects of MSC-exosomes and MSC-conditioned media (MSC-CM) on the function and apoptosis of neutrophils in CGD patients. In this study, neutrophils were isolated from healthy donors and CGD patients and then incubated with exosomes or CM that were prepared from MSCs. Then, neutrophil respiratory burst, apoptosis and phagocytosis capacity were evaluated by NBT assay, Annexin V-PI method and Giemsa staining. It was demonstrated that both MSC-exosomes and CM could improve the phagocytosis capacity and ROS production of neutrophils in CGD patients and healthy donors. In contrast to the healthy group, in CGD patients, exosomes significantly reduced the percentage of viable neutrophils. This report indicated that MSC exosomes and CM could increase the function of the neutrophils isolated from CGD patients. But decreasing the number of the living cells is one of the limitations of them. However, it is hoped that this intervention will be developed in future studies to minimize its limitations.
Collapse
Affiliation(s)
- Mahsa Taghavi-Farahabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Mahdaviani
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorder Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts, Universal Scientific Education and Research Network, Tehran University of Medical Sciences, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK.
| |
Collapse
|
31
|
Bujko K, Cymer M, Adamiak M, Ratajczak MZ. An Overview of Novel Unconventional Mechanisms of Hematopoietic Development and Regulators of Hematopoiesis - a Roadmap for Future Investigations. Stem Cell Rev Rep 2020; 15:785-794. [PMID: 31642043 PMCID: PMC6925068 DOI: 10.1007/s12015-019-09920-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem cells (HSCs) are the best-characterized stem cells in adult tissues. Nevertheless, as of today, many open questions remain. First, what is the phenotype of the most primitive "pre-HSC" able to undergo asymmetric divisions during ex vivo expansion that gives rise to HSC for all hemato-lymphopoietic lineages. Next, most routine in vitro assays designed to study HSC specification into hematopoietic progenitor cells (HPCs) for major hematopoietic lineages are based on a limited number of peptide-based growth factors and cytokines, neglecting the involvement of several other regulators that are endowed with hematopoietic activity. Examples include many hormones, such as pituitary gonadotropins, gonadal sex hormones, IGF-1, and thyroid hormones, as well as bioactive phosphosphingolipids and extracellular nucleotides (EXNs). Moreover, in addition to regulation by stromal-derived factor 1 (SDF-1), trafficking of these cells during mobilization or homing after transplantation is also regulated by bioactive phosphosphingolipids, EXNs, and three ancient proteolytic cascades, the complement cascade (ComC), the coagulation cascade (CoA), and the fibrinolytic cascade (FibC). Finally, it has emerged that bone marrow responds by "sterile inflammation" to signals sent from damaged organs and tissues, systemic stress, strenuous exercise, gut microbiota, and the administration of certain drugs. This review will address the involvement of these unconventional regulators and present a broader picture of hematopoiesis.
Collapse
Affiliation(s)
- Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Monika Cymer
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Adamiak
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA. .,Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
32
|
Yang JH, Liu FX, Wang JH, Cheng M, Wang SF, Xu DH. Mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles: Potential roles in rheumatic diseases. World J Stem Cells 2020; 12:688-705. [PMID: 32843922 PMCID: PMC7415241 DOI: 10.4252/wjsc.v12.i7.688] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been widely investigated in rheumatic disease due to their immunomodulatory and regenerative properties. Recently, mounting studies have implicated the therapeutic potency of MSCs mostly due to the bioactive factors they produce. Extracellular vesicles (EVs) derived from MSCs have been identified as a promising cell-free therapy due to low immunogenicity. Rheumatic disease, primarily including rheumatoid arthritis and osteoarthritis, is a group of diseases in which immune dysregulation and chronic progressive inflammation lead to irreversible joint damage. Targeting MSCs and MSC-derived EVs may be a more effective and promising therapeutic strategy for rheumatic diseases.
AIM To evaluate the potential therapeutic effectiveness of MSCs and EVs generated from MSCs in rheumatic diseases.
METHODS PubMed was searched for the relevant literature using corresponding search terms alone or in combination. Papers published in English language from January 1999 to February 2020 were considered. Preliminary screening of papers concerning analysis of "immunomodulatory function" or "regenerative function" by scrutinizing the titles and abstracts of the literature, excluded the papers not related to the subject of the article. Some other related studies were obtained by manually retrieving the reference lists of papers that comply with the selection criteria, and these studies were screened to meet the final selection and exclusion criteria.
RESULTS Eighty-six papers were ultimately selected for analysis. After analysis of the literature, it was found that both MSCs and EVs generated from MSCs have great potential in multiple rheumatic diseases, such as rheumatoid arthritis and osteoarthritis, in repair and regeneration of tissues, inhibition of inflammatory response, and regulation of body immunity via promoting chondrogenesis, regulating innate and adaptive immune cells, and regulating the secretion of inflammatory factors. But EVs from MSCs exhibit much more advantages over MSCs, which may represent another promising cell-free restorative strategy. Targeting MSCs and MSC-derived EVs may be a more efficient treatment for patients with rheumatic diseases.
CONCLUSION The enormous potential of MSCs and EVs from MSCs in immunomodulation and tissue regeneration offers a new idea for the treatment of rheumatism. However, more in-depth exploration is needed before their clinical application.
Collapse
Affiliation(s)
- Jing-Han Yang
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Feng-Xia Liu
- Department of Allergy, Weifang People’s Hospital, Weifang 261000, Shandong Province, China
| | - Jing-Hua Wang
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Min Cheng
- Department of Physiology, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Shu-Feng Wang
- Medical Experimental Training Center, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Dong-Hua Xu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| |
Collapse
|
33
|
Hu W, Song X, Yu H, Sun J, Zhao Y. Therapeutic Potentials of Extracellular Vesicles for the Treatment of Diabetes and Diabetic Complications. Int J Mol Sci 2020; 21:ijms21145163. [PMID: 32708290 PMCID: PMC7404127 DOI: 10.3390/ijms21145163] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are nano-to-micrometer vesicles released from nearly all cellular types. EVs comprise a mixture of bioactive molecules (e.g., mRNAs, miRNAs, lipids, and proteins) that can be transported to the targeted cells/tissues via the blood or lymph circulation. Recently, EVs have received increased attention, owing to their emerging roles in cell-to-cell communication, or as biomarkers with the therapeutic potential to replace cell-based therapy. Diabetes comprises a group of metabolic disorders characterized by hyperglycemia that cause the development of life-threatening complications. The impacts of conventional clinical treatment are generally limited and are followed by many side effects, including hypoglycemia, obesity, and damage to the liver and kidney. Recently, several studies have shown that EVs released by stem cells and immune cells can regulate gene expression in the recipient cells, thus providing a strategy to treat diabetes and its complications. In this review, we summarize the results from currently available studies, demonstrating the therapeutic potentials of EVs in diabetes and diabetic complications. Additionally, we highlight recommendations for future research.
Collapse
Affiliation(s)
- Wei Hu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (W.H.); (X.S.); (H.Y.)
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA;
| | - Xiang Song
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (W.H.); (X.S.); (H.Y.)
| | - Haibo Yu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (W.H.); (X.S.); (H.Y.)
| | - Jingyu Sun
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA;
| | - Yong Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (W.H.); (X.S.); (H.Y.)
- Correspondence: ; Tel.: +1-201-880-3460
| |
Collapse
|
34
|
Muraca M, Pessina A, Pozzobon M, Dominici M, Galderisi U, Lazzari L, Parolini O, Lucarelli E, Perilongo G, Baraldi E. Mesenchymal stromal cells and their secreted extracellular vesicles as therapeutic tools for COVID-19 pneumonia? J Control Release 2020; 325:135-140. [PMID: 32622963 PMCID: PMC7332437 DOI: 10.1016/j.jconrel.2020.06.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 epidemic represents an unprecedented global health emergency, further aggravated by the lack of effective therapies. For this reason, several clinical trials are testing different off-label drugs, already approved for other pathologies. Mesenchymal stem/stromal cells (MSCs) have been tested during the last two decades for the treatment of various pathologic conditions, including acute and chronic lung diseases, both in animal models and in patients. In particular, promising results have been obtained in the experimental therapy of acute respiratory distress syndrome, which represents the most threatening complication of COVID-19 infection. Furthermore, more recently, great interest has been devoted to the possible clinical applications of extracellular vesicles secreted by MSCs, nanoparticles that convey much of the biological effects and of the therapeutic efficacy of their cells of origin. This review summarizes the experimental evidence underlying the possible use of MSCs and of MSC-EVs in severe COVID-19 infection and underlines the need to evaluate the possible efficacy of these therapeutic approaches through controlled studies under the supervision of the Regulatory Authorities.
Collapse
Affiliation(s)
- Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Italy
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of MilanVia Pascal 3620133 Milano - Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli University, Naples, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Trasfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, and Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Enrico Lucarelli
- Osteoarticolar Regeneration Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giorgio Perilongo
- Department of Women's and Children's Health, University of Padova, Italy
| | - Eugenio Baraldi
- Department of Women's and Children's Health, University of Padova, Italy
| |
Collapse
|
35
|
Bonadies L, Zaramella P, Porzionato A, Perilongo G, Muraca M, Baraldi E. Present and Future of Bronchopulmonary Dysplasia. J Clin Med 2020; 9:jcm9051539. [PMID: 32443685 PMCID: PMC7290764 DOI: 10.3390/jcm9051539] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common respiratory disorder among infants born extremely preterm. The pathogenesis of BPD involves multiple prenatal and postnatal mechanisms affecting the development of a very immature lung. Their combined effects alter the lung's morphogenesis, disrupt capillary gas exchange in the alveoli, and lead to the pathological and clinical features of BPD. The disorder is ultimately the result of an aberrant repair response to antenatal and postnatal injuries to the developing lungs. Neonatology has made huge advances in dealing with conditions related to prematurity, but efforts to prevent and treat BPD have so far been only partially effective. Seeing that BPD appears to have a role in the early origin of chronic obstructive pulmonary disease, its prevention is pivotal also in long-term respiratory outcome of these patients. There is currently some evidence to support the use of antenatal glucocorticoids, surfactant therapy, protective noninvasive ventilation, targeted saturations, early caffeine treatment, vitamin A, and fluid restriction, but none of the existing strategies have had any significant impact in reducing the burden of BPD. New areas of research are raising novel therapeutic prospects, however. For instance, early topical (intratracheal or nebulized) steroids seem promising: they might help to limit BPD development without the side effects of systemic steroids. Evidence in favor of stem cell therapy has emerged from several preclinical trials, and from a couple of studies in humans. Mesenchymal stromal/stem cells (MSCs) have revealed a reparatory capability, preventing the progression of BPD in animal models. Administering MSC-conditioned media containing extracellular vesicles (EVs) have also demonstrated a preventive action, without the potential risks associated with unwanted engraftment or the adverse effects of administering cells. In this paper, we explore these emerging treatments and take a look at the revolutionary changes in BPD and neonatology on the horizon.
Collapse
Affiliation(s)
- Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
| | - Andrea Porzionato
- Human Anatomy Section, Department of Neurosciences, University of Padova, 35128 Padova, Italy;
| | - Giorgio Perilongo
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
| | - Maurizio Muraca
- Institute of Pediatric Research “Città della Speranza”, Stem Cell and Regenerative Medicine Laboratory, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
- Correspondence: ; Tel.: +39-049-821-3560; Fax: +39-049-821-3502
| |
Collapse
|
36
|
Angioni R, Liboni C, Herkenne S, Sánchez-Rodríguez R, Borile G, Marcuzzi E, Calì B, Muraca M, Viola A. CD73 + extracellular vesicles inhibit angiogenesis through adenosine A 2B receptor signalling. J Extracell Vesicles 2020; 9:1757900. [PMID: 32489531 PMCID: PMC7241475 DOI: 10.1080/20013078.2020.1757900] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/04/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
Pathological angiogenesis is a hallmark of several conditions including eye diseases, inflammatory diseases, and cancer. Stromal cells play a crucial role in regulating angiogenesis through the release of soluble factors or direct contact with endothelial cells. Here, we analysed the properties of the extracellular vesicles (EVs) released by bone marrow mesenchymal stromal cells (MSCs) and explored the possibility of using them to therapeutically target angiogenesis. We demonstrated that in response to pro-inflammatory cytokines, MSCs produce EVs that are enriched in TIMP-1, CD39 and CD73 and inhibit angiogenesis targeting both extracellular matrix remodelling and endothelial cell migration. We identified a novel anti-angiogenic mechanism based on adenosine production, triggering of A2B adenosine receptors, and induction of NOX2-dependent oxidative stress within endothelial cells. Finally, in pilot experiments, we exploited the anti-angiogenic EVs to inhibit tumour progression in vivo. Our results identify novel pathways involved in the crosstalk between endothelial and stromal cell and suggest new therapeutic strategies to target pathological angiogenesis.
Collapse
Affiliation(s)
- Roberta Angioni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
- Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Cristina Liboni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | | | - Ricardo Sánchez-Rodríguez
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Giulia Borile
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Elisabetta Marcuzzi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Bianca Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Maurizio Muraca
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
- Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| |
Collapse
|
37
|
Shi Y, Shi H, Nomi A, Lei-Lei Z, Zhang B, Qian H. Mesenchymal stem cell-derived extracellular vesicles: a new impetus of promoting angiogenesis in tissue regeneration. Cytotherapy 2020; 21:497-508. [PMID: 31079806 DOI: 10.1016/j.jcyt.2018.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/25/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Over the past few decades, extracellular vesicles (EVs) have emerged as crucial mediators of intercellular communication. EVs encapsulate and convey information to surrounding cells or distant cells, where they mediate cellular biological responses. Among their multifaceted roles in the modulation of biological responses, the involvement of EVs in vascular development, growth and maturation has been widely documented and their potential therapeutic application in regenerative medicine or in the treatment of angiogenesis-related diseases is drawing increasing interest. In this review, we have summarized the details about the current knowledge on biogenesis of EVs and conventional isolation methods. Evidence supporting the use of EVs derived from mesenchymal stromal cells (MSCs) to enhance angiogenesis in the development of insufficient angiogenesis, such as chronic wounds, stroke and myocardial infarction, will also be discussed critically. Finally, the main challenges and prerequisites for their therapeutic applications will be evaluated.
Collapse
Affiliation(s)
- Yinghong Shi
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, the People's Republic of China; Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, the People's Republic of China
| | - Hui Shi
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, the People's Republic of China; Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, the People's Republic of China
| | - Adnan Nomi
- Department of International Exchange and Cooperation, Jining Medical University, Jining, Shandong, the People's Republic of China
| | - Zhang Lei-Lei
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, the People's Republic of China; Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, the People's Republic of China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, the People's Republic of China.
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, the People's Republic of China; Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, the People's Republic of China.
| |
Collapse
|
38
|
Mesenchymal Stem Cells from Human Exfoliated Deciduous Teeth and the Orbicularis Oris Muscle: How Do They Behave When Exposed to a Proinflammatory Stimulus? Stem Cells Int 2020; 2020:3670412. [PMID: 32184831 PMCID: PMC7060870 DOI: 10.1155/2020/3670412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/04/2020] [Accepted: 02/01/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been studied as a promising type of stem cell for use in cell therapies because of their ability to regulate the immune response. Although they are classically isolated from the bone marrow, many studies have sought to isolate MSCs from noninvasive sources. The objective of this study was to evaluate how MSCs isolated from the dental pulp of human exfoliated deciduous teeth (SHED) and fragments of the orbicularis oris muscle (OOMDSCs) behave when treated with an inflammatory IFN-γ stimulus, specifically regarding their proliferative, osteogenic, and immunomodulatory potentials. The results demonstrated that the proliferation of SHED and OOMDSCs was inhibited by the addition of IFN-γ to their culture medium and that treatment with IFN-γ at higher concentrations resulted in a greater inhibition of the proliferation of these cells than treatment with IFN-γ at lower concentrations. SHED and OOMDSCs maintained their osteogenic differentiation potential after stimulation with IFN-γ. Additionally, SHED and OOMDSCs have been shown to have low immunogenicity because they lack expression of HLA-DR and costimulatory molecules such as CD40, CD80, and CD86 before and after IFN-γ treatment. Last, SHED and OOMDSCs expressed the immunoregulatory molecule HLA-G, and the expression of this antigen increased after IFN-γ treatment. In particular, an increase in intracellular HLA-G expression was observed. The results obtained suggest that SHED and OOMDSCs lack immunogenicity and have immunomodulatory properties that are enhanced when they undergo inflammatory stimulation with IFN-γ, which opens new perspectives for the therapeutic use of these cells.
Collapse
|
39
|
Alcaraz MJ, Compañ A, Guillén MI. Extracellular Vesicles from Mesenchymal Stem Cells as Novel Treatments for Musculoskeletal Diseases. Cells 2019; 9:cells9010098. [PMID: 31906087 PMCID: PMC7017209 DOI: 10.3390/cells9010098] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising therapy for musculoskeletal diseases. There is compelling evidence indicating that MSC effects are mainly mediated by paracrine mechanisms and in particular by the secretion of extracellular vesicles (EVs). Many studies have thus suggested that EVs may be an alternative to cell therapy with MSCs in tissue repair. In this review, we summarize the current understanding of MSC EVs actions in preclinical studies of (1) immune regulation and rheumatoid arthritis, (2) bone repair and bone diseases, (3) cartilage repair and osteoarthritis, (4) intervertebral disk degeneration and (5) skeletal muscle and tendon repair. We also discuss the mechanisms underlying these actions and the perspectives of MSC EVs-based strategies for future treatments of musculoskeletal disorders.
Collapse
Affiliation(s)
- María José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
- Correspondence:
| | - Alvaro Compañ
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
| | - María Isabel Guillén
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
- Department of Pharmacy, Cardenal Herrera-CEU University, Ed. Ciencias de la Salud, 46115 Alfara, Valencia, Spain
| |
Collapse
|
40
|
Medhat D, Rodríguez CI, Infante A. Immunomodulatory Effects of MSCs in Bone Healing. Int J Mol Sci 2019; 20:ijms20215467. [PMID: 31684035 PMCID: PMC6862454 DOI: 10.3390/ijms20215467] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into multilineage cells, thus making them a significant prospect as a cell source for regenerative therapy; however, the differentiation capacity of MSCs into osteoblasts seems to not be the main mechanism responsible for the benefits associated with human mesenchymal stem cells hMSCs when used in cell therapy approaches. The process of bone fracture restoration starts with an instant inflammatory reaction, as the innate immune system responds with cytokines that enhance and activate many cell types, including MSCs, at the site of the injury. In this review, we address the influence of MSCs on the immune system in fracture repair and osteogenesis. This paradigm offers a means of distinguishing target bone diseases to be treated with MSC therapy to enhance bone repair by targeting the crosstalk between MSCs and the immune system.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
41
|
da Costa Gonçalves F, Paz AH. Cell membrane and bioactive factors derived from mesenchymal stromal cells: Cell-free based therapy for inflammatory bowel diseases. World J Stem Cells 2019; 11:618-633. [PMID: 31616539 PMCID: PMC6789183 DOI: 10.4252/wjsc.v11.i9.618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/23/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract associated with multifactorial conditions such as ulcerative colitis and Crohn’s disease. Although the underlying mechanisms of IBD remain unclear, growing evidence has shown that dysregulated immune system reactions in genetically susceptible individuals contribute to mucosal inflammation. However, conventional treatments have been effective in inducing remission of IBD but not in preventing the relapse of them. In this way, mesenchymal stromal cells (MSC) therapy has been recognized as a promising treatment for IBD due to their immunomodulatory properties, ability to differentiate into several tissues, and homing to inflammatory sites. Even so, literature is conflicted regarding the location and persistence of MSC in the body after transplantation. For this reason, recent studies have focused on the paracrine effect of the biofactors secreted by MSC, especially in relation to the immunomodulatory potential of soluble factors (cytokines, chemokines, and growth factors) and extracellular vehicles that are involved in cell communication and in the transfer of cellular material, such as proteins, lipids, and nucleic acids. Moreover, treatment with interferon-γ, tumor necrosis factor-α, and interleukin-1β causes MSC to express immunomodulatory molecules that mediate the suppression via cell-contact dependent mechanisms. Taken together, we present an overview of the role of bioactive factors and cell membrane proteins derived from MSC as a cell-free therapy that can improve IBD treatment.
Collapse
Affiliation(s)
- Fabiany da Costa Gonçalves
- Nephrology and Transplantation, Internal Medicine, Erasmus Medical Center, Rotterdam, GD 3015, Netherlands
| | - Ana Helena Paz
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-903, Brazil
| |
Collapse
|
42
|
Simeoli R, Fierabracci A. Insights into the Role of MicroRNAs in the Onset and Development of Diabetic Neuropathy. Int J Mol Sci 2019; 20:ijms20184627. [PMID: 31540445 PMCID: PMC6770207 DOI: 10.3390/ijms20184627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/30/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetic neuropathy is a serious complication of chronic hyperglycemia in diabetes patients. This complication can involve both peripheral sensorimotor and autonomic nervous system. The precise nature of injury to the peripheral nerves mediated by chronic hyperglycemia is unknown; however, several mechanisms have been proposed including polyol pathway activation, enhanced glycation of proteins and lipids, increased oxidative stress, and cytokine release in the site of injury. MicroRNAs (miRNAs) are small non-coding RNAs that mediate RNA interference by post-transcriptionally modulating gene expression and protein synthesis. Therefore, they have been implicated in several developmental, physiological, and pathophysiological processes where they modulate the expression of different proteins. Recently, miRNAs gained an increasing attention also for their role as diagnostic test in many diseases due to their stability in serum and their easy detection. Furthermore, recent studies suggest that miRNAs may be involved in diabetic neuropathy although their role in the onset and the development of this complication is not fully understood. In this review, we discuss the most recent literature providing evidence for miRNAs role in diabetic neuropathy opening new pathways to improve both early diagnosis and treatment of this complication.
Collapse
Affiliation(s)
- Raffaele Simeoli
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
43
|
Eleuteri S, Fierabracci A. Insights into the Secretome of Mesenchymal Stem Cells and Its Potential Applications. Int J Mol Sci 2019; 20:ijms20184597. [PMID: 31533317 PMCID: PMC6770239 DOI: 10.3390/ijms20184597] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have regenerative, immunoregulatory properties and can be easily isolated and expanded in vitro. Despite being a powerful tool for clinical applications, they present limitations in terms of delivery, safety, and variability of therapeutic response. Interestingly, the MSC secretome composed by cytokines, chemokines, growth factors, proteins, and extracellular vesicles, could represent a valid alternative to their use. It is noteworthy that MSC-derived extracellular vesicles (MSC-EVs) have the same effect and could be advantageous compared to the parental cells because of their specific miRNAs load. MiRNAs could be useful both in diagnostic procedures such as “liquid biopsy” to identify early pathologies and in the therapeutic field. Not only are MSC-EVs’ preservation, transfer, and production easier, but their administration is also safer, hence some clinical trials are ongoing. However, much effort is required to improve the characterization of EVs to avoid artifacts and guarantee reproducibility of the studies.
Collapse
Affiliation(s)
- Sharon Eleuteri
- Infectivology and Clinical Trials Area, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
44
|
Extracellular Microvesicles as New Industrial Therapeutic Frontiers. Trends Biotechnol 2019; 37:707-729. [DOI: 10.1016/j.tibtech.2018.11.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
|
45
|
Sluijter JPG, Davidson SM, Boulanger CM, Buzás EI, de Kleijn DPV, Engel FB, Giricz Z, Hausenloy DJ, Kishore R, Lecour S, Leor J, Madonna R, Perrino C, Prunier F, Sahoo S, Schiffelers RM, Schulz R, Van Laake LW, Ytrehus K, Ferdinandy P. Extracellular vesicles in diagnostics and therapy of the ischaemic heart: Position Paper from the Working Group on Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc Res 2019; 114:19-34. [PMID: 29106545 PMCID: PMC5852624 DOI: 10.1093/cvr/cvx211] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs)—particularly exosomes and microvesicles (MVs)—are attracting considerable interest in the cardiovascular field as the wide range of their functions is recognized. These capabilities include transporting regulatory molecules including different RNA species, lipids, and proteins through the extracellular space including blood and delivering these cargos to recipient cells to modify cellular activity. EVs powerfully stimulate angiogenesis, and can protect the heart against myocardial infarction. They also appear to mediate some of the paracrine effects of cells, and have therefore been proposed as a potential alternative to cell-based regenerative therapies. Moreover, EVs of different sources may be useful biomarkers of cardiovascular disease identities. However, the methods used for the detection and isolation of EVs have several limitations and vary widely between studies, leading to uncertainties regarding the exact population of EVs studied and how to interpret the data. The number of publications in the exosome and MV field has been increasing exponentially in recent years and, therefore, in this ESC Working Group Position Paper, the overall objective is to provide a set of recommendations for the analysis and translational application of EVs focussing on the diagnosis and therapy of the ischaemic heart. This should help to ensure that the data from emerging studies are robust and repeatable, and optimize the pathway towards the diagnostic and therapeutic use of EVs in clinical studies for patient benefit.
Collapse
Affiliation(s)
- Joost Petrus Gerardus Sluijter
- Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, University Utrecht, 3508GA Utrecht, The Netherlands
| | | | | | - Edit Iren Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.,MTA-SE Immunoproteogenomics Research Group, Budapest, Hungary
| | - Dominique Paschalis Victor de Kleijn
- Department of Vascular Surgery, UMC Utrecht, Utrecht University, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| | - Felix Benedikt Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore 119228.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London W1T 7DN, UK.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, W Smithfield, London EC1A 7BE, UK
| | - Raj Kishore
- Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel Hashomer, Israel; Tamman Cardiovascular Research Institute, Heart Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Rosalinda Madonna
- Center of Aging Science and Regenerative Medicine, CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, TX, USA.,Texas Heart Institute, Houston, TX, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Fabrice Prunier
- Institut Mitovasc, CHU d'Angers, Université d'Angers, Angers, France
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ray Michel Schiffelers
- Laboratory Clinical Chemistry and Hematology Division, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Linda Wilhelmina Van Laake
- Division Heart and Lungs, and Hubrecht Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest 1089, Hungary and.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
46
|
Stergar J, Gradisnik L, Velnar T, Maver U. Intervertebral disc tissue engineering: A brief review. Bosn J Basic Med Sci 2019; 19:130-137. [PMID: 30726701 DOI: 10.17305/bjbms.2019.3778] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 07/29/2018] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD) is associated with low back pain and significantly affects the patient's quality of life. Degeneration of the IVD alters disk height and the mechanics of the spine, leading to chronic segmental spinal instability. The pathophysiology of IVD disease is still not well understood. Current therapies for IDD include conservative and invasive approaches, but none of those treatments are able to restore the disc structure and function. Recently, tissue engineering techniques emerged as a possible approach to treat IDD, by replacing a damaged IVD with scaffolds and appropriate cells. Advances in manufacturing techniques, material processing and development, surface functionalization, drug delivery systems and cell incorporation furthered the development of tissue engineering therapies. In this review, biomaterial scaffolds and cell-based therapies for IVD regeneration are briefly discussed.
Collapse
Affiliation(s)
- Janja Stergar
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia Laboratory of Inorganic Chemistry, Faculty of Chemistry and Chemical Technology, University of Maribor, Maribor, Slovenia.
| | | | | | | |
Collapse
|
47
|
Kordelas L, Schwich E, Dittrich R, Horn PA, Beelen DW, Börger V, Giebel B, Rebmann V. Individual Immune-Modulatory Capabilities of MSC-Derived Extracellular Vesicle (EV) Preparations and Recipient-Dependent Responsiveness. Int J Mol Sci 2019; 20:ijms20071642. [PMID: 30987036 PMCID: PMC6479947 DOI: 10.3390/ijms20071642] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/22/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
Treatment with extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have been suggested as novel therapeutic option in acute inflammation-associated disorders due to their immune-modulatory capacities. As we have previously observed differences in the cytokine profile of independent MSC-EV preparations, functional differences of MSC-EV preparations have to be considered. To evaluate the immune-modulatory capabilities of specific MSC-EV preparations, reliable assays are required to characterize the functionality of MSC-EV preparations prior to administration to a patient. To this end, we established an in vitro assay evaluating the immune-modulatory capacities of MSC-EV preparations. Here, we compared the efficacy of four independent MSC-EV preparations to modulate the induction of T cell differentiation and cytokine production after phorbol 12-myristate 13-acetate (PMA)/Ionomycin stimulation of peripheral blood mononuclear cells (PBMC) derived from six healthy donors. Flow cytometric analyses revealed that the four MSC-EV preparations differentially modulate the expression of surface markers, such as CD45RA, on CD4+ and CD8+ T cells, resulting in shifts in the frequencies of effector and effector memory T cells. Moreover, cytokine profile in T cell subsets was affected in a MSC-EV-specific manner exclusively in CD8+ naïve T cells. Strikingly, hierarchical clustering revealed that the T cell response towards the MSC-EV preparations largely varied among the different PBMC donors. Thus, besides defining functional activity of MSC-EV preparations, it will be crucial to test whether patients intended for treatment with MSC-EV preparations are in principal competent to respond to the envisioned MSC-EV therapy.
Collapse
Affiliation(s)
- Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, 45147 Essen, Germany.
| | - Esther Schwich
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| | - Robin Dittrich
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| | - Dietrich W Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, 45147 Essen, Germany.
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| | - Vera Rebmann
- Institute for Transfusion Medicine, University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|
48
|
Interactions between cancer stem cells, immune system and some environmental components: Friends or foes? Immunol Lett 2019; 208:19-29. [DOI: 10.1016/j.imlet.2019.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/02/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
|
49
|
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 2019; 27:55-69. [PMID: 29562775 PMCID: PMC6434476 DOI: 10.1177/0963689717723016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [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
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Chiara Zannini
- 3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy.,4 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Nephrology, Dialysis and Renal Transplant, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elena Olivi
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Riccardo Tassinari
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,3 Ettore Sansavini Health Science Foundation ONLUS-Lab SWITH, Lugo, Italy
| | - Valentina Taglioli
- 2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,6 Department of Experimental, Diagnostic and Specialty Medicine, Laboratory of Experimental Cardiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Martina Rossi
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | | | | | - Giuliana Simonazzi
- 8 Division of Obstetrics and Prenatal Medicine, Department of Medical and Surgical Sciences, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Francesco Alviano
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Laura Bonsi
- 5 Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Carlo Ventura
- 1 GUNA - ATTRE (Advanced Therapies and Tissue Regeneration), Innovation Accelerator at CNR, Via Gobetti 101, 40129 Bologna, Italy.,2 National Institute of Biostructures and Biosystems (NIBB), Rome, Italy.,9 CNR, Institute of Organic Synthesis and Photoreactivity (Istituto per la Sintesi Organica e la Fotoreattività ISOF), Via Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
50
|
Sendon-Lago J, Seoane S, Martinez-Ordoñez A, Eiro N, Saa J, Vizoso FJ, Gonzalez F, Perez-Fernandez R, Bermudez MA. Corneal regeneration by conditioned medium of human uterine cervical stem cells is mediated by TIMP-1 and TIMP-2. Exp Eye Res 2019; 180:110-121. [PMID: 30557571 DOI: 10.1016/j.exer.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 01/15/2023]
Abstract
The aim of the present study was to evaluate the effect and the mechanism of action of the conditioned medium from human uterine cervical stem cells (CM-hUCESC) on corneal wound healing in a rabbit dry eye model. To do this, dry eye and corneal epithelial injuries were induced in rabbits by topical administration of atropine sulfate and NaOH. Hematoxylin-Eosin (H&E) and Ki-67 immunostaining were carried out to evaluate corneal damage and cell proliferation, and real-time PCR was used to evaluate proinflammatory cytokines in the cornea. In addition, in order to investigate possible factors involved in corneal regeneration, primary cultures of rat corneal epithelial cells (rCECs) were used to evaluate cell migration, proliferation, and apoptosis before and after immunoprecipitation of specific factors from the CM-hUCESC. Results showed that CM-hUCESC treatment significantly improved epithelial regeneration in rabbits with dry eye induced by atropine and reduced corneal pro-inflammatory TNF-α, MCP-1, MIP-1α and IL-6 cytokines. In addition, metalloproteinase inhibitors TIMP-1 and TIMP-2, which are present at high levels in CM-hUCESC, mediated corneal regenerative effects by both inducing corneal epithelial cell proliferation and inhibiting apoptosis. In summary, CM-hUCESC induces faster corneal regeneration in a rabbit model of dry eye induced by atropine than conventional treatments, being TIMP-1 and TIMP-2 mediators in this process. The results indicate that an alternative CM-based treatment for some corneal conditions is achievable, although future studies would be necessary to investigate other factors involved in the multiple observed effects of CM-hUCESC.
Collapse
Affiliation(s)
- Juan Sendon-Lago
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Samuel Seoane
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Anxo Martinez-Ordoñez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Noemi Eiro
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain.
| | - Jorge Saa
- Research Unit, Hospital Fundacion de Jove, Gijón, Spain; Service of Ophthalmology, Fundación Hospital de Jove, Gijón, Spain.
| | | | - Francisco Gonzalez
- Department of Surgery and CIMUS, University of Santiago de Compostela, Spain; Service of Ophthalmology and IDIS, Complejo Hospitalario Universitario de Santiago de Compostela, Spain.
| | - Roman Perez-Fernandez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Maria A Bermudez
- Department of Biology, Faculty of Science, University of A Coruña, Spain.
| |
Collapse
|