1
|
Alivernini S, Masserdotti A, Magatti M, Cargnoni A, Papait A, Silini AR, Romoli J, Ficai S, Di Mario C, Gremese E, Tolusso B, Parolini O. Exploring perinatal mesenchymal stromal cells as a potential therapeutic strategy for rheumatoid arthritis. Heliyon 2025; 11:e41438. [PMID: 39811302 PMCID: PMC11732555 DOI: 10.1016/j.heliyon.2024.e41438] [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: 08/28/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by inflammation in the synovial tissue, driven by aberrant activation of both the innate and adaptive immune systems, which can lead to irreversible disability. Despite the increasing therapeutic approaches for RA, only a low percentage of patients achieve sustained disease remission, and the persistence of immune dysregulation is likely responsible for disease recurrence once remission is attained. Cell therapy is an attractive, wide-spectrum strategy to modulate inflammation, and mesenchymal stromal cells (MSC) derived from perinatal tissues provide valuable tools for their use in regenerative medicine, mainly due to their immunomodulatory properties. Several in vitro studies have shown that perinatal MSC modulate the proliferation, maturation, and cytokine secretion profile of both innate and adaptive immune cells. Moreover, different beneficial effects have also been described when perinatal MSC were used to treat animal models of diseases associated with inflammatory conditions and degenerative processes. Specifically, in experimental models of RA, treatment with perinatal MSC resulted in a strong reduction of articular damage, which was associated with the modulation of both inflammation and activation of stromal resident cells in the synovial tissue. Here, we present in vitro and in vivo evidence supporting the use of perinatal MSC in RA. We also highlight the promising results from the few published clinical trials, which demonstrate the safety of perinatal MSC.
Collapse
Affiliation(s)
- Stefano Alivernini
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta R. Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Sara Ficai
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Clara Di Mario
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology Unit, Humanitas Research Hospital, Milan, Italy
| | - Barbara Tolusso
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
2
|
Ołdak M, Kurowska W, Plebańczyk M, Janicka I, Radzikowska A, Skalska U, Kuca-Warnawin E. Adipose-Derived Mesenchymal Stem Cells from Arthritis Patients: Differential Modulation of CD4⁺ T Cell Activation and Cytokine Production. Med Sci Monit 2024; 30:e945273. [PMID: 39543859 PMCID: PMC11575092 DOI: 10.12659/msm.945273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) from intra-articular adipose tissue of osteoarthritis (OA) and rheumatoid arthritis (RA) patients similarly regulate the proliferation of activated CD4⁺ T lymphocytes and exhibit comparable differentiation potential. This study aimed to assess the impact of ASCs from RA patients on CD4⁺ T cell activation and differentiation into Th17 and T regulatory (Treg) cells. MATERIAL AND METHODS Intra-articular adipose tissue samples were obtained from patients with RA and OA, who underwent knee replacement surgery. ASCs were isolated and cultured either with isolated CD4⁺ cells or with peripheral blood mononuclear cells. After culture, CD4⁺ T cell phenotype was evaluated by flow cytometry, and cytokine concentrations in culture supernatants were analyzed via ELISA. Blocking experiments were conducted to identify the soluble agents responsible for the immunomodulatory effects of ASCs. RESULTS RA- and OA-derived ASCs effectively modulated CD25 and CD69 expression on CD4⁺ cells. RA-derived ASCs failed to induce Tregs, decreased HLA-DR expression, and increased IL-35 production. RA- and OA-derived ASCs reduced TNF and IFN-γ production but increased IL-17 production. The immunomodulatory activities of ASCs were linked to the kynurenine pathway and prostaglandin E2. CONCLUSIONS This study indicates that ASCs modulate the phenotype of CD4⁺ T cells and influence the production of both pro-inflammatory and anti-inflammatory cytokines. However, ASCs from RA patients appear to have impaired immunomodulatory abilities, raising concerns about their therapeutic potential. Further research is needed to enhance our understanding of ASCs biology and their therapeutic utility.
Collapse
Affiliation(s)
- Maciej Ołdak
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Weronika Kurowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Iwona Janicka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Radzikowska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Urszula Skalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
3
|
Fu Y, Zhang C, Xie H, Wu Z, Tao Y, Wang Z, Gu M, Wei P, Lin S, Li R, He Y, Sheng J, Xu J, Wang J, Pan Y. Human umbilical cord mesenchymal stem cells alleviated TNBS-induced colitis in mice by restoring the balance of intestinal microbes and immunoregulation. Life Sci 2023; 334:122189. [PMID: 37865178 DOI: 10.1016/j.lfs.2023.122189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
AIMS Human umbilical cord mesenchymal stem cells (HUMSCs) have been documented to be effective for several immune disorders including inflammatory bowel diseases (IBD). However, it remains unclear how HUMSCs function in regulating immune responses and intestinal flora in the trinitrobenzene sulfonic acid (TNBS)-induced IBD model. MATERIALS AND METHODS We assessed the regulatory effects of HUMSCs on the gut microbiota, T lymphocyte subpopulations and related immune cytokines in the TNBS-induced IBD model. The mice were divided into the normal, TNBS, and HUMSC-treated groups. The effect of HUMSCs was evaluated by Hematoxylin and Eosin (H&E) staining, fluorescence-activated cell sorting (FACS), and enzyme-linked immunosorbent assay (ELISA) analyses. Metagenomics Illumina sequencing was conducted for fecal samples. KEY FINDINGS We demonstrated that the disease symptoms and pathological changes in the colon tissues of TNBS-induced colitis mice were dramatically ameliorated by HUMSCs, which improved the gut microbiota and rebalanced the immune system, increasing the abundance of healthy bacteria (such as Lactobacillus murinus and Lactobacillus johnsonii), the Firmicutes/Bacteroidetes ratio, and the proportion of Tregs; the Th1/Th17 ratio was decreased. Consistently, the expression levels of IFN-γ and IL-17 were significantly decreased, and transforming growth factor-β1 (TGF-β1) levels were significantly increased in the plasma of colitis mice HUMSC injection. SIGNIFICANCE Our experiment revealed that HUMSCs mitigate acute colitis by regulating the rebalance of Th1/Th17/Treg cells and related cytokines and remodeling the gut microbiota, providing potential future therapeutic targets in IBD.
Collapse
Affiliation(s)
- Yanxia Fu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- Chinese PLA General Hospital and Medical School, Beijing 100853, China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Hui Xie
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Zisheng Wu
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yurong Tao
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Ziyu Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Meng Gu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Shuye Lin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Ruoran Li
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Junfeng Xu
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| |
Collapse
|
4
|
Tavasolian F, Inman RD. Biology and therapeutic potential of mesenchymal stem cell extracellular vesicles in axial spondyloarthritis. Commun Biol 2023; 6:413. [PMID: 37059822 PMCID: PMC10104809 DOI: 10.1038/s42003-023-04743-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/21/2023] [Indexed: 04/16/2023] Open
Abstract
Axial spondyloarthritis (AxSpA) is a chronic, inflammatory, autoimmune disease that predominantly affects the joints of the spine, causes chronic pain, and, in advanced stages, may result in spinal fusion. Recent developments in understanding the immunomodulatory and tissue-differentiating properties of mesenchymal stem cell (MSC) therapy have raised the possibility of applying such treatment to AxSpA. The therapeutic effectiveness of MSCs has been shown in numerous studies spanning a range of diseases. Several studies have been conducted examining acellular therapy based on MSC secretome. Extracellular vesicles (EVs) generated by MSCs have been proven to reproduce the impact of MSCs on target cells. These EVs are associated with immunological regulation, tissue remodeling, and cellular homeostasis. EVs' biological effects rely on their cargo, with microRNAs (miRNAs) integrated into EVs playing a particularly important role in gene expression regulation. In this article, we will discuss the impact of MSCs and EVs generated by MSCs on target cells and how these may be used as unique treatment strategies for AxSpA.
Collapse
Affiliation(s)
- Fataneh Tavasolian
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
| | - Robert D Inman
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Extracellular Vesicle-Loaded Oncogenic lncRNA NEAT1 from Adipose-Derived Mesenchymal Stem Cells Confers Gemcitabine Resistance in Pancreatic Cancer via miR-491-5p/Snail/SOCS3 Axis. Stem Cells Int 2023; 2023:6510571. [PMID: 36762032 PMCID: PMC9902843 DOI: 10.1155/2023/6510571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023] Open
Abstract
It is becoming increasingly evident that key mechanisms of mesenchymal stem cell (MSC) efficacy appear to associate with paracrine activities, and the delivery of cargos through extracellular vesicles (EVs) controls the mechanistic actions of MSCs. Thus, this study clarified a possible mechanism by which EV-encapsulated NEAT1 from adipose-derived mesenchymal stem cells (ADSCs) might mediate gemcitabine resistance in pancreatic cancer (PCa). Microarray profile suggested a differentially expressed lncRNA NEAT1 in PCa, and we determined its expression in PCa cells. NEAT1 was found to be upregulated in PCa. The binding affinity among NEAT1, miR-491-5p, and Snail was identified through bioinformatic analysis and experimental validation. NEAT1 competitively bound to miR-491-5p to elevate Snail expression and diminish SOCS3 expression. PCa cells were cocultured with EVs extracted from ADSCs, followed by assessment of malignant phenotypes, tumorigenesis, and gemcitabine resistance of PCa cells using gain- or loss-of-function experiments. ADSC-derived EVs carrying NEAT1 promoted PCa cell proliferation, migration, and gemcitabine resistance in vitro and enhanced tumorigenicity in vivo by inhibiting miR-491-5p and SOCS3 and upregulating Snail. Collectively, the findings from our study found a new potential strategy for gemcitabine resistance in PCa by illustrating the mechanistic insights of oncogenic ADSC-derived EVs-loaded NEAT1 via regulating the miR-491-5p/Snail/SOCS3 axis.
Collapse
|
6
|
Fu Y, Li J, Li M, Xu J, Rong Z, Ren F, Wang Y, Sheng J, Chang Z. Umbilical Cord Mesenchymal Stem Cells Ameliorate Inflammation-Related Tumorigenesis via Modulating Macrophages. Stem Cells Int 2022; 2022:1617229. [PMID: 35694239 PMCID: PMC9178412 DOI: 10.1155/2022/1617229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been documented to be effective for the therapy of inflammation-related diseases but raised concerns on possible tumorigenic effects. Since most of the tumors are induced or promoted by chronic inflammation, one could expect that MSCs might be beneficial for the cancer therapy because of their potent roles on inhibiting inflammation. This study is aimed at performing a safety evaluation and evaluating the role of human umbilical cord mesenchymal stem cells (HUC-MSCs) on tumorigenesis. We found that HUC-MSCs cultured within 20 generations had no significant changes in proliferation, cell cycle, cellular senescence, apoptosis, and expression of mesenchymal stem cell markers. HUC-MSCs were unable to form any tumor in immunodeficiency or normal mice with or without inflammatory stimulation. Intriguingly, we observed that HUC-MSCs inhibited tumorigenesis in B16-derived or AOM/DSS-induced colon cancer models. We reasoned that the effect of HUC-MSCs on tumorigenesis might be through regulating the inflammatory response. Indeed, HUC-MSCs dramatically ameliorated the disease symptoms and pathological changes of DSS-induced colitis mice. We deciphered the mechanism that HUC-MSCs inhibited tumorigenesis through reducing the proportion of macrophages, which were decreased in the mice suffered from AOM/DSS-induced colon cancer. Correspondingly, the expression levels of TNF-α and IL-6, which were secreted by macrophages, were significantly decreased in the plasma of colon cancer and colitis mice after injection of HUC-MSCs. This study revealed the role of inhibiting macrophages and shed light on the therapeutic application of HUC-MSCs in inflammation-induced tumorigenesis.
Collapse
Affiliation(s)
- Yanxia Fu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Jun Li
- TsCell Biotech Inc., Beijing 100084, China
| | - Mengdi Li
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zheng Rong
- Department of Gynaecology and Obstetrics, Jishuitan Hospital, Beijing 100096, China
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
7
|
Enantioselectivity of Pentedrone and Methylone on Metabolic Profiling in 2D and 3D Human Hepatocyte-like Cells. Pharmaceuticals (Basel) 2022; 15:ph15030368. [PMID: 35337165 PMCID: PMC8953427 DOI: 10.3390/ph15030368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/12/2022] [Accepted: 03/12/2022] [Indexed: 02/04/2023] Open
Abstract
Pentedrone and methylone can express stereoselectivity in toxicokinetic and toxicodynamic processes. Similarly, their chiral discrimination in metabolism, which was not yet evaluated, can result in different metabolic profiles and subsequent hepatotoxic effects. Therefore, the aim of this work was to assess, for the first time, both the hepatic cytotoxic and metabolic profile of pentedrone and methylone enantiomers using physiologically relevant in vitro models. The hepatotoxicity of these compounds was observed in a concentration-dependent manner in human stem-cell-derived hepatocyte-like cells (HLCs) cultured under 3D (3D-HLCs) and 2D (2D-HLCs) conditions. Enantioselectivity, on the other hand, was only shown for pentedrone (1 mM) in 3D-HLCs, being R-(−)-pentedrone the most cytotoxic. Furthermore, the metabolic profile was initially evaluated in human liver microsomes (HLM) and further demonstrated in 3D-HLCs and 2D-HLCs applying a gas chromatography coupled to a mass spectrometer (GC–MS) technique. Methylone and pentedrone showed distinct and preferential metabolic routes for their enantiomers, resulting in the production of differentiated metabolites; R-(+)-methylone and R-(−)-pentedrone are the most metabolized enantiomers. In conclusion, the results demonstrated enantioselectivity for pentedrone and methylone in the metabolic processes, with enantioselectivity in cytotoxicity for pentedrone.
Collapse
|
8
|
Serras AS, Camões SP, Antunes B, Costa VM, Dionísio F, Yazar V, Vitorino R, Remião F, Castro M, Oliveira NG, Miranda JP. The Secretome of Human Neonatal Mesenchymal Stem Cells Modulates Doxorubicin-Induced Cytotoxicity: Impact in Non-Tumor Cells. Int J Mol Sci 2021; 22:ijms222313072. [PMID: 34884877 PMCID: PMC8657836 DOI: 10.3390/ijms222313072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (Dox) is one of the most widely used treatments for breast cancer, although limited by the well-documented cardiotoxicity and other off-target effects. Mesenchymal stem cell (MSC) secretome has shown immunomodulatory and regenerative properties, further potentiated under 3D conditions. This work aimed to uncover the effect of the MSC-derived secretome from 3D (CM3D) or 2D (CM2D) cultures, in human malignant breast cells (MDA-MB-231), non-tumor breast epithelial cells (MCF10A) and differentiated AC16 cardiomyocytes, co-treated with Dox. A comprehensive proteomic analysis of CM3D/CM2D was also performed to unravel the underlying mechanism. CM3D/CM2D co-incubation with Dox revealed no significant differences in MDA-MB-231 viability when compared to Dox alone, whereas MCF10A and AC16 viability was consistently improved in Dox+CM3D-treated cells. Moreover, neither CM2D nor CM3D affected Dox anti-migratory and anti-invasive effects in MDA-MB-231. Notably, Ge-LC-MS/MS proteomic analysis revealed that CM3D displayed protective features that might be linked to the regulation of cell proliferation (CAPN1, CST1, LAMC2, RANBP3), migration (CCN3, MMP8, PDCD5), invasion (TIMP1/2), oxidative stress (COX6B1, AIFM1, CD9, GSR) and inflammation (CCN3, ANXA5, CDH13, GDF15). Overall, CM3D decreased Dox-induced cytotoxicity in non-tumor cells, without compromising Dox chemotherapeutic profile in malignant cells, suggesting its potential use as a chemotherapy adjuvant to reduce off-target side effects.
Collapse
Affiliation(s)
- Ana S. Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
| | - Sérgio P. Camões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
| | - Bernardo Antunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
| | - Vera M. Costa
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.C.); (F.D.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Flávio Dionísio
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.C.); (F.D.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Volkan Yazar
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Rui Vitorino
- LAQV-REQUIMTE, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- Cardiovascular R&D Center, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Oporto, Portugal
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.C.); (F.D.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.S.S.); (S.P.C.); (B.A.); (M.C.); (N.G.O.)
- Correspondence:
| |
Collapse
|
9
|
An L, Chu T, Wang L, An S, Li Y, Hao H, Zhang Z, Yue H. Frequent injections of high-dose human umbilical cord mesenchymal stem cells slightly aggravate arthritis and skeletal muscle cachexia in collagen-induced arthritic mice. Exp Ther Med 2021; 22:1272. [PMID: 34594409 DOI: 10.3892/etm.2021.10707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
A single injection of low-dose human umbilical cord-derived mesenchymal stem cells (UC-MSCs) has been previously demonstrated to relieve synovitis and bone erosion in animal models of arthritis, but whether frequent injections of high-dose UC-MSCs relieve arthritis and inhibit loss of muscle mass has remained elusive. In the present study, DBA/1 mice were randomly divided into three groups: Normal (wild-type mice; n=11), collagen-induced arthritis (CIA; n=12) and CIA treated with UC-MSCs (n=11; 5x106 UC-MSCs per week for 3 weeks). Arthritis and skeletal muscle cachexia were evaluated until the end of the experiment on day 84. It was indicated that both the CIA and UC-MSC groups had lower body weights compared with the normal mice. Clinical arthritis scores, hind ankle diameters, synovitis and bone erosion progressively increased and were similar between the CIA and UC-MSC groups. Although there was no difference in food intake among the three groups, the normalized food intake of normal group was significantly higher than CIA group and UC-MSC group from day 42 onwards; there was no significance on day 77 but this could be neglected. Furthermore, gastrocnemius muscle weight in the UC-MSC group was significantly reduced compared with that in the CIA and normal groups. The UC-MSC group had higher levels of proinflammatory cytokines, such as TNF-α, IL-6 and IL-1β than those in the CIA group. However, the other cytokines assessed and the fibrosis indices in the CIA and UC-MSC groups were not different from those in the control group and there was no inflammatory cell infiltration. Thus, frequent injections of high-dose UC-MSCs slightly aggravated synovitis and muscle cachexia in the murine CIA model and should therefore be avoided in the treatment of arthritis.
Collapse
Affiliation(s)
- Lemei An
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Tianshu Chu
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Liujun Wang
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Songtao An
- Department of Cardiology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yalong Li
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Stem Cell Research Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Hongbo Hao
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center, City University of New York, New York, NY 10031, USA
| | - Zhuoli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Han Yue
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Stem Cell Research Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
10
|
Mesenchymal stem cells enhance Treg immunosuppressive function at the fetal-maternal interface. J Reprod Immunol 2021; 148:103366. [PMID: 34492568 DOI: 10.1016/j.jri.2021.103366] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/01/2021] [Accepted: 08/20/2021] [Indexed: 12/29/2022]
Abstract
Well-regulated maternal-fetal immune tolerance is a prerequisite for normal pregnancy. Hyperactivated immune cells and overwhelming inflammatory responses trigger adverse gestation outcome, such as recurrent spontaneous abortion (RSA). Local exacerbation of immunomodulatory cells in maternal decidua is a critical event, tightly linked with fetus acceptance. Owning to the notable immunoregulatory potentials, mesenchymal stromal cells (MSCs) and regulatory T cells (Tregs) have been separately reported as promising therapeutic approaches for refractory RSA attributable to certain immune disorders. However, the cross-talk between MSCs and Tregs at the fetal-maternal interface remains poorly understood. Here we revealed, for the first time, that umbilical MSCs could induce expansion of decidual Foxp3+CD4+ T cells with upregulated production of IL-10 and TGF-β. Meanwhile, MSCs reinforced the immune suppressive functions of decidual Tregs (dTregs). More important, MSCs-instructed dTregs gained enhanced capacity to suppress Th1 and Th17 related inflammatory responses. In vivo data demonstrated that adoptive transfer of MSCs obviously promoted accumulation of Foxp3+ dTregs in lipopolysaccharide (LPS)-induced mice abortion model and spontaneous abortion model (DBA/2-mated female CBA/J mice). Furthermore, MSCs treatment effectively ameliorated absorption rate in both models. This study may offer a new insight for the application of MSCs and Tregs in clinical recurrent miscarriage.
Collapse
|
11
|
Lopez-Santalla M, Bueren JA, Garin MI. Mesenchymal stem/stromal cell-based therapy for the treatment of rheumatoid arthritis: An update on preclinical studies. EBioMedicine 2021; 69:103427. [PMID: 34161884 PMCID: PMC8237294 DOI: 10.1016/j.ebiom.2021.103427] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and progressive joint destruction and is a primary cause of disability worldwide. Despite the existence of numerous anti-rheumatic drugs, a significant number of patients with RA do not respond or are intolerant to current treatments. Mesenchymal stem/stromal cell (MSCs) therapy represents a promising therapeutic tool to treat RA, mainly attributable to the immunomodulatory effects of these cells. This review comprises a comprehensive analysis of the scientific literature related to preclinical studies of MSC-based therapy in RA to analyse key aspects of current protocols as well as novel approaches which aim to improve the efficacy of MSC-based therapy.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM).
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM)
| | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM).
| |
Collapse
|
12
|
Lo WC, Tsai LW, Yang YS, Chan RWY. Understanding the Future Prospects of Synergizing Minimally Invasive Transforaminal Lumbar Interbody Fusion Surgery with Ceramics and Regenerative Cellular Therapies. Int J Mol Sci 2021; 22:3638. [PMID: 33807361 PMCID: PMC8037583 DOI: 10.3390/ijms22073638] [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: 02/16/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Transforaminal lumber interbody fusion (TLIF) is the last resort to address the lumber degenerative disorders such as spondylolisthesis, causing lower back pain. The current surgical intervention for these abnormalities includes open TLIF. However, in recent years, minimally invasive TLIF (MIS-TLIF) has gained a high momentum, as it could minimize the risk of infection, blood loss, and post-operative complications pertaining to fusion surgery. Further advancement in visualizing and guiding techniques along with grafting cage and materials are continuously improving the safety and efficacy of MIS-TLIF. These assistive techniques are also playing a crucial role to increase and improve the learning curve of surgeons. However, achieving an appropriate output through TLIF still remains a challenge, which might be synergized through 3D-printing and tissue engineering-based regenerative therapy. Owing to their differentiation potential, biomaterials such as stem/progenitor cells may contribute to restructuring lost or damaged tissues during MIS-TLIF, and this therapeutic efficacy could be further supplemented by platelet-derived biomaterials, leading to improved clinical outcomes. Thus, based on the above-mentioned strategies, we have comprehensively summarized recent developments in MIS-TLIF and its possible combinatorial regenerative therapies for rapid and long-term relief.
Collapse
Affiliation(s)
- Wen-Cheng Lo
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-S.Y.); (R.W.Y.C.)
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Lung-Wen Tsai
- Department of Medical Education and Research, Taipei Medical University Hospital, Taipei 11031, Taiwan;
| | - Yi-Shan Yang
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-S.Y.); (R.W.Y.C.)
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Ryan Wing Yuk Chan
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-S.Y.); (R.W.Y.C.)
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
13
|
Laundos TL, Vasques-Nóvoa F, Gomes RN, Sampaio-Pinto V, Cruz P, Cruz H, Santos JM, Barcia RN, Pinto-do-Ó P, Nascimento DS. Consistent Long-Term Therapeutic Efficacy of Human Umbilical Cord Matrix-Derived Mesenchymal Stromal Cells After Myocardial Infarction Despite Individual Differences and Transient Engraftment. Front Cell Dev Biol 2021; 9:624601. [PMID: 33614654 PMCID: PMC7890004 DOI: 10.3389/fcell.2021.624601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/11/2021] [Indexed: 11/24/2022] Open
Abstract
Human mesenchymal stem cells gather special interest as a universal and feasible add-on therapy for myocardial infarction (MI). In particular, human umbilical cord matrix-derived mesenchymal stromal cells (UCM-MSC) are advantageous since can be easily obtained and display high expansion potential. Using isolation protocols compliant with cell therapy, we previously showed UCM-MSC preserved cardiac function and attenuated remodeling 2 weeks after MI. In this study, UCM-MSC from two umbilical cords, UC-A and UC-B, were transplanted in a murine MI model to investigate consistency and durability of the therapeutic benefits. Both cellular products improved cardiac function and limited adverse cardiac remodeling 12 weeks post-ischemic injury, supporting sustained and long-term beneficial therapeutic effect. Donor associated variability was found in the modulation of cardiac remodeling and activation of the Akt-mTOR-GSK3β survival pathway. In vitro, the two cell products displayed similar ability to induce the formation of vessel-like structures and comparable transcriptome in normoxia and hypoxia, apart from UCM-MSCs proliferation and expression differences in a small subset of genes associated with MHC Class I. These findings support that UCM-MSC are strong candidates to assist the treatment of MI whilst calling for the discussion on methodologies to characterize and select best performing UCM-MSC before clinical application.
Collapse
Affiliation(s)
- Tiago L. Laundos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Cardiovascular RandD Center, Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Internal Medicine, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Rita N. Gomes
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Vasco Sampaio-Pinto
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | | | | | | | | | - Perpétua Pinto-do-Ó
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Diana S. Nascimento
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
14
|
Storozhylova N, Crecente-Campo J, Cabaleiro D, Lugo L, Dussouy C, Simões S, Monteiro M, Grandjean C, Alonso MJ. An In Situ Hyaluronic Acid-Fibrin Hydrogel Containing Drug-Loaded Nanocapsules for Intra-Articular Treatment of Inflammatory Joint Diseases. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00154-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
15
|
Kong CM, Lin HD, Biswas A, Bongso A, Fong CY. Manufacturing of human Wharton's jelly stem cells for clinical use: selection of serum is important. Cytotherapy 2019; 21:483-495. [PMID: 30879965 DOI: 10.1016/j.jcyt.2019.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Human Wharton's jelly-derived mesenchymal stromal cells (hWJSCs) have gained considerable attention for their use in cell therapy. Many of these applications would require manufacturing of millions of hWJSCs. It is, therefore, necessary to develop a Good Manufacturing Practice (GMP)-compliant hWJSC expansion protocol, allowing the generation of a large quantity of cells to meet both clinical and regulatory requirements. Here, we compared human platelet lysate (HPL) and human serum (HS) in supporting clinical-grade hWJSC expansion. METHODS hWJSCs were successfully isolated from six different umbilical cords using GMP-compliant dissociation enzymes. Freshly isolated hWJSCs were cultured in media supplemented with 10% of one of the following sera: fetal bovine serum (FBS), HPL and HS. Properties of the expanded hWJSCs were analyzed. RESULTS We showed that GMP-compliant dissociation enzymes were as efficient as research-grade dissociation enzymes in isolating hWJSCs. hWJSC fresh cell yield and cell viability using HPL and HS supplementations were at greater advantages than FBS. Moreover, hWJSCs expanded in HPL and HS supplementations not only preserved classical MSCs phenotypes and differentiation potential to adipocytes, osteocytes and chondrocytes, they also enhanced the migration of skin fibroblasts. However, HS, unlike HPL, did not alter immunogenicity properties of hWJSCs. hWJSCs expanded in HS supplementation also exerted greater immunosuppressive action in inhibiting T-cell proliferation and increased extracellular matrix (ECM) gene expression, making them useful in tissue repair clinical application. CONCLUSION Our findings indicate that HS can be considered as a promising and safer alternative to FBS, and should be recommended for clinical-grade expansion of hWJSCs.
Collapse
Affiliation(s)
- Chiou Mee Kong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Hao Daniel Lin
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Arijit Biswas
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Ariff Bongso
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore.
| | - Chui-Yee Fong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore.
| |
Collapse
|
16
|
Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, Cruz P, Cruz H, Simões S, Santos JM. The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front Immunol 2019; 10:18. [PMID: 30804924 PMCID: PMC6370626 DOI: 10.3389/fimmu.2019.00018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder whose treatment is mostly restricted to pain and symptom management and to the delay of joint destruction. Mesenchymal stem/stromal cells from the umbilical cord tissue (UC-MSCs) have previously been proven to be immunomodulatory and more efficient than bone marrow-derived MSCs in causing remission of local and systemic arthritic manifestations in vivo. Given the paracrine nature of UC-MSC activity, their application as active substances can be replaced by their secretome, thus avoiding allogeneic rejection and safety issues related to unwanted grafting. In this work, we aimed at demonstrating the viability of applying the 3D-primed UC-MSC secretome for the amelioration of arthritic signs. A proteomic analysis was performed to both, media conditioned by UC-MSC monolayer (CM2D) and 3D cultures (CM3D). The analysis of relevant trophic factors confirmed secretome profiles with very significant differences in terms of therapeutic potential. Whereas, CM3D was characterised by a prevailing expression of anti-inflammatory cytokines such as IL-10 and LIF, along with trophic factors involved in different mechanisms leading to tissue regeneration, such as PDGF-BB, FGF-2, I-309, SCF, and GM-CSF; CM2D presented relatively higher levels of IL-6, MCP-1, and IL-21, with recognised pro-inflammatory roles in joint disease and pleiotropic effects in the progression of rheumatoid arthritis (RA). Accordingly, different motogenic effects over mouse chondrocytes and distinct capacities of inducing glycosaminoglycan synthesis in vitro were observed between CM3D and CM2D. Finally, the evaluation of arthritic manifestations in vivo, using an adjuvant-induced model for arthritis (AIA), suggested a significantly higher therapeutic potential of CM3D over CM2D and even UC-MSCs. Histological analysis confirmed a faster remission of local and systemic arthritic manifestations of CM3D-treated animals. Overall, the results show that the use of UC-MSC CM3D is a viable and better strategy than direct UC-MSC administration for counteracting AIA-related signs. This strategy represents a novel MSC-based but nonetheless cell-free treatment for arthritic conditions such as those characterising RA.
Collapse
Affiliation(s)
- Joana P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Sérgio P Camões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Maria M Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Joana S Rodrigues
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Manuela Carvalheiro
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | | | | | | | - Sandra Simões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Jorge M Santos
- ECBio S.A., Amadora, Portugal.,Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente, Universidade do Porto, Porto, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
17
|
Loy H, Kuok DIT, Hui KPY, Choi MHL, Yuen W, Nicholls JM, Peiris JSM, Chan MCW. Therapeutic Implications of Human Umbilical Cord Mesenchymal Stromal Cells in Attenuating Influenza A(H5N1) Virus-Associated Acute Lung Injury. J Infect Dis 2019; 219:186-196. [PMID: 30085072 PMCID: PMC6306016 DOI: 10.1093/infdis/jiy478] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Background Highly pathogenic avian influenza viruses can cause severe forms of acute lung injury (ALI) in humans, where pulmonary flooding leads to respiratory failure. The therapeutic benefits of bone marrow mesenchymal stromal cells (MSCs) have been demonstrated in a model of ALI due to influenza A(H5N1) virus. However, clinical translation is impractical and limited by a decline in efficacy as the age of the donor increases. Umbilical cord MSCs (UC-MSCs) are easier to obtain by comparison, and their primitive source may offer more-potent therapeutic effects. Methods Here we investigate the therapeutic efficacy of UC-MSCs on the mechanisms of pulmonary edema formation and alveolar fluid clearance and protein permeability of A(H5N1)-infected human alveolar epithelial cells. UC-MSCs were also tested in a mouse model of influenza ALI. Results We found that UC-MSCs were effective in restoring impaired alveolar fluid clearance and protein permeability of A(H5N1)-infected human alveolar epithelial cells. UC-MSCs consistently outperformed bone marrow MSCs, partly because of greater growth factor secretion of angiopoietin 1 and hepatocyte growth factor. Conditioned UC-MSC medium and UC-MSC exosomes were also able to recapitulate these effects. However, UC-MSCs only slightly improved survival of A(H5N1)-infected mice. Conclusions Our results suggest that UC-MSCs are effective in restoring alveolar fluid clearance and protein permeability in A(H5N1)-associated ALI and confer functional in addition to practical advantages over conventional bone marrow MSCs.
Collapse
Affiliation(s)
- Hayley Loy
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Denise I T Kuok
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kenrie P Y Hui
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Miranda H L Choi
- Healthbaby Biotech, Hong Kong Special Administrative Region, China
| | - W Yuen
- Healthbaby Biotech, Hong Kong Special Administrative Region, China
| | - John M Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
18
|
Leyendecker A, Pinheiro CCG, Amano MT, Bueno DF. The Use of Human Mesenchymal Stem Cells as Therapeutic Agents for the in vivo Treatment of Immune-Related Diseases: A Systematic Review. Front Immunol 2018; 9:2056. [PMID: 30254638 PMCID: PMC6141714 DOI: 10.3389/fimmu.2018.02056] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background: One of the greatest challenges for medicine is to find a safe and effective treatment for immune-related diseases. However, due to the low efficacy of the treatment available and the occurrence of serious adverse effects, many groups are currently searching for alternatives to the traditional therapy. In this regard, the use of human mesenchymal stem cells (hMSCs) represents a great promise for the treatment of a variety of immune-related diseases due to their potent immunomodulatory properties. The main objective of this study is, therefore, to present and summarize, through a systematic review of the literature, in vivo studies in which the efficacy of the administration of hMSCs for the treatment of immune-related diseases was evaluated. Methods: The article search was conducted in PubMed/MEDLINE, Scopus and Web of Science databases. Original research articles assessing the therapeutic potential of hMSCs administration for the in vivo treatment immune-related diseases, published from 1984 to December 2017, were selected and evaluated. Results: A total of 132 manuscripts formed the basis of this systematic review. Most of the studies analyzed reported positive results after hMSCs administration. Clinical effects commonly observed include an increase in the survival rates and a reduction in the severity and incidence of the immune-related diseases studied. In addition, hMSCs administration resulted in an inhibition in the proliferation and activation of CD19+ B cells, CD4+ Th1 and Th17 cells, CD8+ T cells, NK cells, macrophages, monocytes, and neutrophils. The clonal expansion of both Bregs and Tregs cells, however, was stimulated. Administration of hMSCs also resulted in a reduction in the levels of pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-1, IL-2, IL-12, and IL-17 and in an increase in the levels of immunoregulatory cytokines such as IL-4, IL-10, and IL-13. Conclusions: The results obtained in this study open new avenues for the treatment of immune-related diseases through the administration of hMSCs and emphasize the importance of the conduction of further studies in this area.
Collapse
|
19
|
Chen ZY, Hu YY, Hu XF, Cheng LX. The conditioned medium of human mesenchymal stromal cells reduces irradiation-induced damage in cardiac fibroblast cells. JOURNAL OF RADIATION RESEARCH 2018; 59:555-564. [PMID: 30010837 PMCID: PMC6151644 DOI: 10.1093/jrr/rry048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Indexed: 05/04/2023]
Abstract
Recently, multipotent mesenchymal stromal cell (MSC) treatment has attracted special attention as a new alternative strategy for stimulating regeneration. Irradiation myocardial fibrosis (IMF) is a major complication associated with total body irradiation for hematopoietic stem cell transplantation, nuclear accidents, and thoracic radiotherapy for lung cancer, esophageal cancer, proximal gastric cancer, breast cancer, thymoma, and lymphoma. The aim of the present study was to assess the therapeutic paracrine effects of human umbilical cord-derived mesenchymal stromal cells (UC-MSCs) in the cell model of IMF. For this purpose, primary human cardiac fibroblasts (HCF) cells were irradiated and cultured with the conditioned medium of UC-MSCs (MSCCM). MSCCM promoted cell viability, reduced collagen deposition as measured by Sircol assay and qPCR (Col1A1 and Col1A2), prevented oxidative stress and increased antioxidant status (as measured by malondialdehyde content and the activities and mRNA levels of antioxidant enzymes), and reduced pro-fibrotic TGF-β1, IL-6 and IL-8 levels (as examined by ELISA kit and qPCR). Pretreatment with inhibitor of NF-κB led to a decrease in the levels of TGF-β1 in cell lysate of HCF cells by ELISA kit. Furthermore, we also found that MSCCM prevented NF-κB signaling pathway activation for its proinflammatory actions induced by irradiation. Taken together, our data suggest that MSCCM could reduce irradiation-induced TGF-β1 production through inhibition of the NF-κB signaling pathway. These data provide new insights into the functional actions of MSCCM on irradiation myocardial fibrosis.
Collapse
Affiliation(s)
- Zhu-Yue Chen
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie-Fang Avenue 1277#, Wuhan, China
| | - Ying-Ying Hu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie-Fang Avenue 1277#, Wuhan, China
| | - Xiao-Fan Hu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie-Fang Avenue 1277#, Wuhan, China
| | - Long-Xian Cheng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie-Fang Avenue 1277#, Wuhan, China
- Corresponding author. Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical Collegeof Huazhong University of Science and Technology, Jie-Fang Avenue 1277#, Wuhan, 430022, China. Tel: +86-27-85726462; Fax: +86-27-85726423;
| |
Collapse
|
20
|
Umbilical cord tissue–derived mesenchymal stromal cells maintain immunomodulatory and angiogenic potencies after cryopreservation and subsequent thawing. Cytotherapy 2017; 19:360-370. [DOI: 10.1016/j.jcyt.2016.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/26/2016] [Accepted: 11/08/2016] [Indexed: 12/22/2022]
|
21
|
Repair of Osteochondral Defects Using Human Umbilical Cord Wharton's Jelly-Derived Mesenchymal Stem Cells in a Rabbit Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8760383. [PMID: 28261617 PMCID: PMC5316442 DOI: 10.1155/2017/8760383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022]
Abstract
Umbilical cord Wharton's jelly-derived mesenchymal stem cell (WJMSC) is a new-found mesenchymal stem cell in recent years with multiple lineage potential. Due to its abundant resources, no damage procurement, and lower immunogenicity than other adult MSCs, WJMSC promises to be a good xenogenous cell candidate for tissue engineering. This in vivo pilot study explored the use of human umbilical cord Wharton's jelly mesenchymal stem cells (hWJMSCs) containing a tissue engineering construct xenotransplant in rabbits to repair full-thickness cartilage defects in the femoral patellar groove. We observed orderly spatial-temporal remodeling of hWJMSCs into cartilage tissues during repair over 16 months, with characteristic architectural features, including a hyaline-like neocartilage layer with good surface regularity, complete integration with adjacent host cartilage, and regenerated subchondral bone. No immune rejection was detected when xenograft hWJMSCs were implanted into rabbit cartilage defects. The repair results using hWJMSCs were superior to those of chondrogenically induced hWJMSCs after assessing gross appearance and histological grading scores. These preliminary results suggest that using novel undifferentiated hWJMSCs as seed cells might be a better approach than using transforming growth factor-β-induced differentiated hWJMSCs for in vivo tissue engineering treatment of cartilage defects. hWJMSC allografts may be promising for clinical applications.
Collapse
|
22
|
Cipriano M, Correia JC, Camões SP, Oliveira NG, Cruz P, Cruz H, Castro M, Ruas JL, Santos JM, Miranda JP. The role of epigenetic modifiers in extended cultures of functional hepatocyte-like cells derived from human neonatal mesenchymal stem cells. Arch Toxicol 2016; 91:2469-2489. [PMID: 27909741 DOI: 10.1007/s00204-016-1901-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/24/2016] [Indexed: 01/06/2023]
Abstract
The development of predictive in vitro stem cell-derived hepatic models for toxicological drug screening is an increasingly important topic. Herein, umbilical cord tissue-derived mesenchymal stem cells (hnMSCs) underwent hepatic differentiation using an optimized three-step core protocol of 24 days that mimicked liver embryogenesis with further exposure to epigenetic markers, namely the histone deacetylase inhibitor trichostatin A (TSA), the cytidine analogue 5-azacytidine (5-AZA) and dimethyl sulfoxide (DMSO). FGF-2 and FGF-4 were also tested to improve endoderm commitment and foregut induction during Step 1 of the differentiation protocol, being HHEX expression increased with FGF-2 (4 ng/mL). DMSO (1%, v/v) when added at day 10 enhanced cell morphology, glycogen storage ability, enzymatic activity and induction capacity. Moreover, the stability of the hepatic phenotype under the optimized differentiation conditions was examined up to day 34. Our findings showed that hepatocyte-like cells (HLCs) acquired the ability to metabolize glucose, produce albumin and detoxify ammonia. Global transcriptional analysis of the HLCs showed a partial hepatic differentiation degree. Global analysis of gene expression in the different cells revealed shared expression of gene groups between HLCs and human primary hepatocytes (hpHeps) that were not observed between HepG2 and hpHeps. In addition, bioinformatics analysis of gene expression data placed HLCs between the HepG2 cell line and hpHeps and distant from hnMSCs. The enhanced hepatic differentiation observed was supported by the presence of the hepatic drug transporters OATP-C and MRP-2 and gene expression of the hepatic markers CK18, TAT, AFP, ALB, HNF4A and CEBPA; and by their ability to display stable UGT-, EROD-, ECOD-, CYP1A1-, CYP2C9- and CYP3A4-dependent activities at levels either comparable with or even higher than those observed in primary hepatocytes and HepG2 cells. Overall, an improvement of the hepatocyte-like phenotype was achieved for an extended culture time suggesting a role of the epigenetic modifiers in hepatic differentiation and maturation and presenting hnMSC-HLCs as an advantageous alternative for drug discovery and in vitro toxicology testing.
Collapse
Affiliation(s)
- M Cipriano
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - J C Correia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - S P Camões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - N G Oliveira
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - P Cruz
- ECBio S.A., Amadora, Portugal
| | - H Cruz
- ECBio S.A., Amadora, Portugal
| | - M Castro
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - J L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - J P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
23
|
Pereira ARS, Mendes TF, Ministro A, Teixeira M, Filipe M, Santos JM, Bárcia RN, Goyri-O'Neill J, Pinto F, Cruz PE, Cruz HJ, Santos SCR. Therapeutic angiogenesis induced by human umbilical cord tissue-derived mesenchymal stromal cells in a murine model of hindlimb ischemia. Stem Cell Res Ther 2016; 7:145. [PMID: 27680210 PMCID: PMC5041588 DOI: 10.1186/s13287-016-0410-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/09/2016] [Indexed: 11/10/2022] Open
Abstract
Background Mesenchymal stem cells derived from human umbilical cord tissue, termed UCX®, have the potential to promote a full range of events leading to tissue regeneration and homeostasis. The main goal of this work was to investigate UCX® action in experimentally induced hindlimb ischemia (HLI). Methods UCX®, obtained by using a proprietary technology developed by ECBio (Amadora, Portugal), were delivered via intramuscular injection to C57BL/6 females after unilateral HLI induction. Perfusion recovery, capillary and collateral density increase were evaluated by laser doppler, CD31 immunohistochemistry and diaphonisation, respectively. The activation state of endothelial cells (ECs) was analysed after EC isolation by laser capture microdissection microscopy followed by RNA extraction, cDNA synthesis and quantitative RT-PCR analysis. The UCX®-conditioned medium was analysed on Gallios flow cytometer. The capacity of UCX® in promoting tubulogenesis and EC migration was assessed by matrigel tubule formation and wound-healing assay, respectively. Results We demonstrated that UCX® enhance angiogenesis in vitro via a paracrine effect. Importantly, after HLI induction, UCX® improve blood perfusion by stimulating angiogenesis and arteriogenesis. This is achieved through a new mechanism in which durable and simultaneous upregulation of transforming growth factor β2, angiopoietin 2, fibroblast growth factor 2, and hepatocyte growth factor, in endothelial cells is induced by UCX®. Conclusions In conclusion, our data demonstrate that UCX® improve the angiogenic potency of endothelial cells in the murine ischemic limb suggesting the potential of UCX® as a new therapeutic tool for critical limb ischemia.
Collapse
Affiliation(s)
- Ana Rita S Pereira
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Teresa F Mendes
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Augusto Ministro
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.,Centro Hospitalar Lisboa Norte, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal
| | - Mariana Teixeira
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Mariana Filipe
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Jorge M Santos
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Rita N Bárcia
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - J Goyri-O'Neill
- Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Fausto Pinto
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.,Faculdade de Medicina da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal
| | - Pedro E Cruz
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Helder J Cruz
- ECBio, Investigação e Desenvolvimento em Biotecnologia S.A., R. Henrique Paiva Couceiro, 27, 2700-4511, Amadora, Portugal
| | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal. .,Faculdade de Medicina da Universidade de Lisboa, Av. Prof Egas Moniz, 1649-028, Lisbon, Portugal.
| |
Collapse
|
24
|
Human Umbilical Cord Mesenchymal Stromal Cell Transplantation in Myocardial Ischemia (HUC-HEART Trial). A Study Protocol of a Phase 1/2, Controlled and Randomized Trial in Combination with Coronary Artery Bypass Grafting. Stem Cell Rev Rep 2016; 11:752-60. [PMID: 26123356 DOI: 10.1007/s12015-015-9601-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs), which may be obtained from the bone marrow, have been studied for more than a decade in the setting of coronary artery disease (CAD). Adipose tissue-derived MSCs have recently come into focus and are being tested in a series of clinical trials. MSC-like cells have also been derived from a variety of sources, including umbilical cord stroma, or HUC-MSCs. The HUC-HEART trail (ClinicalTrials.gov Identifier: NCT02323477) is a phase 1/2, controlled, multicenter, randomized clinical study of the intramyocardial delivery of allogeneic HUC-MSCs in patients with chronic ischemic cardiomyopathy. A total of 79 patients (ages 30-80) with left ventricle ejection fractions ranging between 25 and 45% will be randomized in a 2:1:1 pattern in order to receive an intramyocardial injection of either HUC-MSCs or autologous bone marrow-derived mononuclear cells (BM-MNCs) in combination with coronary arterial bypass grafting (CABG) surgery. The control group of patients will receive no cells and undergo CABG alone. Human HUC-MSCs will be isolated, propagated and banked in accordance with a cGMP protocol, whereas the autologous BM-MNCs will be isolated via aspiration from the iliac crest and subsequently process in a closed-circuit cell purification system shortly before cell transplantation. The cell injections will be implemented in 10 peri-infarct areas. Baseline and post-transplantation outcome measures will be primarily utilized to test both the safety and the efficacy of the administered cells for up to 12 months.
Collapse
|
25
|
Current View on Osteogenic Differentiation Potential of Mesenchymal Stromal Cells Derived from Placental Tissues. Stem Cell Rev Rep 2016; 11:570-85. [PMID: 25381565 PMCID: PMC4493719 DOI: 10.1007/s12015-014-9569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSC) isolated from human term placental tissues possess unique characteristics, including their peculiar immunomodulatory properties and their multilineage differentiation potential. The osteogenic differentiation capacity of placental MSC has been widely disputed, and continues to be an issue of debate. This review will briefly discuss the different MSC populations which can be obtained from different regions of human term placenta, along with their unique properties, focusing specifically on their osteogenic differentiation potential. We will present the strategies used to enhance osteogenic differentiation potential in vitro, such as through the selection of subpopulations more prone to differentiate, the modification of the components of osteo-inductive medium, and even mechanical stimulation. Accordingly, the applications of three-dimensional environments in vitro and in vivo, such as non-synthetic, polymer-based, and ceramic scaffolds, will also be discussed, along with results obtained from pre-clinical studies of placental MSC for the regeneration of bone defects and treatment of bone-related diseases.
Collapse
|
26
|
Shimazu T, Mori Y, Takahashi A, Tsunoda H, Tojo A, Nagamura-Inoue T. Serum- and xeno-free cryopreservation of human umbilical cord tissue as mesenchymal stromal cell source. Cytotherapy 2016; 17:593-600. [PMID: 25881518 DOI: 10.1016/j.jcyt.2015.03.604] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/02/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AIMS Human umbilical cord (UC) has become a notable source for mesenchymal stromal cells (MSCs) that can migrate to areas of inflammation and damaged tissue and can suppress excess immune reactions and to repair, respectively. Although UC is a solid tissue, there are several advantages, including repeatable uses from the same donor sample when needed and the possibility of future explorations for cells with unknown potential, if we could cryopreserve the UC as a living tissue material. However, because the cryoprotectants in the previous reports included animal- or allogeneic human-derived serum or no serum, the frozen-thawed UC-MSCs were inferior to fresh UC-MSCs in cell proliferation. The objective of this study was to find a suitable cryopreservation method of UC for clinical use. METHODS The UC was cut in cross-section and incised longitudinally, immersed in the cryoprotectant and frozen slowly. Later, it was thawed and minced rapidly, and the fragments of UC were cultured by improved explant method. RESULTS The highest yield of cells was obtained from frozen-thawed UC with serum- and xeno-free cryoprotectant, STEM-CELLBANKER, when compared with others. The cells derived from frozen-thawed UC stored in STEM-CELLBANKER expressed the phenotypes of MSCs, retained the immunosuppressive properties in allogeneic mixed lymphocyte reactions and the differentiation potentials (into adipocyte and chondrocytes) comparable to those derived from fresh UC. CONCLUSIONS UC can be cryopreserved in serum- and xeno-free cryoprotectant as a living tissue while keeping its growth and functions equivalent to fresh UC. Our method is simple and feasible for clinical use.
Collapse
Affiliation(s)
- Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | - Arinobu Tojo
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
27
|
Zarbakhsh S, Goudarzi N, Shirmohammadi M, Safari M. Histological Study of Bone Marrow and Umbilical Cord Stromal Cell Transplantation in Regenerating Rat Peripheral Nerve. CELL JOURNAL 2016; 17:668-77. [PMID: 26862526 PMCID: PMC4746417 DOI: 10.22074/cellj.2016.3839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/05/2015] [Indexed: 12/15/2022]
Abstract
Objective Bone marrow and umbilical cord stromal cells are multipotential stem cells
that have the ability to produce growth factors that play an important role in survival and
generation of axons. The goal of this study was to evaluate the effects of the two different
mesenchymal stem cells on peripheral nerve regeneration.
Materials and Methods In this experimental study, a 10 mm segment of the left sciatic
nerve of male Wistar rats (250-300 g) was removed with a silicone tube interposed into
this nerve gap. Bone marrow stromal cells (BMSCs) and human umbilical cord stromal
cells (HUCSCs) were respectively obtained from rat and human. The cells were sepa-
rately cultured and transplanted into the nerve gap. The sciatic nerve regeneration was
evaluated by immunohistochemistry, and light and electron microscopy. Moreover, histo-
morphology of the gastrocnemius muscle was observed.
Results The nerve regeneration in the BMSCs and HUCSCs groups that had received
the stem cells was significantly more favorable than the control group. In addition, the BM-
SCs group was significantly more favorable than the HUCSCs group (P<0.05).
Conclusion The results of this study suggest that both homograft BMSCs and het-
erograft HUCSCs may have the potential to regenerate peripheral nerve injury and
transplantation of BMSCs may be more effective than HUCSCs in rat.
Collapse
Affiliation(s)
- Sam Zarbakhsh
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasim Goudarzi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shirmohammadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Manouchehr Safari
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
28
|
Arutyunyan I, Elchaninov A, Makarov A, Fatkhudinov T. Umbilical Cord as Prospective Source for Mesenchymal Stem Cell-Based Therapy. Stem Cells Int 2016; 2016:6901286. [PMID: 27651799 PMCID: PMC5019943 DOI: 10.1155/2016/6901286] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/14/2016] [Indexed: 02/07/2023] Open
Abstract
The paper presents current evidence on the properties of human umbilical cord-derived mesenchymal stem cells, including origin, proliferative potential, plasticity, stability of karyotype and phenotype, transcriptome, secretome, and immunomodulatory activity. A review of preclinical studies and clinical trials using this cell type is performed. Prospects for the use of mesenchymal stem cells, derived from the umbilical cord, in cell transplantation are associated with the need for specialized biobanking and transplant standardization criteria.
Collapse
Affiliation(s)
- Irina Arutyunyan
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
| | - Andrey Elchaninov
- 2Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, No. 1, Ostrovitianov Street, Moscow 117997, Russia
| | - Andrey Makarov
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
| | - Timur Fatkhudinov
- 1Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, No. 4, Oparin Street, Moscow 117997, Russia
- 2Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, No. 1, Ostrovitianov Street, Moscow 117997, Russia
- *Timur Fatkhudinov:
| |
Collapse
|
29
|
Fazzina R, Mariotti A, Procoli A, Fioravanti D, Iudicone P, Scambia G, Pierelli L, Bonanno G. A new standardized clinical-grade protocol for banking human umbilical cord tissue cells. Transfusion 2015; 55:2864-73. [PMID: 26354088 DOI: 10.1111/trf.13277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/14/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) isolated from human umbilical cord tissue (UCT) can be considered the perfect candidates for cell-based therapies and regenerative medicine. UCT-derived MSCs can be cryogenically stored in cell banks and expanded as needed for therapeutic uses. STUDY DESIGN AND METHODS We developed a new method for UCT-MSC isolation, cryopreservation, and expansion, following all criteria required by a stem cell bank. UCT-MSCs were isolated either by manual dissociation (MM) or by a semiautomatic dissociation system (SAM). In both protocols UCTs were treated enzymatically using Type IV collagenase good manufacturing practices (GMP) graded and hyaluronidase (medicinal product). Isolated UCT-MSCs were cryopreserved and analyzed after thawing for phenotype; for proliferation rate; and for their osteogenic, adipogenic, and chondrogenic differentiation capabilities. RESULTS We found that SAM reduced the time of tissue enzyme exposure and enabled us to obtain a homogeneous single-cell suspension deprived of tissue fragments. The isolated cells in both groups showed high expression of MSC markers CD105, CD73, and CD90 and similar differentiation capabilities, phenotype, and proliferation potential. Moreover, the final yield of MSCs was comparable between the two techniques. CONCLUSION In this study, we have established a reliable and standardized protocol to isolate UCT-MSCs from UCT for cell banking purposes. Processing the whole umbilical tissue with GMP-graded enzymes using a semiautomatic dissociator allowed us to obtain a single-cell suspension product with a known number of isolated cells that can be cryopreserved right after isolation and thawed as needed for expansion and clinical use.
Collapse
Affiliation(s)
| | - Andrea Mariotti
- Institute of Gynecology and Obstetrics, Sacred Heart Catholic University
| | - Annabella Procoli
- Institute of Gynecology and Obstetrics, Sacred Heart Catholic University
| | - Daniela Fioravanti
- Immunohematology and Transfusion Medicine, San Camillo Forlanini Hospital
| | - Paola Iudicone
- Immunohematology and Transfusion Medicine, San Camillo Forlanini Hospital
| | - Giovanni Scambia
- Institute of Gynecology and Obstetrics, Sacred Heart Catholic University
| | - Luca Pierelli
- Immunohematology and Transfusion Medicine, San Camillo Forlanini Hospital.,Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Giuseppina Bonanno
- Institute of Gynecology and Obstetrics, Sacred Heart Catholic University
| |
Collapse
|
30
|
What Makes Umbilical Cord Tissue-Derived Mesenchymal Stromal Cells Superior Immunomodulators When Compared to Bone Marrow Derived Mesenchymal Stromal Cells? Stem Cells Int 2015; 2015:583984. [PMID: 26064137 PMCID: PMC4443932 DOI: 10.1155/2015/583984] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/08/2015] [Accepted: 04/26/2015] [Indexed: 12/15/2022] Open
Abstract
MSCs derived from the umbilical cord tissue, termed UCX, were investigated for their immunomodulatory properties and compared to bone marrow-derived MSCs (BM-MSCs), the gold-standard in immunotherapy. Immunogenicity and immunosuppression were assessed by mixed lymphocyte reactions, suppression of lymphocyte proliferation and induction of regulatory T cells. Results showed that UCX were less immunogenic and showed higher immunosuppression activity than BM-MSCs. Further, UCX did not need prior activation or priming to exert their immunomodulatory effects. This was further corroborated in vivo in a model of acute inflammation. To elucidate the potency differences observed between UCX and BM-MSCs, gene expression related to immune modulation was analysed in both cell types. Several gene expression profile differences were found between UCX and BM-MSCs, namely decreased expression of HLA-DRA, HO-1, IGFBP1, 4 and 6, ILR1, IL6R and PTGES and increased expression of CD200, CD273, CD274, IL1B, IL-8, LIF and TGFB2. The latter were confirmed at the protein expression level. Overall, these results show that UCX seem to be naturally more potent immunosuppressors and less immunogenic than BM-MSCs. We propose that these differences may be due to increased levels of immunomodulatory surface proteins such as CD200, CD273, CD274 and cytokines such as IL1β, IL-8, LIF and TGFβ2.
Collapse
|
31
|
Santos JM, Camões SP, Filipe E, Cipriano M, Barcia RN, Filipe M, Teixeira M, Simões S, Gaspar M, Mosqueira D, Nascimento DS, Pinto-do-Ó P, Cruz P, Cruz H, Castro M, Miranda JP. Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing. Stem Cell Res Ther 2015; 6:90. [PMID: 25956381 PMCID: PMC4448539 DOI: 10.1186/s13287-015-0082-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
Introduction The secretion of trophic factors by mesenchymal stromal cells has gained increased interest given the benefits it may bring to the treatment of a variety of traumatic injuries such as skin wounds. Herein, we report on a three-dimensional culture-based method to improve the paracrine activity of a specific population of umbilical cord tissue-derived mesenchymal stromal cells (UCX®) towards the application of conditioned medium for the treatment of cutaneous wounds. Methods A UCX® three-dimensional culture model was developed and characterized with respect to spheroid formation, cell phenotype and cell viability. The secretion by UCX® spheroids of extracellular matrix proteins and trophic factors involved in the wound-healing process was analysed. The skin regenerative potential of UCX® three-dimensional culture-derived conditioned medium (CM3D) was also assessed in vitro and in vivo against UCX® two-dimensional culture-derived conditioned medium (CM2D) using scratch and tubulogenesis assays and a rat wound splinting model, respectively. Results UCX® spheroids kept in our three-dimensional system remained viable and multipotent and secreted considerable amounts of vascular endothelial growth factor A, which was undetected in two-dimensional cultures, and higher amounts of matrix metalloproteinase-2, matrix metalloproteinase-9, hepatocyte growth factor, transforming growth factor β1, granulocyte-colony stimulating factor, fibroblast growth factor 2 and interleukin-6, when compared to CM2D. Furthermore, CM3D significantly enhanced elastin production and migration of keratinocytes and fibroblasts in vitro. In turn, tubulogenesis assays revealed increased capillary maturation in the presence of CM3D, as seen by a significant increase in capillary thickness and length when compared to CM2D, and increased branching points and capillary number when compared to basal medium. Finally, CM3D-treated wounds presented signs of faster and better resolution when compared to untreated and CM2D-treated wounds in vivo. Although CM2D proved to be beneficial, CM3D-treated wounds revealed a completely regenerated tissue by day 14 after excisions, with a more mature vascular system already showing glands and hair follicles. Conclusions This work unravels an important alternative to the use of cells in the final formulation of advanced therapy medicinal products by providing a proof of concept that a reproducible system for the production of UCX®-conditioned medium can be used to prime a secretome for eventual clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0082-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge M Santos
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Sérgio P Camões
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Elysse Filipe
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Madalena Cipriano
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Rita N Barcia
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Mariana Filipe
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Mariana Teixeira
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Sandra Simões
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Manuela Gaspar
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Diogo Mosqueira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal.
| | - Diana S Nascimento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal.
| | - Perpétua Pinto-do-Ó
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, N° 823, 4150-180, Porto, Portugal. .,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, N° 228, 4050-313, Porto, Portugal. .,Unit for Lymphopoiesis, Immunology Department, INSERM U668, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris, 75015, France.
| | - Pedro Cruz
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Helder Cruz
- ECBio - Investigação e Desenvolvimento em Biotecnologia S.A., Rua Henrique Paiva Couceiro, N° 27, 2700-451, Amadora, Portugal.
| | - Matilde Castro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Joana P Miranda
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
32
|
Mori Y, Ohshimo J, Shimazu T, He H, Takahashi A, Yamamoto Y, Tsunoda H, Tojo A, Nagamura-Inoue T. Improved Explant Method to Isolate Umbilical Cord-Derived Mesenchymal Stem Cells and Their Immunosuppressive Properties. Tissue Eng Part C Methods 2015; 21:367-72. [DOI: 10.1089/ten.tec.2014.0385] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun Ohshimo
- R & D Center, Tsubakimoto Chain Corporation, Saitama, Japan
| | - Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haiping He
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuki Yamamoto
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | - Arinobu Tojo
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Ebrahim NA, Leach L. Temporal studies into attachment, VE-cadherin perturbation, and paracellular migration of human umbilical mesenchymal stem cells across umbilical vein endothelial monolayers. Stem Cells Dev 2014; 24:426-36. [PMID: 25317631 DOI: 10.1089/scd.2014.0207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mesenchymal stem cells from Wharton's jelly of human umbilical cords (WJ-MSC) are a valuable alternate source of stem cells. Their role in situ and whether they can interact and cross intact endothelial monolayers requires elucidation. The aim of this study was to investigate the dynamic interactions between WJ-MSC and human umbilical vein endothelial cells (HUVEC), including attachment, transit times, extravasation pathway, and the effects of WJ-MSC on junctional vascular endothelial (VE)-cadherin. HUVEC were grown to near confluence in endothelial media and to full confluence in mixed media before the addition of PKH26-labelled WJ-MSC. Time lapse fluorescence microscopy showed stem cells undergoing membrane blebbing followed by amoeboid movement on HUVEC monolayers before rounding up and changing shape toward the spindle-shaped morphology during/after transmigration to subendothelial positions. Cells demonstrated a time lag of 60 min before paracellular extravasation, confirmed by confocal microscopy. Forty-six percent of attached cells crossed in the first 2 h. By 16 h, a majority of cells had transmigrated with >96% of cells crossing by 22 h. There were concomitant changes in endothelial junctional VE-cadherin with statistically significant increases in discontinuous staining at 2 h, return to control values at 16 h, even as from 22 h onward HUVEC displayed increased percentage of junctions with continuous staining and upregulation of protein. Our data suggests that WJ-MSC crosses the endothelial barrier through the paracellular pathway and can influence junctional organization of HUVEC with discreet perturbation of VE-cadherin preceding transmigration followed by upregulation once the adluminal side is reached. The latter may reflect a perivascular support function of WJ-MSC in the umbilical cord.
Collapse
Affiliation(s)
- Neven A Ebrahim
- Cardiovascular Research Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham , Nottingham, United Kingdom
| | | |
Collapse
|
34
|
Multiple injections of human umbilical cord-derived mesenchymal stromal cells through the tail vein improve microcirculation and the microenvironment in a rat model of radiation myelopathy. J Transl Med 2014; 12:246. [PMID: 25196350 PMCID: PMC4174271 DOI: 10.1186/s12967-014-0246-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 08/28/2014] [Indexed: 01/26/2023] Open
Abstract
Background At present, no effective clinical treatment is available for the late effects of radiation myelopathy. The aim of the present study was to assess the therapeutic effects of human umbilical cord-derived mesenchymal stromal cells (UC-MSCs) in a rat model of radiation myelopathy. Methods An irradiated cervical spinal cord rat model was generated. UC-MSCs were injected through the tail vein at 90, 97, 104 and 111 days post-irradiation. Behavioral tests were performed using the forelimb paralysis scoring system, and histological damage was examined using Nissl staining. The microcirculation in the spinal cord was assessed using von Willebrand factor (vWF) immunohistochemical analysis and laser-Doppler flowmetry. The microenvironment in the spinal cord was determined by measuring the pro-inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in the serum and the anti-inflammatory cytokines brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) in the spinal cord. Results Multiple injections of UC-MSCs through the tail veil decreased the forelimb paralysis, decreased spinal cord histological damage, increased the number of neurons in the anterior horn of the spinal cord, increased the endothelial cell density and the microvessel density in the white matter and gray matter of the spinal cord, increased the relative magnitude of spinal cord blood flow, down-regulated pro-inflammatory cytokine expression in the serum, and increased anti-inflammatory cytokine expression in the spinal cord. Conclusion Multiple injections of UC-MSCs via the tail vein in a rat model of radiation myelopathy significantly improved the microcirculation and microenvironment through therapeutic paracrine effects.
Collapse
|
35
|
Adami R, Scesa G, Bottai D. Stem cell transplantation in neurological diseases: improving effectiveness in animal models. Front Cell Dev Biol 2014; 2:17. [PMID: 25364724 PMCID: PMC4206985 DOI: 10.3389/fcell.2014.00017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/22/2014] [Indexed: 12/14/2022] Open
Abstract
Neurological diseases afflict a growing proportion of the human population. There are two reasons for this: first, the average age of the population (especially in the industrialized world) is increasing, and second, the diagnostic tools to detect these pathologies are now more sophisticated and can be used on a higher percentage of the population. In many cases, neurological disease has a pharmacological treatment which, as in the case of Alzheimer's disease, Parkinson's disease, Epilepsy, and Multiple Sclerosis can reduce the symptoms and slow down the course of the disease but cannot reverse its effects or heal the patient. In the last two decades the transplantation approach, by means of stem cells of different origin, has been suggested for the treatment of neurological diseases. The choice of slightly different animal models and the differences in methods of stem cell preparation make it difficult to compare the results of transplantation experiments. Moreover, the translation of these results into clinical trials with human subjects is difficult and has so far met with little success. This review seeks to discuss the reasons for these difficulties by considering the differences between human and animal cells (including isolation, handling and transplantation) and between the human disease model and the animal disease model.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Giuseppe Scesa
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| | - Daniele Bottai
- Department of Health Science, Faculty of Medicine, University of Milan Milan, Italy
| |
Collapse
|
36
|
Marto J, Baltazar D, Duarte A, Fernandes A, Gouveia L, Militão M, Salgado A, Simões S, Oliveira E, Ribeiro HM. Topical gels of etofenamate: in vitro and in vivo evaluation. Pharm Dev Technol 2014; 20:710-5. [PMID: 24798887 DOI: 10.3109/10837450.2014.915571] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Non-steroid anti-inflammatory drugs (NSAIDs), such as etofenamate, are among the most prescribed drugs used for their analgesic, anti-rheumatic, antipyretic and anti-inflammatory properties. Topical formulations have the main advantage of targeted delivery. However, drugs must overcome the skin due to its role as a physical and chemical barrier against the penetration of chemicals and microorganisms. This barrier must be altered to allow the permeation of drugs at a suitable rate to the desired site of activity. Permeation modulators can intercalate the skin outer layers causing structure disruption, opening an energetically favourable route for the drug to diffuse through. The aim of this work was the development of hydroalcoholic gels containing 5.0% (w/w) of etofenamate for topical administration with anti-inflammatory activity and enhanced drug delivery. The physical and chemical characterization, in vitro release and permeation studies and in vivo anti-inflammatory activity were assessed. The gel with 30% ethanol showed in vivo anti-inflammatory activity with suitable physical chemical and microbiologic characteristics. In vitro release and permeation studies revealed that the different amounts of ethanol used influenced the release profiles of etofenamate. Moreover, it was demonstrated that this formulation is an adequate vehicle for the etofenamate skin permeation.
Collapse
Affiliation(s)
- Joana Marto
- Pharmaceutics, Faculdade de Farmácia da Universidade de Lisboa, Instituto de Investigação do Medicamento (iMed.ULisboa) , Lisboa , Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gärtner A, Pereira T, Armada-da-Silva P, Amado S, Veloso A, Amorim I, Ribeiro J, Santos J, Bárcia R, Cruz P, Cruz H, Luís A, Santos J, Geuna S, Maurício A. Effects of umbilical cord tissue mesenchymal stem cells (UCX®) on rat sciatic nerve regeneration after neurotmesis injuries. J Stem Cells Regen Med 2014. [PMID: 25075157 PMCID: PMC4112274 DOI: 10.46582/jsrm.1001004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Peripheral nerves have the intrinsic capacity of self-regeneration after traumatic injury but the extent of the regeneration is often very poor. Increasing evidence demonstrates that mesenchymal stem/stromal cells (MSCs) may play an important role in tissue regeneration through the secretion of soluble trophic factors that enhance and assist in repair by paracrine activation of surrounding cells. In the present study, the therapeutic value of a population of umbilical cord tissue-derived MSCs, obtained by a proprietary method (UCX®), was evaluated on end-to-end rat sciatic nerve repair. Furthermore, in order to promote both, end-to-end nerve fiber contacts and MSC cell-cell interaction, as well as reduce the flush away effect of the cells after administration, a commercially available haemostatic sealant, Floseal®, was used as vehicle. Both, functional and morphologic recoveries were evaluated along the healing period using extensor postural thrust (EPT), withdrawal reflex latency (WRL), ankle kinematics analysis, and either histological analysis or stereology, in the hyper-acute, acute and chronic phases of healing. The histological analysis of the hyper-acute and acute phase studies revealed that in the group treated with UCX® alone the Wallerian degeneration was improved for the subsequent process of regeneration, the fiber organization was higher, and the extent of fibrosis was lower. The chronic phase experimental groups revealed that treatment with UCX® induced an increased number of regenerated fibers and thickening of the myelin sheet. Kinematics analysis showed that the ankle joint angle determined for untreated animals was significantly different from any of the treated groups at the instant of initial contact (IC). At opposite toe off (OT) and heel rise (HR), differences were found between untreated animals and the groups treated with either uCx® alone or UCX® administered with Floseal®. Overall, the UCX® application presented positive effects in functional and morphologic recovery, in both the acute and chronic phases of the regeneration process. Kinematics analysis has revealed positive synergistic effects brought by Floseal® as vehicle for MSCs.
Collapse
Affiliation(s)
- A Gärtner
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA) , Rua D. Manuel II, Apartado 55142, 4051-401, Porto, Portugal. ; These authors contributed equally for the results present in this research work
| | - T Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA) , Rua D. Manuel II, Apartado 55142, 4051-401, Porto, Portugal. ; These authors contributed equally for the results present in this research work
| | - Pas Armada-da-Silva
- Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (UL) , Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal. ; CIPER-FMH: Centro Interdisciplinar de Estudo de Performance Humana, Faculdade de Motricidade Humana (FMH) , Universidade de Lisboa (UL), Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal
| | - S Amado
- CIPER-FMH: Centro Interdisciplinar de Estudo de Performance Humana, Faculdade de Motricidade Humana (FMH) , Universidade de Lisboa (UL), Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal. ; UIS-IPL: Unidade de Investigação em Saúde da Escola Superior de Saúde de Leiria , Instituto Politécnico de Leiria, Portugal
| | - Ap Veloso
- Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (UL) , Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal. ; CIPER-FMH: Centro Interdisciplinar de Estudo de Performance Humana, Faculdade de Motricidade Humana (FMH) , Universidade de Lisboa (UL), Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal
| | - I Amorim
- Departamento de Patologia e de Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Instituto Português de Patologia e Imunologia Molecular da niversidade do Porto (IPATIMUP) , Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - J Ribeiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA) , Rua D. Manuel II, Apartado 55142, 4051-401, Porto, Portugal. ; UPVET, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Jd Santos
- CEMUC, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia , Universidade do Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Rn Bárcia
- ECBio - Research and Development in Biotechnology S.A. , Rua Henrique Paiva Couceiro, 27, 2700-451 Amadora, Portugal
| | - P Cruz
- ECBio - Research and Development in Biotechnology S.A. , Rua Henrique Paiva Couceiro, 27, 2700-451 Amadora, Portugal
| | - H Cruz
- ECBio - Research and Development in Biotechnology S.A. , Rua Henrique Paiva Couceiro, 27, 2700-451 Amadora, Portugal
| | - Al Luís
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Jm Santos
- ECBio - Research and Development in Biotechnology S.A. , Rua Henrique Paiva Couceiro, 27, 2700-451 Amadora, Portugal
| | - S Geuna
- Neuroscience Institute of the Cavalieri Ottolenghi Foundation , Turin, Italy. ; Department of Clinical and Biological Sciences , University of Turin, Italy
| | - Ac Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) , Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal. ; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA) , Rua D. Manuel II, Apartado 55142, 4051-401, Porto, Portugal
| |
Collapse
|
38
|
|
39
|
Reynolds G, Cooles FAH, Isaacs JD, Hilkens CMU. Emerging immunotherapies for rheumatoid arthritis. Hum Vaccin Immunother 2014; 10:822-37. [PMID: 24535556 DOI: 10.4161/hv.27910] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Novel treatments in development for rheumatoid arthritis target 3 broad areas: cytokines, cells, and signaling pathways. Therapies from each domain share common advantages (for example previously demonstrated efficacy, potential long-term immunomodulation, and oral administration respectively) that have stimulated research in each area but also common obstacles to their development. In this review recent progress in each area will be discussed alongside the factors that have impeded their path to clinical use.
Collapse
Affiliation(s)
- Gary Reynolds
- Institute of Cellular Medicine; Musculoskeletal Research Group; Newcastle University; Newcastle upon Tyne, Tyne and Wear UK
| | - Faye A H Cooles
- Institute of Cellular Medicine; Musculoskeletal Research Group; Newcastle University; Newcastle upon Tyne, Tyne and Wear UK
| | - John D Isaacs
- Institute of Cellular Medicine; Musculoskeletal Research Group; Newcastle University; Newcastle upon Tyne, Tyne & Wear UK
| | - Catharien M U Hilkens
- Institute of Cellular Medicine; Musculoskeletal Research Group; Newcastle University; Newcastle upon Tyne, Tyne & Wear UK
| |
Collapse
|
40
|
Towards an advanced therapy medicinal product based on mesenchymal stromal cells isolated from the umbilical cord tissue: quality and safety data. Stem Cell Res Ther 2014; 5:9. [PMID: 24438697 PMCID: PMC4055140 DOI: 10.1186/scrt398] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 12/20/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction Standardization of mesenchymal stromal cells (MSCs) manufacturing is urgently needed to enable translational activities and ultimately facilitate comparison of clinical trial results. In this work we describe the adaptation of a proprietary method for isolation of a specific umbilical cord tissue-derived population of MSCs, herein designated by its registered trademark as UCX®, towards the production of an advanced therapy medicinal product (ATMP). Methods The adaptation focused on different stages of production, from cell isolation steps to cell culturing and cryopreservation. The origin and quality of materials and reagents were considered and steps for avoiding microbiological and endotoxin contamination of the final cell product were implemented. Cell isolation efficiency, MSCs surface markers and genetic profiles, originating from the use of different medium supplements, were compared. The ATMP-compliant UCX® product was also cryopreserved avoiding the use of dimethyl sulfoxide, an added benefit for the use of these cells as an ATMP. Cells were analyzed for expansion capacity and longevity. The final cell product was further characterized by flow cytometry, differentiation potential, and tested for contaminants at various passages. Finally, genetic stability and immune properties were also analyzed. Results The isolation efficiency of UCX® was not affected by the introduction of clinical grade enzymes. Furthermore, isolation efficiencies and phenotype analyses revealed advantages in the use of human serum in cell culture as opposed to human platelet lysate. Initial decontamination of the tissue followed by the use of mycoplasma- and endotoxin-free materials and reagents in cell isolation and subsequent culture, enabled the removal of antibiotics during cell expansion. UCX®-ATMP maintained a significant expansion potential of 2.5 population doublings per week up to passage 15 (P15). They were also efficiently cryopreserved in a DMSO-free cryoprotectant medium with approximately 100% recovery and 98% viability post-thaw. Additionally, UCX®-ATMP were genetically stable upon expansion (up to P15) and maintained their immunomodulatory properties. Conclusions We have successfully adapted a method to consistently isolate, expand and cryopreserve a well-characterized population of human umbilical cord tissue-derived MSCs (UCX®), in order to obtain a cell product that is compliant with cell therapy. Here, we present quality and safety data that support the use of the UCX® as an ATMP, according to existing international guidelines.
Collapse
|
41
|
Santos Nascimento D, Mosqueira D, Sousa LM, Teixeira M, Filipe M, Resende TP, Araújo AF, Valente M, Almeida J, Martins JP, Santos JM, Bárcia RN, Cruz P, Cruz H, Pinto-do-Ó P. Human umbilical cord tissue-derived mesenchymal stromal cells attenuate remodeling after myocardial infarction by proangiogenic, antiapoptotic, and endogenous cell-activation mechanisms. Stem Cell Res Ther 2014; 5:5. [PMID: 24411922 PMCID: PMC4055157 DOI: 10.1186/scrt394] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/20/2013] [Indexed: 11/10/2022] Open
Abstract
Introduction Among the plethora of cells under investigation to restore a functional myocardium, mesenchymal stromal cells (MSCs) have been granted considerable interest. However, whereas the beneficial effects of bone marrow MSCs (BM-MSCs) in the context of the diseased heart are widely reported, data are still scarce on MSCs from the umbilical cord matrix (UCM-MSCs). Herein we report on the effect of UCM-MSC transplantation to the infarcted murine heart, seconded by the dissection of the molecular mechanisms at play. Methods Human umbilical cord tissue-derived MSCs (UCX®), obtained by using a proprietary technology developed by ECBio, were delivered via intramyocardial injection to C57BL/6 females subjected to permanent ligation of the left descending coronary artery. Moreover, medium produced by cultured UCX® preconditioned under normoxia (CM) or hypoxia (CMH) was collected for subsequent in vitro assays. Results Evaluation of the effects upon intramyocardial transplantation shows that UCX® preserved cardiac function and attenuated cardiac remodeling subsequent to myocardial infarction (MI). UCX® further led to increased capillary density and decreased apoptosis in the injured tissue. In vitro, UCX®-conditioned medium displayed (a) proangiogenic activity by promoting the formation of capillary-like structures by human umbilical vein endothelial cells (HUVECs), and (b) antiapoptotic activity in HL-1 cardiomyocytes subjected to hypoxia. Moreover, in adult murine cardiac Sca-1+ progenitor cells (CPCs), conditioned medium enhanced mitogenic activity while activating a gene program characteristic of cardiomyogenic differentiation. Conclusions UCX® preserve cardiac function after intramyocardial transplantation in a MI murine model. The cardioprotective effects of UCX® were attributed to paracrine mechanisms that appear to enhance angiogenesis, limit the extent of the apoptosis, augment proliferation, and activate a pool of resident CPCs. Overall, these results suggest that UCX® should be considered an alternative cell source when designing new therapeutic approaches to treat MI.
Collapse
|
42
|
Miranda JP, Filipe E, Fernandes AS, Almeida JM, Martins JP, De la Fuente A, Abal M, Barcia RN, Cruz P, Cruz H, Castro M, Santos JM. The Human Umbilical Cord Tissue-Derived MSC Population UCX(®) Promotes Early Motogenic Effects on Keratinocytes and Fibroblasts and G-CSF-Mediated Mobilization of BM-MSCs When Transplanted In Vivo. Cell Transplant 2013; 24:865-77. [PMID: 24480602 DOI: 10.3727/096368913x676231] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) play an important role in tissue regeneration mainly through the secretion of trophic factors that enhance the repair of damaged tissues. The main goal of this work was to study the paracrine mechanisms by which an umbilical cord tissue-derived MSC population (UCX(®)) promotes the migration capacity of human dermal fibroblasts and keratinocytes, which is highly relevant for skin regeneration. Furthermore, the differences between paracrine activities of MSCs from the umbilical cord tissue and the bone marrow (BM-MSCs) were also evaluated. In vitro scratch assays revealed that conditioned media (CM) obtained from both growing and stationary-phase UCX(®) cultures induced human dermal fibroblast (HDF) and keratinocyte (HaCaT) migration. These assays showed that the motogenic activity of UCX(®) CM to HaCaTs was significantly higher than to HDFs, in opposition to the effect seen with CM produced by BM-MSCs that preferentially induced HDF migration. Accordingly, a comparative quantification of key factors with vital importance in the consecutive stages of wound healing revealed very different secretome profiles between UCX(®) and BM-MSCs. The relatively higher UCX(®) expression of EGF, FGF-2, and KGF strongly supports early induction of keratinocyte migration and function, whereas the UCX(®)-specific expression of G-CSF suggested additional roles in mobilization of healing-related cells including CD34(-)/CD45(-) precursors (MSCs) known to be involved in tissue regeneration. Accordingly, in vitro chemotaxis assays and an in vivo transplantation model for chemoattraction confirmed that UCX(®) are chemotactic to CD34(-)/CD45(-) BM-MSCs via a cell-specific mobilization mechanism mediated by G-CSF. Overall, the results strongly suggest different paracrine activities between MSCs derived from different tissue sources, revealing the potential of UCX(®) to extend the regenerative capacity of the organism by complementing the role of endogenous BM-MSCs.
Collapse
Affiliation(s)
- Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|