1
|
Guillaumin S, Rossoni A, Zeugolis D. State-of the-art and future perspective in co-culture systems for tendon engineering. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100110. [PMID: 40130022 PMCID: PMC11932666 DOI: 10.1016/j.bbiosy.2025.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Tendon is a connective tissue that links bone to muscle, allowing for maintenance of skeleton posture, joint movement, energy storage and transmission of muscle force to bone. Tendon is a hypocellular and hypovascular tissue of poor self-regeneration capacity. Current surgical treatments are of limited success, frequently resulting in reinjury. Upcoming cell therapies are primarily based on tenocytes, a cell population of limited self-renewal capacity in vitro or mesenchymal stromal cells, a cell population prone to ectopic bone formation in vivo. Over the years mono- or multi- factorial cell culture technologies have failed to effectively maintain tenocyte phenotype in culture during expansion or to prime mesenchymal stromal cells towards tenogenic lineage prior to implantation. Upon these limitations the concept of co-culture was conceived. Here, we comprehensively review and discuss tenogenic differentiation of mesenchymal stromal cells through direct or indirect culture with tenocytes in an attempt to generate a tenocyte or a tendon-like cell population for regenerative medicine purposes.
Collapse
Affiliation(s)
- Salomé Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
2
|
Chen S, Liang B, Xu J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med 2024; 22:459. [PMID: 38750573 PMCID: PMC11094970 DOI: 10.1186/s12967-024-05294-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/11/2024] [Indexed: 05/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a heterogeneous cell population distributed throughout various tissues, demonstrating remarkable adaptability to microenvironmental cues and holding immense promise for disease treatment. However, the inherent diversity within MSCs often leads to variability in therapeutic outcomes, posing challenges for clinical applications. To address this heterogeneity, purification of MSC subpopulations through marker-based isolation has emerged as a promising approach to ensure consistent therapeutic efficacy. In this review, we discussed the reported markers of MSCs, encompassing those developed through candidate marker strategies and high-throughput approaches, with the aim of explore viable strategies for addressing the heterogeneity of MSCs and illuminate prospective research directions in this field.
Collapse
Affiliation(s)
- Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Bowei Liang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
3
|
Regenerative Potential of Granulation Tissue in Periodontitis: A Systematic Review and Meta-analysis. Stem Cells Int 2023; 2023:8789852. [PMID: 36926181 PMCID: PMC10014158 DOI: 10.1155/2023/8789852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/27/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Methods Electronic searches were conducted in five databases including CENTRAL, MEDLINE, EMBASE, Web of Science, and Dentistry & Oral Sciences Source using a combination of MeSH terms and keywords up to 21 June 2022. Human studies including patients aged over 18 years with all forms of periodontitis were included. Following the risk of bias assessment, both qualitative and quantitative analyses were performed. Results A total of twelve studies were included in qualitative analysis and six of them in quantitative analyses. The evidence suggested that cells derived from periodontitis granulation tissue have osteogenic, adipogenic, chondrogenic, neurogenic, and angiogenic differentiation abilities as well as immunoregulatory properties. In particular, CD44+, CD73+, CD90+, CD105+, and CD146+ cells were found widely in granulation tissue whilst the only meta-analysis confirmed that CD90+ cells were present in lower numbers within the granulation tissue when compared with healthy periodontal tissue (WMD = -23.43%, 95% CI -30.43 to -16.44, p < 0.00001). Conclusions This review provided further evidence that granulation tissue from patients with periodontitis can be a potential stem cell source for regenerative therapy.
Collapse
|
4
|
Sadeghi B, Ringdén O, Gustafsson B, Castegren M. Mesenchymal stromal cells as treatment for acute respiratory distress syndrome. Case Reports following hematopoietic cell transplantation and a review. Front Immunol 2022; 13:963445. [PMID: 36426365 PMCID: PMC9680556 DOI: 10.3389/fimmu.2022.963445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung disease. It may occur during the pancytopenia phase following allogeneic hematopoietic cell transplantation (HCT). ARDS is rare following HCT. Mesenchymal stromal cells (MSCs) have strong anti-inflammatory effect and first home to the lung following intravenous infusion. MSCs are safe to infuse and have almost no side effects. During the Covid-19 pandemic many patients died from ARDS. Subsequently MSCs were evaluated as a therapy for Covid-19 induced ARDS. We report three patients, who were treated with MSCs for ARDS following HCT. Two were treated with MSCs derived from the bone marrow (BM). The third patient was treated with MSCs obtained from the placenta, so-called decidua stromal cells (DSCs). In the first patient, the pulmonary infiltrates cleared after infusion of BM-MSCs, but he died from multiorgan failure. The second patient treated with BM-MSCs died of aspergillus infection. The patient treated with DSCs had a dramatic response and survived. He is alive after 7 years with a Karnofsky score of 100%. We also reviewed experimental and clinical studies using MSCs or DSCs for ARDS. Several positive reports are using MSCs for sepsis and ARDS in experimental animals. In man, two prospective randomized placebo-controlled studies used adipose and BM-MSCs, respectively. No difference in outcome was seen compared to placebo. Some pilot studies used MSCs for Covid-19 ARDS. Positive results were achieved using umbilical cord and DSCs however, optimal source of MSCs remains to be elucidated using randomized trials.
Collapse
Affiliation(s)
- Behnam Sadeghi
- Translational Cell Therapy Research (TCR), Division of Paediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Behnam Sadeghi,
| | - Olle Ringdén
- Translational Cell Therapy Research (TCR), Division of Paediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Britt Gustafsson
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Markus Castegren
- Center for Clinical Research, Sörmland, Uppsala University, Uppsala, Sweden
- Department of Anesthesiology and Intensive Care, CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Section of Infectious Diseases, Department of Medical Science, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Ivanovska A, Wang M, Arshaghi TE, Shaw G, Alves J, Byrne A, Butterworth S, Chandler R, Cuddy L, Dunne J, Guerin S, Harry R, McAlindan A, Mullins RA, Barry F. Manufacturing Mesenchymal Stromal Cells for the Treatment of Osteoarthritis in Canine Patients: Challenges and Recommendations. Front Vet Sci 2022; 9:897150. [PMID: 35754551 PMCID: PMC9230578 DOI: 10.3389/fvets.2022.897150] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/14/2022] [Indexed: 12/28/2022] Open
Abstract
The recent interest in advanced biologic therapies in veterinary medicine has opened up opportunities for new treatment modalities with considerable clinical potential. Studies with mesenchymal stromal cells (MSCs) from animal species have focused on in vitro characterization (mostly following protocols developed for human application), experimental testing in controlled studies and clinical use in veterinary patients. The ability of MSCs to interact with the inflammatory environment through immunomodulatory and paracrine mechanisms makes them a good candidate for treatment of inflammatory musculoskeletal conditions in canine species. Analysis of existing data shows promising results in the treatment of canine hip dysplasia, osteoarthritis and rupture of the cranial cruciate ligament in both sport and companion animals. Despite the absence of clear regulatory frameworks for veterinary advanced therapy medicinal products, there has been an increase in the number of commercial cell-based products that are available for clinical applications, and currently the commercial use of veterinary MSC products has outpaced basic research on characterization of the cell product. In the absence of quality standards for MSCs for use in canine patients, their safety, clinical efficacy and production standards are uncertain, leading to a risk of poor product consistency. To deliver high-quality MSC products for veterinary use in the future, there are critical issues that need to be addressed. By translating standards and strategies applied in human MSC manufacturing to products for veterinary use, in a collaborative effort between stem cell scientists and veterinary researchers and surgeons, we hope to facilitate the development of quality standards. We point out critical issues that need to be addressed, including a much higher level of attention to cell characterization, manufacturing standards and release criteria. We provide a set of recommendations that will contribute to the standardization of cell manufacturing methods and better quality assurance.
Collapse
Affiliation(s)
- Ana Ivanovska
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Mengyu Wang
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Tarlan Eslami Arshaghi
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Georgina Shaw
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | | | | | | | - Russell Chandler
- Orthopaedic Referral Service, Alphavet Veterinary Centre, Newport, United Kingdom
| | - Laura Cuddy
- Small Animal Surgery, Canine Sports Medicine and Rehabilitation, Veterinary Specialists Ireland, Summerhill, Ireland
| | - James Dunne
- Knocknacarra Veterinary Clinic, Ark Vets Galway, Galway, Ireland
| | - Shane Guerin
- Small Animal Surgery, Gilabbey Veterinary Hospital, Cork, Ireland
| | | | - Aidan McAlindan
- Northern Ireland Veterinary Specialists, Hillsborough, United Kingdom
| | - Ronan A Mullins
- Department of Small Animal Surgery, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
6
|
The cellular prion protein interacts with and promotes the activity of Na,K-ATPases. PLoS One 2021; 16:e0258682. [PMID: 34847154 PMCID: PMC8631662 DOI: 10.1371/journal.pone.0258682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/02/2021] [Indexed: 12/23/2022] Open
Abstract
The prion protein (PrP) is best known for its ability to cause fatal neurodegenerative diseases in humans and animals. Here, we revisited its molecular environment in the brain using a well-developed affinity-capture mass spectrometry workflow that offers robust relative quantitation. The analysis confirmed many previously reported interactions. It also pointed toward a profound enrichment of Na,K-ATPases (NKAs) in proximity to cellular PrP (PrPC). Follow-on work validated the interaction, demonstrated partial co-localization of the ATP1A1 and PrPC, and revealed that cells exposed to cardiac glycoside (CG) inhibitors of NKAs exhibit correlated changes to the steady-state levels of both proteins. Moreover, the presence of PrPC was observed to promote the ion uptake activity of NKAs in a human co-culture paradigm of differentiated neurons and glia cells, and in mouse neuroblastoma cells. Consistent with this finding, changes in the expression of 5’-nucleotidase that manifest in wild-type cells in response to CG exposure can also be observed in untreated PrPC-deficient cells. Finally, the endoproteolytic cleavage of the glial fibrillary acidic protein, a hallmark of late-stage prion disease, can also be induced by CGs, raising the prospect that a loss of NKA activity may contribute to the pathobiology of prion diseases.
Collapse
|
7
|
Hajiesmailpoor A, Emami P, Kondori BJ, Ghorbani M. Stem cell therapy as a recent advanced approach in male infertility. Tissue Cell 2021; 73:101634. [PMID: 34481231 DOI: 10.1016/j.tice.2021.101634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Infertility is one of the most common problems in the world that has negative effects on society and infertile people. Among the various causes of infertility, male infertility accounts for almost half of all infertility cases. Despite advances in medicine, current male infertility treatments such as assisted reproductive technology (ART) have not been successful in treating all types of male infertility. Recently, stem cells have been considered as therapeutic targets for many diseases, including infertility, due to their self-renewing and high differentiation. The purpose of this review is to discuss different types of male infertility and the effect of various stem cells against the treatment of male infertility.
Collapse
Affiliation(s)
- Ayshe Hajiesmailpoor
- Department of Emergency Medical Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Payam Emami
- Department of Emergency Medical Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Masoud Ghorbani
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Nair S, Rocha‐Ferreira E, Fleiss B, Nijboer CH, Gressens P, Mallard C, Hagberg H. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: Role of mitochondria, inflammation, and reactive oxygen species. J Neurochem 2021; 158:59-73. [PMID: 33314066 PMCID: PMC8359360 DOI: 10.1111/jnc.15267] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Preclinical studies have shown that mesenchymal stem cells have a positive effect in perinatal brain injury models. The mechanisms that cause these neurotherapeutic effects are not entirely intelligible. Mitochondrial damage, inflammation, and reactive oxygen species are considered to be critically involved in the development of injury. Mesenchymal stem cells have immunomodulatory action and exert mitoprotective effects which attenuate production of reactive oxygen species and promote restoration of tissue function and metabolism after perinatal insults. This review summarizes the present state, the underlying causes, challenges and possibilities for effective clinical translation of mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eridan Rocha‐Ferreira
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Bobbi Fleiss
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Université de Paris, NeuroDiderotParisFrance
| | - Cora H Nijboer
- Department for Developmental Origins of DiseaseUniversity Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht UniversityUtrechtNetherlands
| | | | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
9
|
Erivan R, Samper N, Villatte G, Boisgard S, Descamps S, Berger M. No Detectable Alteration of Inorganic Allogeneic Bone Matrix Colonizing Mesenchymal Cells: A Step Towards Personalized Bone Grafts. J Bone Metab 2021; 28:161-169. [PMID: 34130368 PMCID: PMC8206612 DOI: 10.11005/jbm.2021.28.2.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background During major bone substance loss, secured allogeneic bone matrix (ABM) is normally utilized for bone repair. Here, we propose a method to colonize ABM using autologous mesenchymal cells (MCs) to improve their integration. Moreover, in this study, the consequences of in vitro colonization on MCs have been evaluated. Methods After in vitro propagation of MCs, their proliferation kinetics on ABM pre-coated with gelatin, fibronectin, collagen IV and human serum (HS) was monitored, and they were compared with cells cultured without ABM for 8 weeks. The effect of ABM on cell phenotype was also assessed. Lastly, the ability of ABM-colonizing MCs to perform hematopoiesis, a function normally preserved in selected culture conditions, and their differentiation towards osteoblastic lineage were evaluated. Results MC and colony-forming unit-fibroblast proliferated 930- and 590-fold, respectively. The proliferation rate of the expanded MCs was higher, forming a 3-dimensional structure in all ABMs. Pre-coating with HS was the most efficient treatment of ABMs to increase the initial adherence of MCs, and it partly explains the reason for the higher propagation of MCs. Flow cytometry analyses revealed subtle alterations in ABM-colonizing cells; however, the ability of MCs to maintain long-term culture initiating cells proliferation and differentiate into osteoblastic lineage was preserved. Conclusions In this study, the in vitro biocompatibility of bone marrow (BM) MCs with ABMs, the role of HS in scaffold coating, and the possibility of initially using a small BM sample for this approach were demonstrated.
Collapse
Affiliation(s)
- Roger Erivan
- Université Clermont Auvergne, CHU Clermont-Ferrand, CNRS, SIGMA Clermont, ICCF, Clermont-Ferrand, France.,Department of Orthopedic and Trauma Surgery, Hôpital Gabriel Montpied, CHU de Clermont Ferrand, Clermont-Ferrand, France
| | - Nicolas Samper
- Université Clermont Auvergne, CHU Clermont-Ferrand, Clermont, Clermont- Ferrand, France
| | - Guillaume Villatte
- Université Clermont Auvergne, CHU Clermont-Ferrand, CNRS, SIGMA Clermont, ICCF, Clermont-Ferrand, France
| | - Stéphane Boisgard
- Université Clermont Auvergne, CHU Clermont-Ferrand, CNRS, SIGMA Clermont, ICCF, Clermont-Ferrand, France
| | - Stéphane Descamps
- Université Clermont Auvergne, CHU Clermont-Ferrand, CNRS, SIGMA Clermont, ICCF, Clermont-Ferrand, France
| | - Marc Berger
- Université Clermont Auvergne, CHU Clermont-Ferrand, GECOM, CRB Auvergne, Clermont-Ferrand, France
| |
Collapse
|
10
|
Galgaro BC, Beckenkamp LR, van den M Nunnenkamp M, Korb VG, Naasani LIS, Roszek K, Wink MR. The adenosinergic pathway in mesenchymal stem cell fate and functions. Med Res Rev 2021; 41:2316-2349. [PMID: 33645857 DOI: 10.1002/med.21796] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) play an important role in tissue homeostasis and damage repair through their ability to differentiate into cells of different tissues, trophic support, and immunomodulation. These properties made them attractive for clinical applications in regenerative medicine, immune disorders, and cell transplantation. However, despite multiple preclinical and clinical studies demonstrating beneficial effects of MSCs, their native identity and mechanisms of action remain inconclusive. Since its discovery, the CD73/ecto-5'-nucleotidase is known as a classic marker for MSCs, but its role goes far beyond a phenotypic characterization antigen. CD73 contributes to adenosine production, therefore, is an essential component of purinergic signaling, a pathway composed of different nucleotides and nucleosides, which concentrations are finely regulated by the ectoenzymes and receptors. Thus, purinergic signaling controls pathophysiological functions such as proliferation, migration, cell fate, and immune responses. Despite the remarkable progress already achieved in considering adenosinergic pathway as a therapeutic target in different pathologies, its role is not fully explored in the context of the therapeutic functions of MSCs. Therefore, in this review, we provide an overview of the role of CD73 and adenosine-mediated signaling in the functions ascribed to MSCs, such as homing and proliferation, cell differentiation, and immunomodulation. Additionally, we will discuss the pathophysiological role of MSCs, via CD73 and adenosine, in different diseases, as well as in tumor development and progression. A better understanding of the adenosinergic pathway in the regulation of MSCs functions will help to provide improved therapeutic strategies applicable in regenerative medicine.
Collapse
Affiliation(s)
- Bruna C Galgaro
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liziane R Beckenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Martha van den M Nunnenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitória G Korb
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Liliana I S Naasani
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| | - Márcia R Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
11
|
Cen YJ, You DB, Wang W, Feng Y. Preliminary studies of constructing a tissue-engineered lamellar corneal graft by culturing mesenchymal stem cells onto decellularized corneal matrix. Int J Ophthalmol 2021; 14:10-18. [PMID: 33469478 DOI: 10.18240/ijo.2021.01.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/28/2020] [Indexed: 12/28/2022] Open
Abstract
AIM To construct a competent corneal lamellar substitute in order to alleviate the shortage of human corneal donor. METHODS Rabbit mesenchymal stem cells (MSCs) were isolated from bone marrow and identified by flow cytometric, osteogenic and adipogenic induction. Xenogenic decellularized corneal matrix (XDCM) was generated from dog corneas. MSCs were seeded and cultured on XDCM to construct the tissue-engineered cornea. Post-transplantation biocompatibility of engineered corneal graft were tested by animal experiment. Rabbits were divided into two groups then underwent lamellar keratoplasty (LK) with different corneal grafts: 1) XDCM group (n=5): XDCM; 2) XDCM-MSCs groups (n=4): tissue-engineered cornea made up with XDCM and MSCs. The ocular surface recovery procedure was observed while corneal transparency, neovascularization and epithelium defection were measured and compared. In vivo on focal exam was performed 3mo postoperatively. RESULTS Rabbit MSCs were isolated and identified. Flow cytometry demonstrated isolated cells were CD90 positive and CD34, CD45 negative. Osteogenic and adipogenic induction verified their multipotent abilities. MSC-XDCM grafts were constructed and observed. In vivo transplantation showed the neovascularization in XDCM-MSC group was much less than that in XDCM group postoperatively. Post-transplant 3-month confocal test showed less nerve regeneration and bigger cell-absent area in XDCM-MSC group. CONCLUSION This study present a novel corneal tissue-engineered graft that could reduce post-operatively neovascularization and remain transparency, meanwhile shows that co-transplantation of MSCs may help increase corneal transplantation successful rate and enlarge the source range of corneal substitute to overcome cornea donor shortage.
Collapse
Affiliation(s)
- Yu-Jie Cen
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - De-Bo You
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wang
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
12
|
Jurek S, Sandhu MA, Trappe S, Bermúdez-Peña MC, Kolisek M, Sponder G, Aschenbach JR. Optimizing adipogenic transdifferentiation of bovine mesenchymal stem cells: a prominent role of ascorbic acid in FABP4 induction. Adipocyte 2020; 9:35-50. [PMID: 31996081 PMCID: PMC6999845 DOI: 10.1080/21623945.2020.1720480] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adipocyte differentiation of bovine adipose-derived stem cells (ASC) was induced by foetal bovine serum (FBS), biotin, pantothenic acid, insulin, rosiglitazone, dexamethasone and 3-isobutyl-1-methylxanthine, followed by incubation in different media to test the influence of ascorbic acid (AsA), bovine serum lipids (BSL), FBS, glucose and acetic acid on transdifferentiation into functional adipocytes. Moreover, different culture plate coatings (collagen-A, gelatin-A or poly-L-lysine) were tested. The differentiated ASC were subjected to Nile red staining, DAPI staining, immunocytochemistry and quantitative reverse transcription PCR (for NT5E, THY1, ENG, PDGFRα, FABP4, PPARγ, LPL, FAS, GLUT4). Nile red quantification showed a significant increase in the development of lipid droplets in treatments with AsA and BSL without FBS. The presence of BSL induced a prominent increase in FABP4 mRNA abundance and in FABP4 immunofluorescence signals in coincubation with AsA. The abundance of NT5E, ENG and THY1 mRNA decreased or tended to decrease in the absence of FBS, and ENG was additionally suppressed by AsA. DAPI fluorescence was higher in cells cultured in poly-L-lysine or gelatin-A coated wells. In additional experiments, the multi-lineage differentiation potential to osteoblasts was verified in medium containing ß-glycerophosphate, dexamethasone and 1,25-dihydroxyvitamin D3 using alizarin red staining. In conclusion, bovine ASC are capable of multi-lineage differentiation. Poly-L-lysine or gelatin-A coating, the absence of FBS, and the presence of BSL and AsA favour optimal transdifferentiation into adipocytes. AsA supports transdifferentiation via a unique role in FABP4 induction, but this is not linearly related to the primarily BSL-driven lipid accumulation. Abbreviations: AcA: acetic acid; AsA: ascorbic acid; ASC: adipose-derived stem cells; BSL: bovine serum lipids; DAPI: 4´,6-diamidino-2-phenylindole; DLK: delta like non-canonical notch ligand; DMEM: Dulbecco’s modified Eagle’s medium; DPBS: Dulbecco’s phosphate-buffered saline; ENG: endoglin; FABP: fatty acid binding protein; FAS: fatty acid synthase; GLUT4: glucose transporter type 4; IBMX: 3-isobutyl-1-methylxanthine; LPL: lipoprotein lipase; MSC: mesenchymal stem cells; α-MEM: α minimum essential medium; NT5E: ecto-5ʹ-nucleotidase; PDGFRα: platelet derived growth factor receptor α; PPARγ: peroxisome proliferator activated receptor γ; RPS19: ribosomal protein S19; SEM: standard error of the mean; THY1: Thy-1 cell surface antigen; TRT: treatment; TRT-Con: treatment negative control; YWHAZ: tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta
Collapse
Affiliation(s)
- Sandra Jurek
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - Mansur A. Sandhu
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
- Department of Veterinary Biomedical Sciences, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Susanne Trappe
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - M. Carmen Bermúdez-Peña
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
- Nursing Faculty, Autonomous University of Queretaro, Querétaro City, Mexico
| | - Martin Kolisek
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
- Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Gerhard Sponder
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
13
|
Coelho A, Alvites RD, Branquinho MV, Guerreiro SG, Maurício AC. Mesenchymal Stem Cells (MSCs) as a Potential Therapeutic Strategy in COVID-19 Patients: Literature Research. Front Cell Dev Biol 2020; 8:602647. [PMID: 33330498 PMCID: PMC7710935 DOI: 10.3389/fcell.2020.602647] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
In 2019, an outbreak of an unknown coronavirus - SARS-CoV-2 - responsible for COVID-19 disease, was first reported in China, and evolved into a pandemic of huge dimensions and raised serious concerns for global health. The number of critical cases continues to increase dramatically, while vaccines and specific treatments are not yet available. There are several strategies currently being studied for the treatment of adverse symptoms of COVID-19, that encompass Acute Lung Injury (ALI)/Acute Respiratory Distress Syndrome (ARDS), extensive pulmonary inflammation, cytokine storm, and pulmonary edema, due to virus-induced pneumonia. Mesenchymal stem cells (MSCs) are at the origin of new revolutionary treatments, which may come to be applied in such as Regenerative Medicine, Immunotherapy, Tissue Engineering, and Cell and Molecular Biology due to immunomodulation and anti-inflammatory activity. MSCs have already been studied with positive outcomes for other lung pathologies, thus representing and being identified as an important opportunity for the treatment of COVID-19. It has recently been shown that these cells allow hopeful and effective therapies for serious or critical COVID-19, minimizing its adverse symptoms. In this study we will analyze the MSCs, their origin, differentiation, and therapeutic potential, making a bridge with the COVID-19 disease and its characteristics, as a potential therapeutic strategy but also reporting recent studies where these cell-based therapies were used for the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- André Coelho
- Biotecnologia Medicinal, Escola Superior de Saúde do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Rui Damásio Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
| | - Mariana Vieira Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
| | - Susana G. Guerreiro
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
| |
Collapse
|
14
|
Gomez-Salazar M, Gonzalez-Galofre ZN, Casamitjana J, Crisan M, James AW, Péault B. Five Decades Later, Are Mesenchymal Stem Cells Still Relevant? Front Bioeng Biotechnol 2020; 8:148. [PMID: 32185170 PMCID: PMC7058632 DOI: 10.3389/fbioe.2020.00148] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mario Gomez-Salazar
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zaniah N Gonzalez-Galofre
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Joan Casamitjana
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mihaela Crisan
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Bruno Péault
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Improved Isolation of Mesenchymal Stem Cells Based on Interactions between N-Acetylglucosamine-Bearing Polymers and Cell-Surface Vimentin. Stem Cells Int 2019; 2019:4341286. [PMID: 31814834 PMCID: PMC6878802 DOI: 10.1155/2019/4341286] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) in bone marrow and adipose tissues are expected to be effective tools for regenerative medicine to treat various diseases. To obtain MSCs that possess both high differentiation and tissue regenerative potential, it is necessary to establish an isolation system that does not require long-term culture. It has previously been reported that the cytoskeletal protein vimentin, expressed on the surfaces of multiple cell types, possesses N-acetylglucosamine- (GlcNAc-) binding activity. Therefore, we tried to exploit this interaction to efficiently isolate MSCs from rat bone marrow cells using GlcNAc-bearing polymer-coated dishes. Cells isolated by this method were identified as MSCs because they were CD34-, CD45-, and CD11b/c-negative and CD90-, CD29-, CD44-, CD54-, CD73-, and CD105-positive. Osteoblast, adipocyte, and chondrocyte differentiation was observed in these cells. In total, yields of rat MSCs were threefold to fourfold higher using GlcNAc-bearing polymer-coated dishes than yields using conventional tissue-culture dishes. Interestingly, MSCs isolated with GlcNAc-bearing polymer-coated dishes strongly expressed CD106, whereas those isolated with conventional tissue-culture dishes had low CD106 expression. Moreover, senescence-associated β-galactosidase activity in MSCs from GlcNAc-bearing polymer-coated dishes was lower than that in MSCs from tissue-culture dishes. These results establish an improved isolation method for high-quality MSCs.
Collapse
|
16
|
Barry F. MSC Therapy for Osteoarthritis: An Unfinished Story. J Orthop Res 2019; 37:1229-1235. [PMID: 31081558 DOI: 10.1002/jor.24343] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 04/26/2019] [Indexed: 02/04/2023]
Abstract
Mesenchymal stromal cells (MSCs) have firmly occupied the attention of orthopedic clinicians and scientists for most of the last 25 years. Hundreds of laboratories worldwide have carried out research aimed at unraveling the biological characteristics of these cells and probing the manner in which they potentially contribute to cartilage and bone repair. Clinical trials registries indicate that they are also being tested in patient studies for a wide range of conditions such as osteoarthritis, rheumatoid arthritis, fracture repair, regeneration of articular cartilage, tendon repair, and for treatment of degenerative disc disease. Despite these efforts, the effectiveness of MSCs as a treatment modality for these conditions is still uncertain and market authorizations have been limited. In addition, critical and clear phenotypic parameters for defining MSCs are uncertain and a coherent biological framework surrounding the therapeutic mechanism of action is not yet available. Added to this, cell manufacturing protocols are complex and costly and present substantial challenges in terms of regulatory oversight and standardization. Despite these obstacles, MSCs still remain at the forefront of efforts in Regenerative Medicine, based on a conviction that this technology can provide an effective treatment paradigm for major diseases where there is still an unmet need. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1229-1235, 2019.
Collapse
Affiliation(s)
- Frank Barry
- Regenerative Medicine Institute, National University of Ireland Galway, Arthritis Programme, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Robb KP, Fitzgerald JC, Barry F, Viswanathan S. Mesenchymal stromal cell therapy: progress in manufacturing and assessments of potency. Cytotherapy 2018; 21:289-306. [PMID: 30528726 DOI: 10.1016/j.jcyt.2018.10.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cell (MSC) therapies have been pursued for a broad spectrum of indications but mixed reports on clinical efficacy have given rise to some degree of skepticism regarding the effectiveness of this approach. However, recent reports of successful clinical outcomes and regulatory approvals for graft-versus-host disease, Crohn's disease and critical limb ischemia have prompted a shift in this perspective. With hundreds of clinical trials involving MSCs currently underway and an increasing demand for large-scale manufacturing protocols, there is a critical need to develop standards that can be applied to processing methods and to establish consensus assays for both MSC processing control and MSC product release. Reference materials and validated, uniformly applied tests for quality control of MSC products are needed. Here, we review recent developments in MSC manufacturing technologies, release testing and potency assays. We conclude that, although MSCs hold considerable promise clinically, economies of scale have yet to be achieved although numerous bioreactor technologies for scalable production of MSCs exist. Additionally, rigorous disease-specific product testing and comprehensive understanding of mechanisms of action, which are linked to relevant process and product release potency assays, will be required to ensure that these therapies continue to be successful.
Collapse
Affiliation(s)
- Kevin P Robb
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Joan C Fitzgerald
- Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Frank Barry
- The Arthritis Program, University Health Network, Toronto, Canada;; Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Sowmya Viswanathan
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Cell Therapy Program, University Health Network, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
18
|
Huynh NPT, Zhang B, Guilak F. High-depth transcriptomic profiling reveals the temporal gene signature of human mesenchymal stem cells during chondrogenesis. FASEB J 2018; 33:358-372. [PMID: 29985644 DOI: 10.1096/fj.201800534r] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) provide an attractive cell source for cartilage repair and cell therapy; however, the underlying molecular pathways that drive chondrogenesis of these populations of adult stem cells remain poorly understood. We generated a rich data set of high-throughput RNA sequencing of human MSCs throughout chondrogenesis at 6 different time points. Our data consisted of 18 libraries with 3 individual donors as biologic replicates, with each library possessing a sequencing depth of 100 million reads. Computational analyses with differential gene expression, gene ontology, and weighted gene correlation network analysis identified dynamic changes in multiple biologic pathways and, most importantly, a chondrogenic gene subset, whose functional characterization promises to further harness the potential of MSCs for cartilage tissue engineering. Furthermore, we created a graphic user interface encyclopedia built with the goal of producing an open resource of transcriptomic regulation for additional data mining and pathway analysis of the process of MSC chondrogenesis.-Huynh, N. P. T., Zhang, B., Guilak, F. High-depth transcriptomic profiling reveals the temporal gene signature of human mesenchymal stem cells during chondrogenesis.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and.,Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and
| |
Collapse
|
19
|
Sorrell JM, Somoza RA, Caplan AI. Human mesenchymal stem cells induced to differentiate as chondrocytes follow a biphasic pattern of extracellular matrix production. J Orthop Res 2018; 36:1757-1766. [PMID: 29194731 PMCID: PMC5976510 DOI: 10.1002/jor.23820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
Regenerative medicine and tissue engineering studies are actively developing novel means to repair adult articular cartilage defects using biological approaches. One such approach is the harnessing of adult human therapeutic cells such as those referred to as mesenchymal stem cells. Upon exposure to chondrogenic signals, these cells differentiate and initiate the production of a complex and voluminous cartilaginous matrix that is crucial to both the structure and function of cartilage. Furthermore, this complexity requires the time-sensitive activation of a large number of genes to produce the components of this matrix. The current study analyzed the kinetics of matrix production in an aggregate culture model where adult human mesenchymal stem cells were induced to differentiate as chondrocytes. The results indicate the existence of a biphasic mode of differentiation and maturation during which matrix genes and molecules are differentially activated and secreted. These results have important implications for developing novel approaches for the creation of tissue engineered articular cartilage. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1757-1766, 2018.
Collapse
Affiliation(s)
- J. Michael Sorrell
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Rodrigo A. Somoza
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Arnold I. Caplan
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| |
Collapse
|
20
|
Pourmoghadam Z, Aghebati‐Maleki L, Motalebnezhad M, Yousefi B, Yousefi M. Current approaches for the treatment of male infertility with stem cell therapy. J Cell Physiol 2018; 233:6455-6469. [DOI: 10.1002/jcp.26577] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/27/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Zahra Pourmoghadam
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Leili Aghebati‐Maleki
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | | | - Bahman Yousefi
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
21
|
Hoffmann A, Floerkemeier T, Melzer C, Hass R. Comparison of in vitro-cultivation of human mesenchymal stroma/stem cells derived from bone marrow and umbilical cord. J Tissue Eng Regen Med 2017; 11:2565-2581. [PMID: 27125777 DOI: 10.1002/term.2153] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 12/15/2015] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Cell-mediated therapy is currently considered as a novel approach for many human diseases. Potential uses range from topic applications with the regeneration of confined tissue areas to systemic applications. Stem cells including mesenchymal stroma/stem cells (MSCs) represent a highly attractive option. Their potential to cure or alleviate human diseases is investigated in a number of clinical trials. A wide variety of methods has been established in the past years for isolation, cultivation and characterization of human MSCs as expansion is presently deemed a prerequisite for clinical application with high numbers of cells carrying reproducible properties. MSCs have been retrieved from various tissues and used in a multitude of settings whereby numerous experimental protocols are available for expansion of MSCs in vitro. Accordingly, different isolation, culture and upscaling techniques contribute to the heterogeneity of MSC characteristics and the, sometimes, controversial results. Therefore, this review discusses and summarizes certain experimental conditions for MSC in vitro culture focusing on adult bone marrow-derived and neonatal umbilical cord-derived MSCs in order to enhance our understanding for MSC tissue sources and to stratify different procedures. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andrea Hoffmann
- Department of Orthopaedic Surgery, OE 8893, Hannover Medical School, Hannover, Germany
| | - Thilo Floerkemeier
- Department of Orthopaedic Surgery (Annastift), OE 6270, Hannover Medical School, Hannover, Germany
| | - Catharina Melzer
- Biochemistry and Tumour Biology Laboratory, Department of Obstetrics and Gynecology, OE 6411, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumour Biology Laboratory, Department of Obstetrics and Gynecology, OE 6411, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Payushina OV, Butorina NN, Sheveleva ON, Bukhinnik SS, Berezina AA, Ramazanova SG, Domaratskaya EI. Comparative study of bone marrow mesenchymal stromal cells at different stages of ontogeny. Russ J Dev Biol 2017. [DOI: 10.1134/s1062360417040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Kovac M, Vasicek J, Kulikova B, Bauer M, Curlej J, Balazi A, Chrenek P. Different RNA and protein expression of surface markers in rabbit amniotic fluid-derived mesenchymal stem cells. Biotechnol Prog 2017; 33:1601-1613. [DOI: 10.1002/btpr.2519] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/25/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Michal Kovac
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture; Nitra Slovak Republic
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
| | - Jaromir Vasicek
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
- Research Centre AgroBioTech, Slovak University of Agriculture; Nitra Slovak Republic
| | - Barbora Kulikova
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
| | - Miroslav Bauer
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
- Faculty of Natural Sciences; Constantine the Philosopher University; Nitra Slovak republic
| | - Jozef Curlej
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture; Nitra Slovak Republic
| | - Andrej Balazi
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
| | - Peter Chrenek
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture; Nitra Slovak Republic
- Research Inst. for Animal Production, National Agricultural and Food Centre; Lužianky Slovak Republic
| |
Collapse
|
24
|
Cell Connections by Tunneling Nanotubes: Effects of Mitochondrial Trafficking on Target Cell Metabolism, Homeostasis, and Response to Therapy. Stem Cells Int 2017; 2017:6917941. [PMID: 28659978 PMCID: PMC5474251 DOI: 10.1155/2017/6917941] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/21/2017] [Accepted: 03/30/2017] [Indexed: 01/16/2023] Open
Abstract
Intercellular communications play a major role in tissue homeostasis and responses to external cues. Novel structures for this communication have recently been described. These tunneling nanotubes (TNTs) consist of thin-extended membrane protrusions that connect cells together. TNTs allow the cell-to-cell transfer of various cellular components, including proteins, RNAs, viruses, and organelles, such as mitochondria. Mesenchymal stem cells (MSCs) are both naturally present and recruited to many different tissues where their interaction with resident cells via secreted factors has been largely documented. Their immunosuppressive and repairing capacities constitute the basis for many current clinical trials. MSCs recruited to the tumor microenvironment also play an important role in tumor progression and resistance to therapy. MSCs are now the focus of intense scrutiny due to their capacity to form TNTs and transfer mitochondria to target cells, either in normal physiological or in pathological conditions, leading to changes in cell energy metabolism and functions, as described in this review.
Collapse
|
25
|
Holton J, Imam MA, Snow M. Bone Marrow Aspirate in the Treatment of Chondral Injuries. Front Surg 2016; 3:33. [PMID: 27379241 PMCID: PMC4909728 DOI: 10.3389/fsurg.2016.00033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023] Open
Abstract
The ability of mesenchymal stem cells (MSCs) to transdifferentiate into a desired cell lineage has captured the imagination of scientists and clinicians alike. The limited ability for chondrocytes to regenerate in chondral injuries has raised the concept of using MSCs to help regenerate and repair damaged tissue. The expansion of cells in a laboratory setting to be delivered back to the patient is too costly for clinical use in the present tough economic climate. This process is slow with due to the complexity of trying to imitate the natural environment and biological stimulation of chondral cell replication and proliferation. Bone marrow aspirate concentrate (BMAC) has the potential to provide an easily accessible and readily available source of MSCs with key growth factors that can be used in treating chondral injuries. This review summarizes the underlying basic science of MSCs and the therapeutic potential of BMAC.
Collapse
Affiliation(s)
- James Holton
- Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK
| | - Mohamed A. Imam
- Royal Orthopaedic Hospital, Birmingham, UK
- Suez Canal University, Ismailia, Egypt
| | | |
Collapse
|
26
|
Mesenchymal stem cells: Immunomodulatory capability and clinical potential in immune diseases. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2014.12.001] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
Abstract
Stem cells possess the extraordinary capacity of self-renewal and differentiation to various cell types, thus to form original tissues and organs. Stem cell heterogeneity including genetic and nongenetic mechanisms refers to biological differences amongst normal and stem cells originated within the same tissue. Cell differentiation hierarchy and stochasticity in gene expression and signaling pathways may result in phenotypic differences of stem cells. The maintenance of stemness and activation of differentiation potential are fundamentally orchestrated by microenvironmental stem cell niche-related cellular and humoral signals.
Collapse
Affiliation(s)
- Györgyi Műzes
- 2nd Department of Medicine, Immunology Division, Semmelweis University, Szentkirályi u. 46., Budapest, 1088, Hungary.
| | - Ferenc Sipos
- 2nd Department of Medicine, Immunology Division, Semmelweis University, Szentkirályi u. 46., Budapest, 1088, Hungary
| |
Collapse
|
28
|
Caplan AI, Sorrell JM. The MSC curtain that stops the immune system. Immunol Lett 2015; 168:136-9. [DOI: 10.1016/j.imlet.2015.06.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/04/2015] [Indexed: 01/08/2023]
|
29
|
Jeon YJ, Kim J, Cho JH, Chung HM, Chae JI. Comparative Analysis of Human Mesenchymal Stem Cells Derived From Bone Marrow, Placenta, and Adipose Tissue as Sources of Cell Therapy. J Cell Biochem 2015; 117:1112-25. [PMID: 26448537 DOI: 10.1002/jcb.25395] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022]
Abstract
Various source-derived mesenchymal stem cells (MSCs) with multipotent capabilities were considered for cell therapeutics of incurable diseases. The applicability of MSCs depends on the cellular source and on their different in vivo functions, despite having similar phenotypic and cytological characteristics. We characterized MSCs from different sources, including human bone marrow (BM), placenta (PL), and adipose tissue (AT), in terms of the phenotype, surface antigen expression, differentiation ability, proteome reference map, and blood flow recovery in a hindlimb ischemic disease model. The MSCs exhibit different differentiation potentials depending on the cellular source despite having similar phenotypic and surface antigen expression. We identified approximately 90 differentially regulated proteins. Most up- or down-regulated proteins show cytoskeletal or oxidative stress, peroxiredoxin, and apoptosis roles according to their functional involvement. In addition, the PL-MSCs retained a higher therapeutic efficacy than the BM- and AT-MSCs in the hindlimb ischemic disease model. In summary, we examined differentially expressed key regulatory factors for MSCs that were obtained from several cellular sources and demonstrated their differentially expressed proteome profiles. Our results indicate that primitive PL-MSCs have biological advantages relative to those from other sources, making PL-MSCs a useful model for clinical applications of cell therapy.
Collapse
Affiliation(s)
- Young-Joo Jeon
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju, 561-756, Republic of Korea
| | - Jumi Kim
- Samsung Advanced Institute of Technology, Well Aging Research Center, Suwon, Republic of Korea
| | - Jin Hyoung Cho
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju, 561-756, Republic of Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju, 561-756, Republic of Korea
| |
Collapse
|
30
|
Sun Q, Li F, Li H, Chen RH, Gu YZ, Chen Y, Liang HS, You XR, Ding SS, Gao L, Wang YL, Qin MD, Zhang XG. Amniotic fluid stem cells provide considerable advantages in epidermal regeneration: B7H4 creates a moderate inflammation microenvironment to promote wound repair. Sci Rep 2015; 5:11560. [PMID: 26101181 PMCID: PMC4477371 DOI: 10.1038/srep11560] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/18/2015] [Indexed: 11/09/2022] Open
Abstract
The current treatments for severe skin injury all involve skin grafting. However, there is a worldwide shortage of donor skin tissue. In this study, we examined the advantages of using human amniotic fluid stem (hAFS) cells in skin wound healing. In vitro, hAFS cells differentiate into keratinocytes (termed hAFS-K). Like keratinocytes, hAFS-K cells express the markers K5, K14, K10 and involucrin; display typical cellular structure, including a tonofibril-rich cytoplasm; and construct a completely pluristratified epithelium in 3D culture. In vivo, in a mouse excisional wound model, GFP-positive hAFS cells participate in wound repair. Co-localization of GFP/K14 and GFP/K10 in the repaired epidermis demonstrated that hAFS cells can differentiate into keratinocytes. Real-time PCR results confirmed that hAFS cells can initiate and promote early-stage repair of skin damage. During wound repair, hAFS cells did not directly secrete repair-related factors, such as bFGF, VEGF, CXCL12, TGF-β1 and KGF, and provided a moderate inflammation reaction with lower expression of IL-1β, IL-6, TNF-α, Cox2 and Mac3. In hAFS cells, the negative co-stimulatory molecule B7H4 regulates low immunogenicity, which can provide a modest inflammatory reaction microenvironment for wound repair. Furthermore, with their uniquely high proliferation rate, hAFS cells offer a promising alternative for epidermal regeneration.
Collapse
Affiliation(s)
- Qing Sun
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Fang Li
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Hong Li
- Center for Reproduction and Genetics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province,P.R. China
| | - Rui-Hua Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Yan-Zheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ying Chen
- Center for Reproduction and Genetics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province,P.R. China
| | - Han-Si Liang
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Xin-Ran You
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Si-Si Ding
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ling Gao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Yun-Liang Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ming-De Qin
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Xue-Guang Zhang
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| |
Collapse
|
31
|
Açil Y, Yang F, Gulses A, Ayna M, Wiltfang J, Gierloff M. Isolation, characterization and investigation of differentiation potential of human periodontal ligament cells and dental follicle progenitor cells and their response to BMP-7 in vitro. Odontology 2015; 104:123-35. [PMID: 25757659 DOI: 10.1007/s10266-015-0198-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/18/2015] [Indexed: 12/18/2022]
Abstract
The aim of this study was to assess the factors, mechanisms and the differences between periodontal ligament (PDL) cells and denta l follicle (DF) progenitor cells towards the osteoblastic/cementoblastic differentiation and to investigate the effects of BMP-7 on developmental (DF) and mature tissue-derived (PDL) cells, respectively. Primary cell culture of PDL cells and DF progenitor cells was performed. Osteogenic differentiation was evaluated using von Kossa, Alizarin Red S and immuno-histo-chemistry staining of osteocalcin. Gene expression pattern was evaluated via real-time PCR. A series of CD surface marks were tested using flow cytometry and fluorescence-activated cell-sorting analysis was performed. Real-time RT-PCR demonstrated similar gene expression pattern of PDL cells and DF progenitor cells: the expression of OPN and OCN significantly was elevated when incubated with osteogenic components, Runx2 was unaffected, and Osteorix was hardly expressed whether in basic medium or induction medium. In addition, BMP-7 induced osteoblast/cementoblast differentiation of PDLSCs and DF progenitor cells in a dose- and time-dependent manner, as reflected by enhanced Runx2 and (OCN) mRNA transcript expression. BMP-7 triggers PDL cells and DF progenitor cells to differentiate towards an osteoblast/cementoblast phenotype.
Collapse
Affiliation(s)
- Yahya Açil
- Department of Oral and Maxillofacial Surgery, Christian Albrechts University, Kiel, Germany
| | - Fan Yang
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, China
| | - Aydin Gulses
- Centre for Oral Health and Dental Management, Mevki Military Hospital, 06130, Diskapi Altindag, Ankara, Turkey.
| | | | - Jörg Wiltfang
- Department of Oral and Maxillofacial Surgery, Christian Albrechts University, Kiel, Germany
| | - Matthias Gierloff
- Department of Oral and Maxillofacial Surgery, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
32
|
Tissue non-specific alkaline phosphatase production by human dental pulp stromal cells is enhanced by high density cell culture. Cell Tissue Res 2015; 361:529-40. [PMID: 25636587 PMCID: PMC4529449 DOI: 10.1007/s00441-014-2106-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
The cell surface hydrolase tissue non-specific alkaline phosphatase (TNAP) (also known as MSCA-1) is used to identify a sub-population of bone marrow stromal cells (BMSCs) with high mineralising potential and is found on subsets of cells within the dental pulp. We aim to determine whether TNAP is co-expressed by human dental pulp stromal cells (hDPSCs) alongside a range of BMSC markers, whether this is an active form of the enzyme and the effects of culture duration and cell density on its expression. Cells from primary dental pulp and culture expanded hDPSCs expressed TNAP. Subsequent analyses revealed persistent TNAP expression and co-expression with BMSC markers such as CD73 and CD90. Flow cytometry and biochemical assays showed that increased culture durations and cell densities enhanced TNAP expression by hDPSCs. Arresting the hDPSC cell cycle also increased TNAP expression. These data confirm that TNAP is co-expressed by hDPSCs together with other BMSC markers and show that cell density affects TNAP expression levels. We conclude that TNAP is a potentially useful marker for hDPSC selection especially for uses in mineralised tissue regenerative therapies.
Collapse
|
33
|
Hashemian SJ, Kouhnavard M, Nasli-Esfahani E. Mesenchymal Stem Cells: Rising Concerns over Their Application in Treatment of Type One Diabetes Mellitus. J Diabetes Res 2015; 2015:675103. [PMID: 26576437 PMCID: PMC4630398 DOI: 10.1155/2015/675103] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 01/17/2015] [Accepted: 01/18/2015] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disorder that leads to beta cell destruction and lowered insulin production. In recent years, stem cell therapies have opened up new horizons to treatment of diabetes mellitus. Among all kinds of stem cells, mesenchymal stem cells (MSCs) have been shown to be an interesting therapeutic option based on their immunomodulatory properties and differentiation potentials confirmed in various experimental and clinical trial studies. In this review, we discuss MSCs differential potentials in differentiation into insulin-producing cells (IPCs) from various sources and also have an overview on currently understood mechanisms through which MSCs exhibit their immunomodulatory effects. Other important issues that are provided in this review, due to their importance in the field of cell therapy, are genetic manipulations (as a new biotechnological method), routes of transplantation, combination of MSCs with other cell types, frequency of transplantation, and special considerations regarding diabetic patients' autologous MSCs transplantation. At the end, utilization of biomaterials either as encapsulation tools or as scaffolds to prevent immune rejection, preparation of tridimensional vascularized microenvironment, and completed or ongoing clinical trials using MSCs are discussed. Despite all unresolved concerns about clinical applications of MSCs, this group of stem cells still remains a promising therapeutic modality for treatment of diabetes.
Collapse
Affiliation(s)
- Seyed Jafar Hashemian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Seyed Jafar Hashemian:
| | - Marjan Kouhnavard
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Choi JK, Hwang HI, Jang YJ. The efficiency of the in vitro osteo/dentinogenic differentiation of human dental pulp cells, periodontal ligament cells and gingival fibroblasts. Int J Mol Med 2014; 35:161-8. [PMID: 25355399 DOI: 10.3892/ijmm.2014.1986] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 10/20/2014] [Indexed: 11/06/2022] Open
Abstract
Although the primary cell cultures from dental pulp and other oral tissue are frequently used to study osteogenic potential and stem cell responses, few systematic and comparative studies on stemness for the dentinogenic differentiation of these cells have been conducted. In the present study, to investigate the stemness of oral primary cells during extended culture, human adult dental pulp cells (hDPCs), periodontal ligament stem cells (hPDLSCs) and gingival fibroblasts (hGFs) were obtained and cultured from pulp tissue, periodontal ligaments, and marginal and attached gingival tissue of extracted third molars, respectively. As shown by fluorescence-activated cell sorting analysis and immunophenotyping, the mesenchymal stem cell markers, CD44, CD73, CD90, CD146 and CD166, were highly expressed in early passage hDPCs, hPDLSCs and hGFs. However, when the cells were treated with osteogenic additives, mineralization markedly increased in the hDPCs and hPDLSCs, but not in the hGFs. Moreover, the expression of dentinogenic markers, such as dentin sialophosphoprotein and dentin matrix protein-1, appeared to decrease during extended culture past passage number 8 of the hDPCs and hPDLSCs. These data suggest that hDPCs and hPDLSCs may have differentiation potential during the early passages, and that their progenitor potential is diminished during extended culture. The hGFs did not show differentiation capability during culture, even though they contained general mesenchymal stem cell surface proteins. The transcriptional expression of dentinogenic markers in hDPCs was not affected by co-culture with hPDLSCs and/or hGFs.
Collapse
Affiliation(s)
- Jung-Kwon Choi
- Laboratory of Cell Cycle and Signal Transduction, Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan-si, Chungnam 330-714, Republic of Korea
| | - Hyo-In Hwang
- Laboratory of Cell Cycle and Signal Transduction, Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan-si, Chungnam 330-714, Republic of Korea
| | - Young-Joo Jang
- Laboratory of Cell Cycle and Signal Transduction, Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan-si, Chungnam 330-714, Republic of Korea
| |
Collapse
|
35
|
Rahimzadeh A, Mirakabad FST, Movassaghpour A, Shamsasenjan K, Kariminekoo S, Talebi M, Shekari A, Zeighamian V, Ghalhar MG, Akbarzadeh A. Biotechnological and biomedical applications of mesenchymal stem cells as a therapeutic system. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 44:559-70. [PMID: 25340260 DOI: 10.3109/21691401.2014.968823] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic, multipotent progenitor cells which reside in bone marrow (BM), support homing of hematopoietic stem cells (HSCs) and self-renewal in the BM. These cells have the potential to differentiate into tissues of mesenchymal origin, such as fibroblasts, adipocytes, cardiomyocytes, and stromal cells. MSCs can express surface molecules like CD13, CD29, CD44, CD73, CD90, CD166, CXCL12 and toll-like receptors (TLRs). Different factors, such as TGF-β, IL-10, IDO, PGE-2, sHLA-G5, HO, and Galectin-3, secreted by MSCs, induce interaction in cell to cell immunomodulatory effects on innate and adaptive cells of the immune system. Furthermore, these cells can stimulate and increase the TH2 and regulatory T-cells through inhibitory effects on the immune system. MSCs originate from the BM and other tissues including the brain, adipose tissue, peripheral blood, cornea, thymus, spleen, fallopian tube, placenta, Wharton's jelly and umbilical cord blood. Many studies have focused on two significant features of MSC therapy: (I) MSCs can modulate T-cell-mediated immunological responses, and (II) systemically administered MSCs home in to sites of ischemia or injury. In this review, we describe the known mechanisms of immunomodulation and homing of MSCs. As a result, this review emphasizes the functional role of MSCs in modulating immune responses, their capability in homing to injured tissue, and their clinical therapeutic potential.
Collapse
Affiliation(s)
- Amirbahman Rahimzadeh
- a Hematology and Oncology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee, Tabriz University of Medical Sciences , Tabriz , Iran
| | | | - Aliakbar Movassaghpour
- a Hematology and Oncology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Karim Shamsasenjan
- d Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine , Tabriz , Iran
| | - Saber Kariminekoo
- a Hematology and Oncology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Talebi
- e Hematology and Blood Banking Department, Faculty of Medical Science , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Abolfazl Shekari
- f Department Of Medical Genetic , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Vahideh Zeighamian
- g Department of Medical Biotechnology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Masoud Gandomkar Ghalhar
- g Department of Medical Biotechnology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Abolfazl Akbarzadeh
- h Department of Medical Nanotechnology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
36
|
Bara JJ, Richards RG, Alini M, Stoddart MJ. Concise Review: Bone Marrow-Derived Mesenchymal Stem Cells Change Phenotype Following In Vitro Culture: Implications for Basic Research and the Clinic. Stem Cells 2014; 32:1713-23. [DOI: 10.1002/stem.1649] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/12/2013] [Accepted: 12/21/2013] [Indexed: 12/18/2022]
Affiliation(s)
| | | | - Mauro Alini
- AO Research Institute Davos; Davos Platz 7270 Davos Switzerland
| | | |
Collapse
|
37
|
Fausther M, Lavoie EG, Goree JR, Baldini G, Dranoff JA. NT5E mutations that cause human disease are associated with intracellular mistrafficking of NT5E protein. PLoS One 2014; 9:e98568. [PMID: 24887587 PMCID: PMC4041762 DOI: 10.1371/journal.pone.0098568] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/05/2014] [Indexed: 11/21/2022] Open
Abstract
Ecto-5′-nucleotidase/CD73/NT5E, the product of the NT5E gene, is the dominant enzyme in the generation of adenosine from degradation of AMP in the extracellular environment. Nonsense (c.662C→A, p.S221X designated F1, c.1609dupA, p.V537fsX7 designated F3) and missense (c.1073G→A, p.C358Y designated F2) NT5E gene mutations in three distinct families have been shown recently to cause premature arterial calcification disease in human patients. However, the underlying mechanisms by which loss-of-function NT5E mutations cause human disease are unknown. We hypothesized that human NT5E gene mutations cause mistrafficking of the defective proteins within cells, ultimately blocking NT5E catalytic function. To test this hypothesis, plasmids encoding cDNAs of wild type and mutant human NT5E tagged with the fluorescent probe DsRed were generated and used for transfection and heterologous expression in immortalized monkey COS-7 kidney cells that lack native NT5E protein. Enzyme histochemistry and Malachite green assays were performed to assess the biochemical activities of wild type and mutant fusion NT5E proteins. Subcellular trafficking of fusion NT5E proteins was monitored by confocal microscopy and western blot analysis of fractionated cell constituents. All 3 F1, F2, and F3 mutations result in a protein with significantly reduced trafficking to the plasma membrane and reduced ER retention as compared to wild type protein. Confocal immunofluorescence demonstrates vesicles containing DsRed-tagged NT5E proteins (F1, F2 and F3) in the cell synthetic apparatus. All 3 mutations resulted in absent NT5E enzymatic activity at the cell surface. In conclusion, three familial NT5E mutations (F1, F2, F3) result in novel trafficking defects associated with human disease. These novel genetic causes of human disease suggest that the syndrome of premature arterial calcification due to NT5E mutations may also involve a novel “trafficking-opathy”.
Collapse
Affiliation(s)
- Michel Fausther
- Division of Gastroenterology & Hepatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Research Service, Central Arkansas VA Healthcare System, Little Rock, Arkansas, United States of America
| | - Elise G. Lavoie
- Division of Gastroenterology & Hepatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Research Service, Central Arkansas VA Healthcare System, Little Rock, Arkansas, United States of America
| | - Jessica R. Goree
- Division of Gastroenterology & Hepatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Research Service, Central Arkansas VA Healthcare System, Little Rock, Arkansas, United States of America
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jonathan A. Dranoff
- Division of Gastroenterology & Hepatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Research Service, Central Arkansas VA Healthcare System, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Sempere JM, Martinez-Peinado P, Arribas MI, Reig JA, De La Sen ML, Zubcoff JJ, Fraga MF, Fernández AF, Santana A, Roche E. Single cell-derived clones from human adipose stem cells present different immunomodulatory properties. Clin Exp Immunol 2014; 176:255-65. [PMID: 24666184 PMCID: PMC3992038 DOI: 10.1111/cei.12270] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2014] [Indexed: 12/18/2022] Open
Abstract
Human adipose mesenchymal stem cells are a heterogeneous population, where cell cultures derived from single-cell-expanded clones present varying degrees of differential plasticity. This work focuses on the immunomodulatory/anti-inflammatory properties of these cells. To this end, five single-cell clones were isolated (generally called 1.X and 3.X) from two volunteers. Regarding the expression level of the lineage-characteristic surface antigens, clones 1·10 and 1·22 expressed the lowest amounts, while clones 3·10 and 3·5 expressed more CD105 than the rest and clone 1·7 expressed higher amounts of CD73 and CD44. Regarding cytokine secretion, all clones were capable of spontaneously releasing high levels of interleukin (IL)-6 and low to moderate levels of IL-8. These differences can be explained in part by the distinct methylation profile exhibited by the clones. Furthermore, and after lipopolysaccharide stimulation, clone 3.X produced the highest amounts of proinflammatory cytokines such as IL-1β, while clones 1·10 and 1·22 highly expressed IL-4 and IL-5. In co-culture experiments, clones 1.X are, together, more potent inhibitors than clones 3.X for proliferation of total, CD3(+) T, CD4(+) T and CD8(+) T lymphocytes and natural killer (NK) cells. The results of this work indicate that the adipose stem cell population is heterogeneous in cytokine production profile, and that isolation, characterization and selection of the appropriate cell clone is a more exact method for the possible treatment of different patients or pathologies.
Collapse
Affiliation(s)
- J M Sempere
- Immunology Division, Biotechnology Department, University of AlicanteSan Vicente del Raspeig, Alicante, Spain
| | - P Martinez-Peinado
- Immunology Division, Biotechnology Department, University of AlicanteSan Vicente del Raspeig, Alicante, Spain
| | - M I Arribas
- Biochemistry and Cell Therapy Unit, Institute of Bioengineering, University Miguel HernandezElche, Alicante, Spain
| | - J A Reig
- Biochemistry and Cell Therapy Unit, Institute of Bioengineering, University Miguel HernandezElche, Alicante, Spain
| | - M L De La Sen
- Immunology Division, Biotechnology Department, University of AlicanteSan Vicente del Raspeig, Alicante, Spain
| | - J J Zubcoff
- Department of Statistics, University of AlicanteSan Vicente del Raspeig, Alicante, Spain
| | - M F Fraga
- Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSICCantoblanco, Madrid, Spain
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, University of OviedoOviedo, Asturias, Spain
| | - A F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, University of OviedoOviedo, Asturias, Spain
| | - A Santana
- Research Unit, Gran Canaria Hospital Dr Negrin and Genetic Unit, Childhood Hospital ComplexLas Palmas, Canary Islands, Spain
| | - E Roche
- Biochemistry and Cell Therapy Unit, Institute of Bioengineering, University Miguel HernandezElche, Alicante, Spain
- CIBERobn (Fisiopatología de la Obesidad y la Nutrición CB12/03/30038), Instituto de Salud Carlos IIIMajadahonda, Madrid, Spain
| |
Collapse
|
39
|
Cordeiro-Spinetti E, de Mello W, Trindade LS, Taub DD, Taichman RS, Balduino A. Human bone marrow mesenchymal progenitors: perspectives on an optimized in vitro manipulation. Front Cell Dev Biol 2014; 2:7. [PMID: 25364715 PMCID: PMC4207019 DOI: 10.3389/fcell.2014.00007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/24/2014] [Indexed: 01/14/2023] Open
Abstract
When it comes to regenerative medicine, mesenchymal stem cells (MSCs) are considered one of the most promising cell types for use in many cell therapies and bioengineering protocols. The International Society of Cellular Therapy recommended minimal criteria for defining multipotential MSC is based on adhesion and multipotency in vitro, and the presence or absence of select surface markers. Though these criteria help minimize discrepancies and allow some comparisons of data generated in different laboratories, the conditions in which cells are isolated and expanded are often not considered. Herein, we propose and recommend a few procedures to be followed to facilitate the establishment of quality control standards when working with mesenchymal progenitors isolation and expansion. Following these procedures, the classic Colony-Forming Unit-Fibroblast (CFU-f) assay is revisited and three major topics are considered to define conditions and to assist on protocol optimization and data interpretation. We envision that the creation of a guideline will help in the identification and isolation of long-term stem cells and short-term progenitors to better explore their regenerative potential for multiple therapeutic purposes.
Collapse
Affiliation(s)
| | - Wallace de Mello
- LaBioTeC, Universidade Veiga de Almeida Rio de Janeiro, Brazil ; Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz Fiocruz, Rio de Janeiro, Brazil
| | | | - Dennis D Taub
- Department of Vetarans Affairs, Hematology and Immunology Research, Washington DC Veterans Affairs Medical Center Washington, DC, USA
| | - Russell S Taichman
- School of Dentistry, Department of Periodontics and Oral Medicine, University of Michigan Ann Arbor, MI, USA
| | - Alex Balduino
- LaBioTeC, Universidade Veiga de Almeida Rio de Janeiro, Brazil ; Excellion Serviços Biomédicos Petrópolis, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Payushina OV, Butorina NN, Sheveleva ON, Bukhinnik SS, Domaratskaya EI. Clonal growth, phenotype, and differentiation potential of mesenchymal stromal cells derived from the rat fetal bone. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2014; 453:394-6. [PMID: 24385180 DOI: 10.1134/s0012496613060148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Indexed: 11/22/2022]
Affiliation(s)
- O V Payushina
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | |
Collapse
|
41
|
Wang T, Liao T, Wang H, Deng W, Yu D. Transplantation of bone marrow stromal cells overexpressing human vascular endothelial growth factor 165 enhances tissue repair in a rat model of radiation-induced injury. Chin Med J (Engl) 2014; 127:1093-1099. [PMID: 24622441 DOI: 10.3760/cma.j.issn.0366-6999.20132337] [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: 01/05/2025] Open
Abstract
BACKGROUND The multilineage differentiation potential ability of bone marrow stromal cells (BMSCs) showed great potential in tissue engineering, while vascular endothelial growth factor 165 (VEGF165) promotes vasculogenesis and further promotes tissue regeneration. This study aimed to assess the ability of rat BMSCs expressing human VEGF A165 (hVEGF165) to promote tissue repair in rat model of radiation-induced injury. METHODS Rat BMSCs were isolated from the tibia. Plasmid DNA expressing hVEGF165 was stably transfected into BMSCs using liposomes. The right hindlimb muscle of 40 rats was irradiated using a (60)Co γ source (total dose 30 Gy). The animals were divided into four groups (n = 10): not injected with BMSCs (control; group 1) or intramuscularly injected two times (once in 2 weeks) with pcDNA(TM)3.1-transfected BMSCs (group 2), untransfected BMSCs (group 3), or hVEGF165-transfected BMSCs (group 4). Angiography was performed 1 week after the last injection of BMSCs; samples of the hindlimb muscle were subjected to transmission electron microscopy, ultrastructural analysis, reverse transcription-PCR (RT-PCR), Western blotting, and immunohistochemistry. RESULTS Rat BMSCs with multipotent differentiation capacity were isolated. hVEGF165-transfected BMSCs overexpressed hVEGF165 mRNA and protein. Injection of BMSCs (groups 2-4) increased the average vessel number, density, diameter, and cross-sectional area; mRNA expression of the myogenic markers including myoblast determination protein, myogenin, and a-smooth muscle actin; and CD31 protein expression; and promoted the repair of blood vessels and myofibers after radiation-induced injury compared to group 1; each of these parameters and hVEGF165 mRNA or protein expression were markedly improved in rats injected with hVEGF165-transfected BMSCs compared to groups 2 and 3. CONCLUSIONS BMSCs expressing hVEGF165 enhanced the repair of radiation-induced tissue injury by promoting vasculogenesis and muscle fiber regeneration. BMSCs expressing hVEGF165 may have potential clinical applications.
Collapse
Affiliation(s)
- Tao Wang
- Department of Oral and Maxillofacial Surgery, People's Hospital of Hainan Province, Haikou, Hainan 570311, China.
| | - Tian'an Liao
- Department of Oral and Maxillofacial Surgery, People's Hospital of Hainan Province, Haikou, Hainan 570311, China
| | - Hong Wang
- Department of Oral and Maxillofacial Surgery, People's Hospital of Hainan Province, Haikou, Hainan 570311, China
| | - Wei Deng
- Department of Oral and Maxillofacial Surgery, People's Hospital of Hainan Province, Haikou, Hainan 570311, China
| | - Dahai Yu
- Department of Oral Maxillofacial Surgery, Stomatological Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
42
|
Diogo MM, da Silva CL, Cabral JMS. Separation Technologies for Stem Cell Bioprocessing. CELL ENGINEERING 2014. [DOI: 10.1007/978-94-007-7196-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
43
|
Fan L, Hu K, Ji K, Sun Q, Xiong J, Yang L, Liu H. Directed differentiation of aged human bone marrow multipotent stem cells effectively generates dopamine neurons. In Vitro Cell Dev Biol Anim 2013; 50:304-12. [PMID: 24163158 DOI: 10.1007/s11626-013-9701-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/27/2013] [Indexed: 12/11/2022]
Abstract
This study aimed to isolate aged human bone marrow multipotent stem cells (hAMSCs) with the potential for multilineage differentiation and to directly induce the cells to generate dopamine neurons, which could be used for Parkinson's disease therapy. We compared different culture methods for stem cells from aged human bone marrow and identified hAMSCs that could proliferate in vitro for at least 60 doubling times. Using RT-PCR and IHC, we found that these hAMSCs expressed pluripotent genes, such as Oct4, Sox2, and Nanog. In vitro studies also proved that hAMSCs could differentiate into three germ layer-derived cell types, such as osteogenic, chondrogenic, adipogenic, and hepatocyte-liked cells. After induction for more than 20 d in vitro with retinoic acid, basic fibroblast growth factor, and sonic hedgehog using a two-step method and withdrawal of serum, hAMSCs could differentiate into dopamine neurons at the positive ratio of 70%, which showed DA secretion function upon depolarization. In conclusion, we suggest that hAMSCs can be used as cell sources to develop medical treatments to prevent the progression of Parkinson's disease, especially in aged persons.
Collapse
Affiliation(s)
- Lixing Fan
- Research Center of Developmental Biology, Second Military Medical University, XiangYin road 800, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Chick stem cells: current progress and future prospects. Stem Cell Res 2013; 11:1378-92. [PMID: 24103496 PMCID: PMC3989061 DOI: 10.1016/j.scr.2013.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 09/06/2013] [Accepted: 09/13/2013] [Indexed: 12/15/2022] Open
Abstract
Chick embryonic stem cells (cESCs) can be derived from cells obtained from stage X embryos (blastoderm stage); these have the ability to contribute to all somatic lineages in chimaeras, but not to the germ line. However, lines of stem cells that are able to contribute to the germ line can be established from chick primordial germ cells (cPGCs) and embryonic germ cells (cEGCs). This review provides information on avian stem cells, emphasizing different sources of cells and current methods for derivation and culture of pluripotent cells from chick embryos. We also review technologies for isolation and derivation of chicken germ cells and the production of transgenic birds. Chick embryonic stem cells (cESCs) can be derived from a variety of sources. cESCs can contribute to all somatic cell types but not to the germ line. germ cells can be isolated from early embryos, embryonic blood and gonads. germ cells can establish self-renewing lines and contribute to the germline.
Collapse
|
45
|
Watson JT, Foo T, Wu J, Moed BR, Thorpe M, Schon L, Zhang Z. CD271 as a marker for mesenchymal stem cells in bone marrow versus umbilical cord blood. Cells Tissues Organs 2013; 197:496-504. [PMID: 23689142 DOI: 10.1159/000348794] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2013] [Indexed: 11/19/2022] Open
Abstract
CD271 has been applied to isolate mesenchymal stem cells (MSCs) from bone marrow and other tissues. Umbilical cord blood is a unique resource of stem cells and endothelial progenitor cells. Isolation of MSCs from umbilical cord blood, however, has been inefficient and inconsistent. This study was designed to examine the potential application of CD271 as a marker for the isolation of MSCs from umbilical cord blood. CD271+ cells were isolated from umbilical cord blood and bone marrow using CD271 antibody-conjugated microbeads, and characterized in osteogenic, chondrogenic and adipogenic differentiation. CD271+ cells from umbilical cord blood were slow to proliferate compared with those isolated from bone marrow. While CD271+ cells from bone marrow differentiated into osteogenic, chondrogenic and adipogenic lineages, there were no sound indications of differentiation by CD271+ cells from umbilical cord blood under the same differentiation conditions applied to the CD271+ cells from bone marrow. The study also found that bone marrow CD271+ cells remarkably upregulated the expression of chondrogenic genes under chondrogenic differentiation induction. When implanted into bone defects in mice, CD271+ cells from bone marrow regenerated significant bone, but the counterparts in umbilical cord blood formed little bone in the bone defects. In conclusion, CD271 is an efficient marker for MSC isolation from bone marrow but has failed to isolate MSCs from umbilical cord blood. CD271+ cells in bone marrow are particularly chondrogenic. The property of CD271+ cells is unique but varies from different tissues.
Collapse
Affiliation(s)
- J Tracy Watson
- Department of Orthopedic Surgery, Saint Louis University, St. Louis, MO, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Gaafar TM, Abdel Rahman HA, Attia W, Hamza HS, Brockmeier K, El Hawary RE. Comparative characteristics of endothelial-like cells derived from human adipose mesenchymal stem cells and umbilical cord blood-derived endothelial cells. Clin Exp Med 2013; 14:177-84. [PMID: 23649875 DOI: 10.1007/s10238-013-0238-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/25/2013] [Indexed: 01/22/2023]
Abstract
Adult peripheral blood contains a limited number of endothelial progenitor cells that can be isolated for treatment of ischemic diseases. The adipose tissue became an interesting source of stem cells for regenerative medicine. This study aimed to investigate the phenotype of cells obtained by culturing adipose-derived mesenchymal stem cells (ad-MSCs) in the presence of endothelial growth supplements compared to endothelial cells obtained from umbilical cord blood (UCB). Passage 3 ad-MSCs and mononuclear layer from UCB were cultured in presence of endothelial growth media for 3 weeks followed by their characterization by flow cytometry and polymerase chain reaction. After culture in endothelial inductive media, ad-MSCs expressed endothelial genes and some endothelial marker proteins as CD31 and CD34, respectively. Adipose tissue could be a reliable source for easy obtaining, expanding and differentiating MSCs into endothelial-like cells for autologous cell-based therapy.
Collapse
Affiliation(s)
- Taghrid M Gaafar
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, 16 Street 107, Maadi, Cairo, Egypt,
| | | | | | | | | | | |
Collapse
|
47
|
Choi WY, Jeon HG, Chung Y, Lim JJ, Shin DH, Kim JM, Ki BS, Song SH, Choi SJ, Park KH, Shim SH, Moon J, Jung SJ, Kang HM, Park S, Chung HM, Ko JJ, Cha KY, Yoon TK, Kim H, Lee DR. Isolation and characterization of novel, highly proliferative human CD34/CD73-double-positive testis-derived stem cells for cell therapy. Stem Cells Dev 2013; 22:2158-73. [PMID: 23509942 DOI: 10.1089/scd.2012.0385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human adult stem cells are a readily available multipotent cell source that can be used in regenerative medicine. Despite many advantages, including low tumorigenicity, their rapid senescence and limited plasticity have curtailed their use in cell-based therapies. In this study, we isolated CD34/CD73-double-positive (CD34(+)/CD73(+)) testicular stromal cells (HTSCs) and found that the expression of CD34 was closely related to the cells' stemness and proliferation. The CD34(+)/CD73(+) cells grew in vitro for an extended period of time, yielding a multitude of cells (5.6×10(16) cells) without forming tumors in vivo. They also differentiated into all three germ layer lineages both in vitro and in vivo, produced cartilage more efficiently compared to bone marrow stem cells and, importantly, restored erectile function in a cavernous nerve crush injury rat model. Thus, these HTSCs may represent a promising new autologous cell source for clinical use.
Collapse
Affiliation(s)
- Won Yun Choi
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Meric A, Yenigun A, Yenigun VB, Dogan R, Ozturan O. Comparison of Chondrocytes Produced from Adipose Tissue–Derived Stem Cells and Cartilage Tissue. J Craniofac Surg 2013; 24:830-3. [DOI: 10.1097/scs.0b013e3182902779] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
49
|
Lin CS, Xin ZC, Dai J, Lue TF. Commonly used mesenchymal stem cell markers and tracking labels: Limitations and challenges. Histol Histopathol 2013; 28:1109-16. [PMID: 23588700 DOI: 10.14670/hh-28.1109] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Early observations that cultured mesenchymal stem cells (MSCs) could be induced to exhibit certain characteristics of osteocytes and chondrocytes led to the proposal that they could be transplanted for tissue repair through cellular differentiation. Therefore, many subsequent preclinical studies with transplanted MSCs have strived to demonstrate that cellular differentiation was the underlying mechanism for the therapeutic effect. These studies generally followed the minimal criteria set by The International Society for Cellular Therapy in assuring MSC identity by using CD70, CD90, and CD105 as positive markers and CD34 as a negative marker. However, the three positive markers are co-expressed in a wide variety of cells, and therefore, even when used in combination, they are certainly incapable of identifying MSCs in vivo. Another frequently used MSC marker, Stro-1, has been shown to be an endothelial antigen and whether it can identify MSCs in vivo remains unknown. On the other hand, the proposed negative marker CD34 has increasingly been shown to be expressed in native MSCs, such as in the adipose tissue. It has also helped establish that MSCs are likely vascular stem cells (VSCs) that reside in the capillaries and in the adventitia of larger blood vessels. These cells do not express CD31, CD104b, or α-SMA, and therefore are designated as CD34+CD31-CD140b-SMA-. Many preclinical MSC transplantation studies have also attempted to demonstrate cellular differentiation by using labeled MSCs. However, all commonly used labels have shortcomings that often complicate data interpretation. The β-gal (LacZ) gene as a label is problematic because many mammalian tissues have endogenous β-gal activities. The GFP gene is similarly problematic because many mammalian tissues are endogenously fluorescent. The cell membrane label DiI can be adsorbed by host cells, and nuclear stains Hoechst dyes and DAPI can be transferred to host cells. Thymidine analog BrdU is associated with loss of cellular protein antigenicity due to harsh histological conditions. Newer thymidine analog EdU is easier to detect by chemical reaction to azide-conjugated Alexa fluors, but certain bone marrow cells are reactive to these fluors in the absence of EdU. These caveats need to be taken into consideration when designing or interpreting MSC transplantation experiments.
Collapse
Affiliation(s)
- Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California 94143-0738, USA.
| | | | | | | |
Collapse
|
50
|
Fernández Vallone VB, Romaniuk MA, Choi H, Labovsky V, Otaegui J, Chasseing NA. Mesenchymal stem cells and their use in therapy: what has been achieved? Differentiation 2013; 85:1-10. [PMID: 23314286 DOI: 10.1016/j.diff.2012.08.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 07/10/2012] [Accepted: 08/16/2012] [Indexed: 12/13/2022]
Abstract
The considerable therapeutic potential of human multipotent mesenchymal stromal cells or mesenchymal stem cells (MSCs) has generated increasing interest in a wide variety of biomedical disciplines. Nevertheless, researchers report studies on MSCs using different methods of isolation and expansion, as well as different approaches to characterize them; therefore, it is increasingly difficult to compare and contrast study outcomes. To begin to address this issue, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy proposed minimal criteria to define human MSCs. First, MSCs must be plastic-adherent when maintained in standard culture conditions (α minimal essential medium plus 20% fetal bovine serum). Second, MSCs must express CD105, CD73 and CD90, and MSCs must lack expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface molecules. Third, MSCs must differentiate into osteoblasts, adipocytes and chondroblasts in vitro. MSCs are isolated from many adult tissues, in particular from bone marrow and adipose tissue. Along with their capacity to differentiate and transdifferentiate into cells of different lineages, these cells have also generated great interest for their ability to display immunomodulatory capacities. Indeed, a major breakthrough was the finding that MSCs are able to induce peripheral tolerance, suggesting that they may be used as therapeutic tools in immune-mediated disorders. Although no significant adverse events have been reported in clinical trials to date, all interventional therapies have some inherent risks. Potential risks for undesirable events, such as tumor development, that might occur while using these stem cells for therapy must be taken into account and contrasted against the potential benefits to patients.
Collapse
|