1
|
Osteoclastogenesis of human peripheral blood, bone marrow, and cord blood monocytes. Sci Rep 2023; 13:3763. [PMID: 36882450 PMCID: PMC9992388 DOI: 10.1038/s41598-023-30701-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Osteoclasts are multinucleated bone resorbing cells that can be differentiated from human monocytes in vitro. There are few studies comparing osteoclastogenesis of different monocyte sources. We compared monocytes from human bone marrow (BM), peripheral blood (PB), and umbilical cord blood (CB) and their osteoclastogenic potential by culturing them with RANKL (20 and 80 ng/ml) and M-CSF (10 ng/ml) for 14 days. We also cultured cells without growth factors, as umbilical cord blood monocytes have been reported to be able to fuse spontaneously into osteoclasts. The data was analysed on d4, d8, d11, and d14. After culture with RANKL and M-CSF, all types of cell cultures developed TRACP -positive multinuclear cells that were able to form resorption pits on human bone slices. Only occasional multinuclear cells and small infrequent resorbed areas could be found in PB and CB-derived cultures without growth factors. BM-derived cells formed greater resorption areas than PB- and CB-derived monocytes. The greatest monocyte population in BM samples were intermediate (CD14++CD16+) and in PB and CB classical monocytes (76.3% and 54.4%, respectively). In conclusion, our data demonstrates that bone resorbing osteoclasts can be differentiated from BM, PB and CB. However, the osteoclast precursor origin can affect the osteoclast properties and function.
Collapse
|
2
|
Um S, Ha J, Choi SJ, Oh W, Jin HJ. Prospects for the therapeutic development of umbilical cord blood-derived mesenchymal stem cells. World J Stem Cells 2020; 12:1511-1528. [PMID: 33505598 PMCID: PMC7789129 DOI: 10.4252/wjsc.v12.i12.1511] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/23/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is a primitive and abundant source of mesenchymal stem cells (MSCs). UCB-derived MSCs have a broad and efficient therapeutic capacity to treat various diseases and disorders. Despite the high latent self-renewal and differentiation capacity of these cells, the safety, efficacy, and yield of MSCs expanded for ex vivo clinical applications remains a concern. However, immunomodulatory effects have emerged in various disease models, exhibiting specific mechanisms of action, such as cell migration and homing, angiogenesis, anti-apoptosis, proliferation, anti-cancer, anti-fibrosis, anti-inflammation and tissue regeneration. Herein, we review the current literature pertaining to the UCB-derived MSC application as potential treatment strategies, and discuss the concerns regarding the safety and mass production issues in future applications.
Collapse
Affiliation(s)
- Soyoun Um
- Research Team for Immune Cell Therapy, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Jueun Ha
- Research Team for Osteoarthritis, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| |
Collapse
|
3
|
Current Status of Canine Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Veterinary Medicine. Stem Cells Int 2018; 2018:8329174. [PMID: 30123294 PMCID: PMC6079340 DOI: 10.1155/2018/8329174] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/19/2018] [Indexed: 12/26/2022] Open
Abstract
Stem cell therapy has prompted the expansion of veterinary medicine both experimentally and clinically, with the potential to contribute to contemporary treatment strategies for various diseases and conditions for which limited or no therapeutic options are presently available. Although the application of various types of stem cells, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue-derived mesenchymal stem cells (AT-MSCs), and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs), has promising potential to improve the health of different species, it is crucial that the benefits and drawbacks are completely evaluated before use. Umbilical cord blood (UCB) is a rich source of stem cells; nonetheless, isolation of mesenchymal stem cells (MSCs) from UCB presents technical challenges. Although MSCs have been isolated from UCB of diverse species such as human, equine, sheep, goat, and canine, there are inherent limitations of using UCB from these species for the expansion of MSCs. In this review, we investigated canine UCB (cUCB) and compared it with UCB from other species by reviewing recent articles published from February 2003 to June 2017 to gain an understanding of the limitations of cUCB in the acquisition of MSCs and to determine other suitable sources for the isolation of MSCs from canine. Our review indicates that cUCB is not an ideal source of MSCs because of insufficient volume and ethical issues. However, canine reproductive organs discarded during neutering may help broaden our understanding of effective isolation of MSCs. We recommend exploring canine reproductive and adipose tissue rather than UCB to fulfill the current need in veterinary medicine for the well-designed and ethically approved source of MSCs.
Collapse
|
4
|
The effects of culture conditions on the functionality of efficiently obtained mesenchymal stromal cells from human cord blood. Cytotherapy 2016; 18:423-37. [PMID: 26857232 DOI: 10.1016/j.jcyt.2015.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/02/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Cord blood (CB) is an attractive source of mesenchymal stromal cells (MSCs) because of its abundant availability and ease of collection. However, the success rate of generating CB-MSCs is low. In this study, our aim was to demonstrate the efficiency of our previously described method to obtain MSCs from CB and further characterize them and to study the effects of different culture conditions on MSCs. METHODS CB-MSC cultures were established in low oxygen (3%) conditions on fibronectin in 10% fetal bovine serum containing culture medium supplemented with combinations of growth factors. Cells were characterized for their adipogenic, osteogenic and chondrogenic differentiation capacity; phenotype; and HOX gene expression profile. The functionality of the cells cultured in different media was tested in vitro with angiogenesis and T-cell proliferation assays. RESULTS We demonstrate 87% efficacy in generating MSCs from CB. The established cells had typical MSC characteristics with reduced adipogenic differentiation potential and a unique HOX gene fingerprint. Growth factor-rich medium and a 3% oxygen condition enhanced cell proliferation; however, the growth factor-rich medium had a negative effect on the expression of CD90. Dexamethasone-containing medium improved the capacity of the cells to suppress T-cell proliferation, whereas the cells grown without dexamethasone were more able to support angiogenesis. CONCLUSIONS Our results demonstrate that the composition of expansion medium is critical for the functionality of MSCs and should always be appropriately defined for each purpose.
Collapse
|
5
|
Liedtke S, Sacchetti B, Laitinen A, Donsante S, Klöckers R, Laitinen S, Riminucci M, Kogler G. Low oxygen tension reveals distinct HOX codes in human cord blood-derived stromal cells associated with specific endochondral ossification capacities in vitro and in vivo. J Tissue Eng Regen Med 2016; 11:2725-2736. [PMID: 27214005 DOI: 10.1002/term.2167] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/21/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022]
Abstract
Effects of oxygen tension on the generation, expansion, proliferation and differentiation of stromal cell types is widely described in the literature. However, data on the internal heterogeneity of applied cell populations at different O2 levels and possible impacts on differentiation potentials are controversial. Here, the expression of 39 human HOX genes was determined in neonatal cord blood stromal cells and linked to differentiation-associated signatures. In cord blood, unrestricted somatic stromal cells (USSCs), lacking HOX gene expression, and cord blood-derived multipotent stromal cells (CB-MSCs), expressing about 20 HOX genes, are distinguished by their specific HOX code. Interestingly, 74% of the clones generated at 21% O2 were HOX-negative USSCs, whereas 73% of upcoming clones at 3% O2 were HOX-positive CB-MSCs. In order to better categorize distinct cell lines generated at 3% O2 , the expression of all 39 HOX genes within HOX clusters A, B, C and D were tested and new subtypes defined: cells negative in all four HOX clusters (USSCs); cells positive in all four clusters (CB-MSCsABCD ); and subpopulations missing a single cluster (CB-MSCsACD and CB-MSCsBCD ). Comprehensive qPCR analyses of established chondro-osteomarkers revealed subtype-specific signatures verifiably associated with in vitro and in vivo differentiation capacity. The data presented here underline the necessity of better characterizing distinct cell populations at a clonal level, taking advantage of the inherent specific HOX code as a distinguishing feature between individual subtypes. Moreover, the correlation of subtype-specific molecular signatures with in vitro and in vivo bone formation is discussed. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Stefanie Liedtke
- Institute of Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Medical Centre, Düsseldorf, Germany
| | - Benedetto Sacchetti
- Stem Cell Laboratory, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Anita Laitinen
- Research and Development, Medical Services, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Samantha Donsante
- Stem Cell Laboratory, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Robert Klöckers
- Institute of Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Medical Centre, Düsseldorf, Germany
| | - Saara Laitinen
- Research and Development, Medical Services, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Mara Riminucci
- Stem Cell Laboratory, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Gesine Kogler
- Institute of Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Medical Centre, Düsseldorf, Germany
| |
Collapse
|
6
|
Kerkelä E, Laitinen A, Räbinä J, Valkonen S, Takatalo M, Larjo A, Veijola J, Lampinen M, Siljander P, Lehenkari P, Alfthan K, Laitinen S. Adenosinergic Immunosuppression by Human Mesenchymal Stromal Cells Requires Co-Operation with T cells. Stem Cells 2016; 34:781-90. [PMID: 26731338 DOI: 10.1002/stem.2280] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 10/06/2015] [Accepted: 10/30/2015] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) have the capacity to counteract excessive inflammatory responses. MSCs possess a range of immunomodulatory mechanisms, which can be deployed in response to signals in a particular environment and in concert with other immune cells. One immunosuppressive mechanism, not so well-known in MSCs, is mediated via adenosinergic pathway by ectonucleotidases CD73 and CD39. In this study, we demonstrate that adenosine is actively produced from adenosine 5'-monophosphate (AMP) by CD73 on MSCs and MSC-derived extracellular vesicles (EVs). Our results indicate that although MSCs express CD39 at low level and it colocalizes with CD73 in bulge areas of membranes, the most efficient adenosine production from adenosine 5'-triphosphate (ATP) requires co-operation of MSCs and activated T cells. Highly CD39 expressing activated T cells produce AMP from ATP and MSCs produce adenosine from AMP via CD73 activity. Furthermore, adenosinergic signaling plays a role in suppression of T cell proliferation in vitro. In conclusion, this study shows that adenosinergic signaling is an important immunoregulatory mechanism of MSCs, especially in situations where ATP is present in the extracellular environment, like in tissue injury. An efficient production of immunosuppressive adenosine is dependent on the concerted action of CD39-positive immune cells with CD73-positive cells such as MSCs or their EVs.
Collapse
Affiliation(s)
- Erja Kerkelä
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | | | - Sami Valkonen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Antti Larjo
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Johanna Veijola
- Institute of Clinical Medicine, Division of Surgery.,Institute of Biomedicine, Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland.,Clinical Research Center, Department of Surgery and Intensive Care, Oulu University Hospital, Oulu, Finland
| | | | - Pia Siljander
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Petri Lehenkari
- Institute of Clinical Medicine, Division of Surgery.,Institute of Biomedicine, Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland.,Clinical Research Center, Department of Surgery and Intensive Care, Oulu University Hospital, Oulu, Finland
| | | | | |
Collapse
|
7
|
Barilani M, Lavazza C, Viganò M, Montemurro T, Boldrin V, Parazzi V, Montelatici E, Crosti M, Moro M, Giordano R, Lazzari L. Dissection of the cord blood stromal component reveals predictive parameters for culture outcome. Stem Cells Dev 2015; 24:104-14. [PMID: 25046283 DOI: 10.1089/scd.2014.0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In regenerative medicine, human cord blood-derived multipotent mesenchymal stromal cells (CBMSCs) stand out for their biological peculiarities demonstrated in in vitro and in vivo preclinical studies. Here, we present our 9-year experience for the consistent isolation of CBMSCs. Although nearly one CB unit out of two retains the potential to give rise to MSC colonies, only 46% of them can be cultured till low passages (P≥4), but one-fourth of those reaches even higher passages (P≥8). Subsequent characterization for morphological, clonal, differentiation, and proliferation properties revealed two divergent CBMSC behaviors. In particular, a cumulative population doublings cut-off (CPD=15) was identified that undoubtedly distinguishes two growth curves, and different degrees of commitment toward osteogenesis were observed. These data clearly show the existence of at least two distinct CBMSC subsets: one mainly short-living and less proliferative (SL-CBMSCs), the other long-living, with higher growth rate, and, very importantly, with significantly (P≤0.01) longer telomere (LL-CBMSCs). Moreover, significant differences in the immunoprofile before seeding were found among CB units giving rise to LL-CBMSCs or SL-CBMSCs or showing no colony formation. Finally, all the aforementioned results provided a peculiar and useful set of parameters potentially predictive for CBMSC culture outcome.
Collapse
Affiliation(s)
- Mario Barilani
- 1 Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Peters EB, Liu B, Christoforou N, West JL, Truskey GA. Umbilical Cord Blood-Derived Mononuclear Cells Exhibit Pericyte-Like Phenotype and Support Network Formation of Endothelial Progenitor Cells In Vitro. Ann Biomed Eng 2015; 43:2552-68. [PMID: 25777295 DOI: 10.1007/s10439-015-1301-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/11/2015] [Indexed: 01/17/2023]
Abstract
Umbilical cord blood represents a promising cell source for pro-angiogenic therapies. The present study examined the potential of mononuclear cells (MNCs) from umbilical cord blood to support endothelial progenitor cell (EPC) microvessel formation. MNCs were isolated from the cord blood of 20 separate donors and selected for further characterization based upon their proliferation potential and morphological resemblance to human vascular pericytes (HVPs). MNCs were screened for their ability to support EPC network formation using an in vitro assay (Matrigel™) as well as a reductionist, coculture system consisting of no additional angiogenic cytokines beyond those present in serum. In less than 15% of the isolations, we identified a population of highly proliferative MNCs that phenotypically resembled HVPs as assessed by expression of PDGFR-β, NG2, α-SMA, and ephrin-B2. Within a Matrigel™ system, MNCs demonstrated pericyte-like function through colocalization to EPC networks and similar effects as HVPs upon total EPC tubule length (p = 0.95) and number of branch points (p = 0.93). In a reductionist coculture system, MNCs served as pro-angiogenic mural cells by supporting EPC network formation to a significantly greater extent than HVP cocultures, by day 14 of coculture, as evidenced through EPC total tubule length (p < 0.0001) and number of branch points (p < 0.0001). Our findings are significant as we demonstrate mural cell progenitors can be isolated from umbilical cord blood and develop culture conditions to support their use in microvascular tissue engineering applications.
Collapse
|
9
|
Suila H, Hirvonen T, Kotovuori A, Ritamo I, Kerkelä E, Anderson H, Natunen S, Tuimala J, Laitinen S, Nystedt J, Räbinä J, Valmu L. Human Umbilical Cord Blood-Derived Mesenchymal Stromal Cells Display a Novel Interaction between P-Selectin and Galectin-1. Scand J Immunol 2014; 80:12-21. [DOI: 10.1111/sji.12179] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/27/2014] [Indexed: 12/13/2022]
Affiliation(s)
- H. Suila
- Finnish Red Cross Blood Service; Helsinki Finland
| | - T. Hirvonen
- Finnish Red Cross Blood Service; Helsinki Finland
| | - A. Kotovuori
- Finnish Red Cross Blood Service; Helsinki Finland
| | - I. Ritamo
- Finnish Red Cross Blood Service; Helsinki Finland
| | - E. Kerkelä
- Finnish Red Cross Blood Service; Helsinki Finland
| | - H. Anderson
- Finnish Red Cross Blood Service; Helsinki Finland
| | - S. Natunen
- Finnish Red Cross Blood Service; Helsinki Finland
| | - J. Tuimala
- Finnish Red Cross Blood Service; Helsinki Finland
| | - S. Laitinen
- Finnish Red Cross Blood Service; Helsinki Finland
| | - J. Nystedt
- Finnish Red Cross Blood Service; Helsinki Finland
| | - J. Räbinä
- Finnish Red Cross Blood Service; Helsinki Finland
| | - L. Valmu
- Finnish Red Cross Blood Service; Helsinki Finland
| |
Collapse
|
10
|
Suila H, Hirvonen T, Ritamo I, Natunen S, Tuimala J, Laitinen S, Anderson H, Nystedt J, Räbinä J, Valmu L. Extracellular o-linked N-acetylglucosamine is enriched in stem cells derived from human umbilical cord blood. Biores Open Access 2014; 3:39-44. [PMID: 24804163 PMCID: PMC3995142 DOI: 10.1089/biores.2013.0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stem cells have a unique ability to self-renew and differentiate into diverse cell types. Currently, stem cells from various sources are being explored as a promising new treatment for a variety of human diseases. A diverse set of functional and phenotypical markers are used in the characterization of specific therapeutic stem cell populations. The glycans on the stem cell surface respond rapidly to alterations in cellular state and signaling and are therefore ideal for identifying even minor changes in cell populations. Many stem cell markers are based on cell surface glycan epitopes including the widely used markers SSEA-3, SSEA-4, Tra 1-60, and Tra 1-81. We have now discovered by mRNA analysis that a novel glycosyltranferase, epidermal growth factor (EGF) domain-specific O-linked GlcNAc transferase (EOGT), is highly expressed in stem cells. EOGT is responsible for adding O-linked N-acetylglucosamine (O-GlcNAc) to folded EGF domains on extracellular proteins, such as those on the Notch receptors. We were able to show by immunological assays that human umbilical cord blood–derived mesenchymal stromal cells display O-GlcNAc, the product of EOGT, and that O-GlcNAc is further elongated with galactose to form O-linked N-acetyllactosamine. We suggest that these novel glycans are involved in the fine tuning of Notch receptor signaling pathways in stem cells.
Collapse
Affiliation(s)
- Heli Suila
- Finnish Red Cross Blood Service , Helsinki, Finland
| | - Tia Hirvonen
- Finnish Red Cross Blood Service , Helsinki, Finland
| | - Ilja Ritamo
- Finnish Red Cross Blood Service , Helsinki, Finland
| | - Suvi Natunen
- Finnish Red Cross Blood Service , Helsinki, Finland
| | | | | | | | | | | | - Leena Valmu
- Finnish Red Cross Blood Service , Helsinki, Finland
| |
Collapse
|
11
|
Karlupia N, Manley NC, Prasad K, Schäfer R, Steinberg GK. Intraarterial transplantation of human umbilical cord blood mononuclear cells is more efficacious and safer compared with umbilical cord mesenchymal stromal cells in a rodent stroke model. Stem Cell Res Ther 2014; 5:45. [PMID: 24690461 PMCID: PMC4055161 DOI: 10.1186/scrt434] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 10/21/2013] [Accepted: 03/20/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Stroke is the second leading cause of death worldwide, claims six lives every 60 seconds, and is a leading cause of adult disability across the globe. Tissue plasminogen activator, the only United States Food and Drug Administration (FDA)-approved drug currently available, has a narrow therapeutic time window of less than 5 hours. In the past decade, cells derived from the human umbilical cord (HUC) have emerged as a potential therapeutic alternative for stroke; however, the most effective HUC-derived cell population remains unknown. METHODS We compared three cell populations derived from the human umbilical cord: cord blood mononuclear cells (cbMNCs); cord blood mesenchymal stromal cells (cbMSCs), a subpopulation of cbMNCs; and cord matrix MSCs (cmMSCs). We characterized these cells in vitro with flow cytometry and assessed the cells' in vivo efficacy in a 2-hour transient middle cerebral artery occlusion (MCAo) rat model of stroke. cbMNCs, cbMSCs, and cmMSCs were each transplanted intraarterially at 24 hours after stroke. RESULTS A reduction in neurologic deficit and infarct area was observed in all three cell groups; however, this reduction was significantly enhanced in the cbMNC group compared with the cmMSC group. At 2 weeks after stroke, human nuclei-positive cells were present in the ischemic hemispheres of immunocompetent stroke rats in all three cell groups. Significantly decreased expression of rat brain-derived neurotrophic factor mRNA was observed in the ischemic hemispheres of all three cell-treated and phosphate-buffered saline (PBS) group animals compared with sham animals, although the decrease was least in cbMNC-treated animals. Significantly decreased expression of rat interleukin (IL)-2 mRNA and IL-6 mRNA was seen only in the cbMSC group. Notably, more severe complications (death, eye inflammation) were observed in the cmMSC group compared with the cbMNC and cbMSC groups. CONCLUSIONS All three tested cell types promoted recovery after stroke, but cbMNCs showed enhanced recovery and fewer complications compared with cmMSCs.
Collapse
Affiliation(s)
- Neha Karlupia
- Department of Neurosurgery, R281, Stanford School of Medicine, Stanford University, Stanford, CA 94305-5487, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Neurology, All India Institute of medical Sciences, New Delhi 110029, India
| | - Nathan C Manley
- Department of Neurosurgery, R281, Stanford School of Medicine, Stanford University, Stanford, CA 94305-5487, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kameshwar Prasad
- Department of Neurology, All India Institute of medical Sciences, New Delhi 110029, India
| | - Richard Schäfer
- Department of Neurosurgery, R281, Stanford School of Medicine, Stanford University, Stanford, CA 94305-5487, USA
| | - Gary K Steinberg
- Department of Neurosurgery, R281, Stanford School of Medicine, Stanford University, Stanford, CA 94305-5487, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Kilpinen L, Impola U, Sankkila L, Ritamo I, Aatonen M, Kilpinen S, Tuimala J, Valmu L, Levijoki J, Finckenberg P, Siljander P, Kankuri E, Mervaala E, Laitinen S. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J Extracell Vesicles 2013; 2:21927. [PMID: 24349659 PMCID: PMC3860334 DOI: 10.3402/jev.v2i0.21927] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/31/2013] [Accepted: 11/12/2013] [Indexed: 12/21/2022] Open
Abstract
Background Mesenchymal stromal cells (MSC) are shown to have a great therapeutic potential in many immunological disorders. Currently the therapeutic effect of MSCs is considered to be mediated via paracrine interactions with immune cells. Umbilical cord blood is an attractive but still less studied source of MSCs. We investigated the production of extracellular membrane vesicles (MVs) from human umbilical cord blood derived MSCs (hUCBMSC) in the presence (MVstim) or absence (MVctrl) of inflammatory stimulus. Methods hUCBMSCs were cultured in serum free media with or without IFN-γ and MVs were collected from conditioned media by ultracentrifugation. The protein content of MVs were analyzed by mass spectrometry. Hypoxia induced acute kidney injury rat model was used to analyze the in vivo therapeutic potential of MVs and T-cell proliferation and induction of regulatory T cells were analyzed by co-culture assays. Results Both MVstim and MVctrl showed similar T-cell modulation activity in vitro, but only MVctrls were able to protect rat kidneys from reperfusion injury in vivo. To clarify this difference in functionality we made a comparative mass spectrometric analysis of the MV protein contents. The IFN-γ stimulation induced dramatic changes in the protein content of the MVs. Complement factors (C3, C4A, C5) and lipid binding proteins (i.e apolipoproteins) were only found in the MVctrls, whereas the MVstim contained tetraspanins (CD9, CD63, CD81) and more complete proteasome complex accompanied with MHCI. We further discovered that differently produced MV pools contained specific Rab proteins suggesting that same cells, depending on external signals, produce vesicles originating from different intracellular locations. Conclusions We demonstrate by both in vitro and in vivo models accompanied with a detailed analysis of molecular characteristics that inflammatory conditioning of MSCs influence on the protein content and functional properties of MVs revealing the complexity of the MSC paracrine regulation.
Collapse
Affiliation(s)
| | - Ulla Impola
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Ilja Ritamo
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Maria Aatonen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | | | | | - Leena Valmu
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Piet Finckenberg
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Pia Siljander
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland ; Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
13
|
Hirvonen T, Suila H, Tiitinen S, Natunen S, Laukkanen ML, Kotovuori A, Reinman M, Satomaa T, Alfthan K, Laitinen S, Takkinen K, Räbinä J, Valmu L. Production of a recombinant antibody specific for i blood group antigen, a mesenchymal stem cell marker. Biores Open Access 2013; 2:336-45. [PMID: 24083089 PMCID: PMC3777189 DOI: 10.1089/biores.2013.0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) offer great promise for future regenerative and anti-inflammatory therapies. Panels of functional and phenotypical markers are currently used in characterization of different therapeutic stem cell populations from various sources. The i antigen (linear poly-N-acetyllactosamine) from the Ii blood group system has been suggested as a marker for MSCs derived from umbilical cord blood (UCB). However, there are currently no commercially available antibodies recognizing the i antigen. In the present study, we describe the use of antibody phage display technology to produce recombinant antibodies recognizing a structure from the surface of mesenchymal stem cells. We constructed IgM phage display libraries from the lymphocytes of a donor with an elevated serum anti-i titer. Antibody phage display technology is not dependent on immunization and thus allows the generation of antibodies against poorly immunogenic molecules, such as carbohydrates. Agglutination assays utilizing i antigen–positive red blood cells (RBCs) from UCB revealed six promising single-chain variable fragment (scFv) antibodies, three of which recognized epitopes from the surface of UCB-MSCs in flow cytometric assays. The amino acid sequence of the VH gene segment of B12.2 scFv was highly similar to the VH4.21 gene segment required to encode anti-i specificities. Further characterization of binding properties revealed that the binding of B12.2 hyperphage was inhibited by soluble linear lactosamine oligosaccharide. Based on these findings, we suggest that the B12.2 scFv we have generated is a prominent anti-i antibody that recognizes i antigen on the surface of both UCB-MSCs and RBCs. This binder can thus be utilized in UCB-MSC detection and isolation as well as in blood group serology.
Collapse
Affiliation(s)
- Tia Hirvonen
- Finnish Red Cross Blood Service , Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jin HJ, Bae YK, Kim M, Kwon SJ, Jeon HB, Choi SJ, Kim SW, Yang YS, Oh W, Chang JW. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci 2013; 14:17986-8001. [PMID: 24005862 PMCID: PMC3794764 DOI: 10.3390/ijms140917986] [Citation(s) in RCA: 445] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/18/2013] [Accepted: 08/22/2013] [Indexed: 02/07/2023] Open
Abstract
Various source-derived mesenchymal stem cells (MSCs) have been considered for cell therapeutics in incurable diseases. To characterize MSCs from different sources, we compared human bone marrow (BM), adipose tissue (AT), and umbilical cord blood-derived MSCs (UCB-MSCs) for surface antigen expression, differentiation ability, proliferation capacity, clonality, tolerance for aging, and paracrine activity. Although MSCs from different tissues have similar levels of surface antigen expression, immunosuppressive activity, and differentiation ability, UCB-MSCs had the highest rate of cell proliferation and clonality, and significantly lower expression of p53, p21, and p16, well known markers of senescence. Since paracrine action is the main action of MSCs, we examined the anti-inflammatory activity of each MSC under lipopolysaccharide (LPS)-induced inflammation. Co-culture of UCB-MSCs with LPS-treated rat alveolar macrophage, reduced expression of inflammatory cytokines including interleukin-1α (IL-1α), IL-6, and IL-8 via angiopoietin-1 (Ang-1). Using recombinant Ang-1 as potential soluble paracrine factor or its small interference RNA (siRNA), we found that Ang-1 secretion was responsible for this beneficial effect in part by preventing inflammation. Our results demonstrate that primitive UCB-MSCs have biological advantages in comparison to adult sources, making UCB-MSCs a useful model for clinical applications of cell therapy.
Collapse
Affiliation(s)
- Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
- Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea; E-Mail:
| | - Yun Kyung Bae
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Miyeon Kim
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Soon-Jae Kwon
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Hong Bae Jeon
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Seong Who Kim
- Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea; E-Mail:
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
| | - Jong Wook Chang
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Korea; E-Mails: (H.J.J.); (Y.K.B.); (M.K.); (S.-J.K.); (H.B.J.); (S.J.C.); (Y.S.Y.); (W.O.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +82-2-3465-6771; Fax: +82-2-475-1991
| |
Collapse
|
15
|
Nystedt J, Anderson H, Tikkanen J, Pietilä M, Hirvonen T, Takalo R, Heiskanen A, Satomaa T, Natunen S, Lehtonen S, Hakkarainen T, Korhonen M, Laitinen S, Valmu L, Lehenkari P. Cell surface structures influence lung clearance rate of systemically infused mesenchymal stromal cells. Stem Cells 2013; 31:317-26. [PMID: 23132820 DOI: 10.1002/stem.1271] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/13/2012] [Indexed: 12/13/2022]
Abstract
The promising clinical effects of mesenchymal stromal/stem cells (MSCs) rely especially on paracrine and nonimmunogenic mechanisms. Delivery routes are essential for the efficacy of cell therapy and systemic delivery by infusion is the obvious goal for many forms of MSC therapy. Lung adhesion of MSCs might, however, be a major obstacle yet to overcome. Current knowledge does not allow us to make sound conclusions whether MSC lung entrapment is harmful or beneficial, and thus we wanted to explore MSC lung adhesion in greater detail. We found a striking difference in the lung clearance rate of systemically infused MSCs derived from two different clinical sources, namely bone marrow (BM-MSCs) and umbilical cord blood (UCB-MSCs). The BM-MSCs and UCB-MSCs used in this study differed in cell size, but our results also indicated other mechanisms behind the lung adherence. A detailed analysis of the cell surface profiles revealed differences in the expression of relevant adhesion molecules. The UCB-MSCs had higher expression levels of α4 integrin (CD49d, VLA-4), α6 integrin (CD49f, VLA-6), and the hepatocyte growth factor receptor (c-Met) and a higher general fucosylation level. Strikingly, the level of CD49d and CD49f expression could be functionally linked with the lung clearance rate. Additionally, we saw a possible link between MSC lung adherence and higher fibronectin expression and we show that the expression of fibronectin increases with MSC culture confluence. Future studies should aim at developing methods of transiently modifying the cell surface structures in order to improve the delivery of therapeutic cells.
Collapse
Affiliation(s)
- Johanna Nystedt
- Advanced Therapies and Product Development, Finnish Red Cross Blood Service, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kerkelä E, Hakkarainen T, Mäkelä T, Raki M, Kambur O, Kilpinen L, Nikkilä J, Lehtonen S, Ritamo I, Pernu R, Pietilä M, Takalo R, Juvonen T, Bergström K, Kalso E, Valmu L, Laitinen S, Lehenkari P, Nystedt J. Transient proteolytic modification of mesenchymal stromal cells increases lung clearance rate and targeting to injured tissue. Stem Cells Transl Med 2013; 2:510-20. [PMID: 23734061 DOI: 10.5966/sctm.2012-0187] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Systemic infusion of therapeutic cells would be the most practical and least invasive method of administration in many cellular therapies. One of the main obstacles especially in intravenous delivery of cells is a massive cell retention in the lungs, which impairs homing to the target tissue and may decrease the therapeutic outcome. In this study we showed that an alternative cell detachment of mesenchymal stromal/stem cells (MSCs) with pronase instead of trypsin significantly accelerated the lung clearance of the cells and, importantly, increased their targeting to an area of injury. Cell detachment with pronase transiently altered the MSC surface protein profile without compromising cell viability, multipotent cell characteristics, or immunomodulative and angiogenic potential. The transient modification of the cell surface protein profile was sufficient to produce effective changes in cell rolling behavior in vitro and, importantly, in the in vivo biodistribution of the cells in mouse, rat, and porcine models. In conclusion, pronase detachment could be used as a method to improve the MSC lung clearance and targeting in vivo. This may have a major impact on the bioavailability of MSCs in future therapeutic regimes.
Collapse
Affiliation(s)
- Erja Kerkelä
- Advanced Therapies and Product Development, Finnish.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Natunen S, Lampinen M, Suila H, Ritamo I, Pitkänen V, Nairn AV, Räbinä J, Laitinen S, Moremen KW, Reutter W, Valmu L. Metabolic glycoengineering of mesenchymal stromal cells with N-propanoylmannosamine. Glycobiology 2013; 23:1004-12. [PMID: 23708401 DOI: 10.1093/glycob/cwt039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
There is an increasing interest in the modification of cell surface glycosylation to improve the properties of therapeutic cells. For example, glycosylation affects the biodistribution of mesenchymal stromal cells (MSCs). Metabolic glycoengineering is an efficient way to modify the cell surface. The mammalian biosynthetic machinery tolerates the unnatural sialic acid precursor, N-propanoylmannosamine (ManNProp), and incorporates it into cell surface glycoconjugates. We show here by mass spectrometric analysis of cell surface N-glycans that about half of N-acetylneuraminic acid was replaced by N-propanoylneuraminic acid in the N-glycans of human umbilical cord blood-derived MSCs supplemented with ManNProp. In addition, the N-glycan profile was altered. ManNProp-supplemented cells had more multiply fucosylated N-glycan species than control cells. The fucosylated epitopes were shown in tandem mass spectrometric analysis to be Lewis x or blood group H epitopes, but not sialyl Lewis x (sLex). The amounts of tri- and tetra-antennary and polylactosamine-containing N-glycans also increased in ManNProp supplementation. In accordance with previous studies of other cell types, increased expression of the sLex epitope in ManNProp-supplemented MSCs was demonstrated by flow cytometry. In light of the N-glycan analysis, the sLex epitope in these cells is likely to be carried by O-glycans or glycolipids. sLex has been shown to target MSCs to bone marrow, which may be desirable in therapeutic applications. The present results represent the first structural analysis of an N-glycome of ManNProp-supplemented cells and demonstrate the feasibility of modifying cell surface glycosylation of therapeutic cells by this type of metabolic glycoengineering.
Collapse
Affiliation(s)
- Suvi Natunen
- Finnish Red Cross Blood Service, 00310 Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Walter J, Hausmann S, Drepper T, Puls M, Eggert T, Dihné M. Flavin mononucleotide-based fluorescent proteins function in mammalian cells without oxygen requirement. PLoS One 2012; 7:e43921. [PMID: 22984451 PMCID: PMC3439463 DOI: 10.1371/journal.pone.0043921] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/27/2012] [Indexed: 11/23/2022] Open
Abstract
Usage of the enhanced green fluorescent protein (eGFP) in living mammalian cells is limited to aerobic conditions due to requirement of oxygen during chromophore formation. Since many diseases or disease models are associated with acute or chronic hypoxia, eGFP-labeling of structures of interest in experimental studies might be unreliable leading to biased results. Thus, a chromophore yielding a stable fluorescence under hypoxic conditions is desirable. The fluorescence of flavin mononucleotide (FMN)-based fluorescent proteins (FbFPs) does not require molecular oxygen. Recently, the advantages of FbFPs for several bacterial strains and yeasts were described, specifically, their usage as a real time fluorescence marker in bacterial expression studies and their ability of chromophore formation under anaerobic conditions. Our objective was to verify if FbFPs also function in mammalian cells in order to potentially broaden the repertoire of chromophores with ones that can reliably be used in mammalian studies under hypoxic conditions. In the present study, we demonstrate for the first time, that FbFPs can be expressed in different mammalian cells, among them murine neural stem cells during proliferative and differentiated stages. Fluorescence intensities were comparable to eGFP. In contrast to eGFP, the FbFP fluorescence did not decrease when cells were exposed to defined hypoxic conditions neither in proliferating nor in differentiated cells. Thus, FbFPs can be regarded as an alternative to eGFP in studies that target cellular structures which are exposed to hypoxic conditions.
Collapse
Affiliation(s)
- Janine Walter
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Baden-Württemberg, Germany.
| | | | | | | | | | | |
Collapse
|
19
|
Pietilä M, Lehtonen S, Tuovinen E, Lähteenmäki K, Laitinen S, Leskelä HV, Nätynki A, Pesälä J, Nordström K, Lehenkari P. CD200 positive human mesenchymal stem cells suppress TNF-alpha secretion from CD200 receptor positive macrophage-like cells. PLoS One 2012; 7:e31671. [PMID: 22363701 PMCID: PMC3282758 DOI: 10.1371/journal.pone.0031671] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 01/11/2012] [Indexed: 01/14/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) display immunosuppressive properties in vitro and the potential has also been transferred successfully to clinical trials for treatment of autoimmune diseases. OX-2 (CD200), a member of the immunoglobulin superfamily, is widely expressed in several tissues and has recently been found from hMSCs. The CD200 receptor (CD200R) occurs only in myeloid-lineage cells. The CD200-CD200R is involved in down-regulation of several immune cells, especially macrophages. The present study on 20 hMSC lines shows that the CD200 expression pattern varied from high (CD200Hi) to medium (CD200Me) and low (CD200Lo) in bone marrow-derived mesenchymal stem cell (BMMSC) lines, whereas umbilical cord blood derived mesenchymal stem cells (UCBMSCs) were constantly negative for CD200. The role of the CD200-CD200R axis in BMMSCs mediated immunosuppression was studied using THP-1 human macrophages. Interestingly, hMSCs showed greater inhibition of TNF-α secretion in co-cultures with IFN-γ primed THP-1 macrophages when compared to LPS activated cells. The ability of CD200Hi BMMSCs to suppress TNF-α secretion from IFN-γ stimulated THP-1 macrophages was significantly greater when compared to CD200Lo whereas UCBMSCs did not significantly reduce TNF-α secretion. The interference of CD200 binding to the CD200R by anti-CD200 antibody weakened the capability of BMMSCs to inhibit TNF-α secretion from IFN-γ activated THP-1 macrophages. This study clearly demonstrated that the efficiency of BMMSCs to suppress TNF-α secretion of THP-1 macrophages was dependent on the type of stimulus. Moreover, the CD200-CD200r axis could have a previously unidentified role in the BMMSC mediated immunosuppression.
Collapse
Affiliation(s)
- Mika Pietilä
- Institute of Biomedicine, Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hirvonen T, Suila H, Kotovuori A, Ritamo I, Heiskanen A, Sistonen P, Anderson H, Satomaa T, Saarinen J, Tiitinen S, Räbinä J, Laitinen S, Natunen S, Valmu L. The i blood group antigen as a marker for umbilical cord blood-derived mesenchymal stem cells. Stem Cells Dev 2011; 21:455-64. [PMID: 21933024 DOI: 10.1089/scd.2011.0405] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) offer great promise for future regenerative and anti-inflammatory therapies. However, there is a lack of methods to quickly and efficiently isolate, characterize, and ex vivo expand desired cell populations for therapeutic purposes. Single markers to identify cell populations have not been characterized; instead, all characterizations rely on panels of functional and phenotypical properties. Glycan epitopes can be used for identifying and isolating specific cell types from heterogeneous populations, on the basis of their cell-type specific expression and prominent cell surface localization. We have now studied in detail the cell surface expression of the blood group i epitope (linear poly-N-acetyllactosamine chain) in umbilical cord blood (UCB)-derived MSCs. We used flow cytometry and mass spectrometric glycan analysis and discovered that linear poly-N-acetyllactosamine structures are expressed in UCB-derived MSCs, but not in cells differentiated from them. We further verified the findings by mass spectrometric glycan analysis. Gene expression analysis indicated that the stem-cell specific expression of the i antigen is determined by β3-N-acetylglucosaminyltransferase 5. The i antigen is a ligand for the galectin family of soluble lectins. We found concomitant cell surface expression of galectin-3, which has been reported to mediate the immunosuppressive effects exerted by MSCs. The i antigen may serve as an endogenous ligand for this immunosuppressive agent in the MSC microenvironment. Based on these findings, we suggest that linear poly-N-acetyllactosamine could be used as a novel UCB-MSC marker either alone or within an array of MSC markers.
Collapse
Affiliation(s)
- Tia Hirvonen
- Department of Advanced Therapy and Product Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pietilä M, Palomäki S, Lehtonen S, Ritamo I, Valmu L, Nystedt J, Laitinen S, Leskelä HV, Sormunen R, Pesälä J, Nordström K, Vepsäläinen A, Lehenkari P. Mitochondrial function and energy metabolism in umbilical cord blood- and bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2011; 21:575-88. [PMID: 21615273 DOI: 10.1089/scd.2011.0023] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive choice for a variety of cellular therapies. hMSCs can be isolated from many different tissues and possess unique mitochondrial properties that can be used to determine their differentiation potential. Mitochondrial properties may possibly be used as a quality measure of hMSC-based products. Accordingly, the present work focuses on the mitochondrial function of hMSCs from umbilical cord blood (UCBMSC) cells and bone marrow cells from donors younger than 18 years of age (BMMSC <18) and those more than 50 years of age (BMMSC >50). Changes of ultrastructure and energy metabolism during osteogenic differentiation in all hMSC types were studied in detail. Results show that despite similar surface antigen characteristics, the UCBMSCs had smaller cell surface area and possessed more abundant rough endoplasmic reticulum than BMMSC >50. BMMSC <18 were morphologically more UCBMSC-like. UCBMSC showed dramatically higher mitochondrial-to-cytoplasm area ratio and elevated superoxide and manganese superoxide dismutase (MnSOD) levels as compared with BMMSC >50 and BMMSC <18. All hMSCs types showed changes indicative of mitochondrial activation after 2 weeks of osteogenic differentiation, and the increase in mitochondrial-to-cytoplasm area ratio appears to be one of the first steps in the differentiation process. However, BMMSC >50 showed a lower level of mitochondrial maturation and differentiation capacity. UCBMSCs and BMMSCs also showed a different pattern of exocytosed proteins and glycoproteoglycansins. These results indicate that hMSCs with similar cell surface antigen expression have different mitochondrial and functional properties, suggesting different maturation levels and other significant biological variations of the hMSCs. Therefore, it appears that mitochondrial analysis presents useful characterization criteria for hMSCs intended for clinical use.
Collapse
Affiliation(s)
- Mika Pietilä
- Department of Anatomy and Cell Biology, Institute of Biomedicine, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|